Abstract

There has been an alarming increase in the prevalence of obesity in people with type 1 diabetes in recent years. Although obesity has long been recognized as a major risk factor for the development of type 2 diabetes and a catalyst for complications, much less is known about the role of obesity in the initiation and pathogenesis of type 1 diabetes. Emerging evidence suggests that obesity contributes to insulin resistance, dyslipidemia, and cardiometabolic complications in type 1 diabetes. Unique therapeutic strategies may be required to address these comorbidities within the context of intensive insulin therapy, which promotes weight gain. There is an urgent need for clinical guidelines for the prevention and management of obesity in type 1 diabetes. The development of these recommendations will require a transdisciplinary research strategy addressing metabolism, molecular mechanisms, lifestyle, neuropsychology, and novel therapeutics. In this review, the prevalence, clinical impact, energy balance physiology, and potential mechanisms of obesity in type 1 diabetes are described, with a special focus on the substantial gaps in knowledge in this field. Our goal is to provide a framework for the evidence base needed to develop type 1 diabetes–specific weight management recommendations that account for the competing outcomes of glycemic control and weight management.

Essential Points
  • Obesity has long been known to be a concurrent phenotype and predisposing factor for type 2 diabetes, but its prevalence, consequences, and physiological attributes in type 1 diabetes are poorly understood

  • There is an urgent need to develop type 1 diabetes–specific guidelines for the prevention and treatment of obesity and achievement of optimal glycemic control

  • The characteristics of energy balance in type 1 diabetes are not fully defined and will be impacted on a clinical level by both glycemic control and medical therapy

  • The mechanisms that drive obesity in type 1 diabetes are likely to include genetics, epigenetics, enteroendocrine hormones, microbiome, and effects on multiple tissues, organs, cells, and organelles

  • Developing weight management strategies in individuals with type 1 diabetes will require a transdisciplinary research approach that combines expertise in epidemiology, endocrinology, clinical psychology, translational metabolism, nutrition, exercise physiology, mathematical modeling, and advanced analytics

It has long been known that obesity contributes to the pathophysiology and complications of type 2 diabetes (T2D) (1). Importantly, specific guidelines emphasize the management of obesity in individuals with both prediabetes and T2D due to the strong evidence that treatment of obesity can delay the onset of T2D and improve glycemic control (2). Obesity, once rare in type 1 diabetes (T1D), is now an increasingly frequent problem (3–5). Accumulating evidence on the prevalence and consequences of obesity in T1D shows very clearly that this comorbidity is both underappreciated and poorly understood. Obesity is likely due to changes in the environment impacting the population as a whole and an unintended consequence of modern, more intensive approaches to the management of hyperglycemia in T1D, as evidenced by the Diabetes Control and Complications Trial (DCCT) and other studies (4, 5). Obesity is a major public health concern (6) that influences health in multiple ways, from cardiometabolic disease (7–10) to cancer (11, 12). This review defines the current state of the evidence related to the prevalence, contributing factors, adverse outcomes, physiology, and mechanisms connecting obesity and T1D. The substantial gaps in our mechanistic understanding of obesity in T1D are a major focus of this review. We conclude with a description of the transdisciplinary research paradigm needed for the development of the first T1D-specific recommendations for the prevention and treatment of obesity that concurrently address energy balance and glycemic control.

Epidemiology of Obesity in T1D

T1D is one of the most common chronic diseases of childhood. In the United States, both incidence and prevalence are increasing: prevalence increased by 21.1% (95% CI, 15.6% to 27.0%) between 2001 and 2008 (13), and incidence increased by 1.8% per year (95% CI, 1.0% to 2.6%) between 2002 and 2012 (14). The burden of T1D is expected to continue to rise during the next several decades (15). T1D complications, including hospitalization due to hypoglycemic and hyperglycemic crises, retinopathy, neuropathy, and kidney disease/failure, are major causes of morbidity and health care expenditures, although cardiovascular disease is the leading cause of death (16).

In parallel with the recent increase in prevalence of T1D, there has been a concerning increase in obesity (4, 5, 15, 17). Although the physiology and consequences of obesity in T2D have been extensively studied (18), the relatively new problem of obesity in T1D has received little attention. The SEARCH for Diabetes in Youth Study, using anthropometric measures conducted under a standardized protocol, found that 22.1% of youth with T1D were overweight compared with 16.1% of youth without T1D from the National Health and Nutrition Examination Survey (NHANES). The strength of these findings lies in the comparison of the same time period and ages, while contrasting individuals with and without T1D (19). The T1D Exchange Registry (T1DX) and the Prospective Diabetes Follow-up Registry showed that youth 2 to 18 years of age diagnosed with T1D for at least 1 year from Germany, Austria, and the United States had greater body mass index (BMI) z scores as compared with World Health Organization international norms and respective country-specific national norms (20). The considerable obesity burden in youth with T1D was confirmed, also in the T1DX, where the prevalence of obesity (BMI ≥95th percentile) was 13.5% in adolescents (mean age 15.4 years), with higher prevalences in those of black/African American descent (17.9%) and Hispanic/Latino descent (15.9%) between the years of 2010 and 2012 (5). In comparison, NHANES data from 2011 to 2014 show that the prevalence of obesity in adolescents in the United States (age 12 to 19 years) is 20.5% with the highest prevalences in those of Hispanic (22.8%) and non-Hispanic black (22.6%) descent (21). In adults with T1D from the Pittsburgh Epidemiology of Diabetes Complications (Pittsburgh EDC) Study, the prevalence of obesity increased across time, reaching 22.7% during the final reported time point (2004 to 2007) (3). In comparative data from adults in NHANES, rates of obesity also increased over time, with a prevalence of 33.8% during the 2007 to 2008 data collection period (22).

A study of temporal trends shows an increase in obesity in adults with T1D from 3.4% at baseline (1986 to 1988) to 22.7% (2004 to 2007). Importantly, the rise in prevalence of overweight and obesity was not due to aging of the cohort and occurred faster than the increases in the general population (3). In a study that considered the longitudinal clinical course of T1D, obesity prevalence increased from 1% at baseline (1983 to 1989) to 31% at year 12 (2005) of the Epidemiology of Diabetes Interventions and Complications (EDIC) trial in the group originally randomized to intensive therapy. The rate of change was much higher than in the general population (23). Although limited studies have directly compared obesity in T1D vs healthy populations matched for age, sex, and time frame (19, 20), the studies presented illustrate a concerning trend of obesity in individuals with T1D. Whether and how factors that impact obesity in the general population (e.g., aging, individual biological factors, environmental pressures, and socioeconomic status) modulate the effects of intensive insulin therapy on secular obesity trends in T1D is an area that requires further investigation.

Key Therapeutic and Psychosocial Factors Affecting Body Weight in T1D

Lifestyle interventions

Nutrition and exercise are important therapeutic avenues to improve glycemic control and manage body weight. There is extensive research and expert consensus on how to manage the often co-occurring phenotypes of obesity and T2D through lifestyle interventions (2). These guidelines are individualized in nature to account for metabolic status, clinical goals, preferences, and sociocultural considerations (2, 24). However, optimal dietary approaches for weight management and metabolic control remain controversial. The available literature is essentially all on T2D and varies in dietary approach, sample population, T2D duration, and treatment regimen (25, 26). Current American Diabetes Association (ADA) Standards of Medical Care emphasize that in patients with T2D, weight loss can be achieved with hypocaloric diets with macronutrient composition based on the individual’s medical status and preferences (2). These current guidelines lack the evidence to address the competing clinical outcomes of weight management and optimal glycemic control that are so prominent in people with T1D. Randomized controlled clinical trials that evaluate dietary approaches to optimize both weight management and glycemic control in T1D have not been conducted.

In conjunction with reduced energy intake and optimized macronutrient distribution, physical activity offers benefits for weight management and health in the general population (27). For adults with diabetes, 150 minutes of physical activity is recommended throughout the week, with no more than 2 consecutive days devoid of physical activity (28). Most pediatric and adult patients with T1D (both normal weight and overweight) do not achieve the minimum physical activity recommendations (28–30). Reduced physical activity levels increase adipose tissue mass in patients with T1D and negatively impact muscle mass and quality. This further deteriorates metabolic control, cardiovascular health, and cardiorespiratory fitness (31). Those patients who do meet physical activity guidelines have better body composition, lower HbA1c levels, and a better lipid profile (32). Therefore, increased physical activity levels could dramatically improve the health of individuals with T1D. A recent consensus statement provides the most up-to-date exercise parameters for people with T1D with respect to glycemic targets, safety, efficacy, nutrition, and insulin adjustments to avoid complications, as well as the barriers to exercise (33). However, there is a pressing need for studies to address exercise efficacy and responsiveness in lean individuals vs those with obesity, particularly ones that concurrently address advanced medical therapies and differences in energy balance physiology. The major challenge for optimizing both exercise and nutritional interventions is understanding the specific metabolic parameters that drive energy and macronutrient balance (discussed in subsequent sections) so that fuel oxidation can be restored to a normal state while providing the caloric inputs needed to achieve the desired weight outcome.

Behavioral and psychosocial factors

In the general population, psychosocial factors such as socioeconomic status, access to health care and healthy foods, unhealthy diet of friends and family, low self-esteem, depression, peer relationships, and level of family/social support influence the risk for obesity and success with weight loss strategies (34–36). These factors are tightly linked to eating behaviors in youth with T1D and T2D (37). Although a body of evidence exists that is focused on the behavioral and psychosocial context of obesity and T2D (38–40), the knowledge base is emerging regarding obesity and weight management in people with T1D. Our complementary review of the biopsychosocial aspects of weight management in T1D focuses on the multitude of factors, including depression, fear of hypoglycemia, stress, quality of life, cultural ideals of body image, body dissatisfaction, and past dieting behaviors, that could influence the risk for obesity, present challenges for weight maintenance, and promote disordered eating behaviors similarly in T1D (41).

In brief, numerous studies have described disordered eating behavior patterns and eating disorders in individuals with T1D (41–45). For example, a unique maladaptive approach to body weight management is the use of insulin restriction/manipulation as an effective, yet extremely dangerous means to manage weight. This volitional omission of insulin to control weight is a well-described phenomenon and involves completely omitting insulin or purposefully delivering less insulin than is needed (46). It is estimated that 20% to 40% of youth with T1D have engaged in insulin manipulation to prevent weight gain (43, 47), and insulin manipulation has been shown to persist into adulthood (45). Female sex, higher BMI, infrequent family meals, high family attention to weight, depression, low self-esteem, and body dissatisfaction are associated with an increased risk of behaviors where insulin restriction is used as a means to control body weight in individuals with T1D (43, 47, 48).

Strategic insulin restriction as a means of “calorie purging” to prevent weight gain causes chronic hyperglycemia with symptoms of dehydration, loss of lean body tissue, and glycosuria. The loss of glucose prevents it from being used by muscle and fat, which leads to further deterioration of glycemic control, the breakdown of muscle and fat for energy, and ketogenesis (49). Furthermore, it promotes deleterious medical outcomes, including high rates of diabetic ketoacidosis, chronic daily hyperglycemia, suboptimal glycemic control, nephropathy, and foot problems (44, 49). Insulin manipulation behaviors lead to an increase in morbidity as evidenced by a threefold increase in relative risk of death for individuals who restrict insulin (49). With an increased need to achieve and maintain a healthy body weight in people with T1D, simultaneously with achieving euglycemia, a greater focus will likely need to be placed on mitigating potentially harmful behaviors that are a response to these two competing clinical outcomes.

Current medical approaches

Differences between physiological and endogenous insulin delivery

There are gaps in our current ability to replace the physiological deficits that emerge with endocrine pancreatic dysfunction. The demise of insulin-producing pancreatic β-cells is the central pathophysiological defect in T1D and is associated with α-cell dysfunction and impaired glucagon secretion. There is a lack of glucagon suppression in the postprandial state (50) and an inability to release glucagon in response to hypoglycemia (51). The nonphysiologic subcutaneous delivery of insulin in T1D results in an imbalance between the hepatic and peripheral effects of insulin. This can lead to suboptimal control of hepatic glucose production, primarily via aberrant regulation of fasting glycogenesis and gluconeogenesis and postprandial suppression of glucagon secretion as well as peripheral hyperinsulinemia, which could contribute to both weight gain and hypoglycemia (52, 53). Glucagon dysregulation contributes to fasting and postprandial hyperglycemia in people with T1D (50), but also to an increased risk of hypoglycemia (51), particularly in the later stages of T1D. Additionally, amylin, which is cosecreted from β-cells, is deficient in T1D. Upon a meal stimulus, amylin functions to suppress glucagon and reduce gastric emptying. Amylin deficiency contributes to the inability to control postprandial glucose levels and has anorexigenic effects (54–57). This suggests that amylin is important for overall carbohydrate homeostasis and energy balance and these functions are not replaced with insulin monotherapy. Studies of the amylin analog pramlintide have demonstrated that adjunctive use with prandial insulin in patients with T1D is associated with improvements in postprandial and overall glucose control, as well as weight loss (55, 57–60)

Intensive insulin therapy

A key contributor to obesity in T1D is the weight gain that occurs with intensive insulin therapy. One of the earliest direct demonstrations of this association was in the DCCT, which was a randomized controlled trial of conventional vs intensive insulin therapy, where obesity emerged as a result of treatment-associated weight gain (61). EDIC, the follow-up study to the DCCT, extended those findings by showing that participants continued to gain weight during the course of 30 years of follow-up (17, 62). In a multicenter study that gathered post hoc observational data between 2006 and 2009, BMI was higher in youth (ages 9 to 14.9 years) who were placed on intensive insulin regimens to address suboptimal glycemic control based on each center’s standards of care (63). Within the Pittsburgh EDC cohort, an investigation of long-term temporal patterns of weight change in relationship to intensive insulin therapy revealed that in a period of 18 years, the percentage of individuals on intensive insulin therapy increased 10-fold in parallel to a sevenfold increase in obesity and a 47% increase in overweight status (3). In contrast, in a cross-sectional investigation of four cohorts in a single center (1999, 2002, 2006, and 2009), the use of intensive insulin therapy increased from 52% to 97%, but the prevalence of obesity remained stable: neither the mean z BMI nor the distribution of weight status differed significantly across the four cohorts. Although weight did not differ based on treatment regimen, the authors noted that the prevalence of obesity paralleled that of the general population [based on Ref. (64)]. Glycemic control did not improve with greater use of intensive insulin therapy, which could partially explain the stability of weight (4).

The DCCT demonstrated the importance of intensive insulin therapy for achieving optimal glycemic control to decrease long-term complications, including retinopathy, nephropathy, and cardiovascular disease (61). Thus, even with the demonstration of a clear association between intensive insulin therapy and weight gain, insulin is the foundation of T1D medical management. Current ADA Standards of Care recommend intensive insulin therapy that consists of multiple-dose insulin injections (three to four injections per day of basal and prandial insulin) or insulin pump therapy (65). If obesity rates continue to rise in the general population due to various environmental and biological factors (21, 22, 66) as they have since the initial obesity reports from the DCCT (61), modulators other than insulin regimen could mask treatment-associated weight gain or lead to a compounding of the problem. There is a pressing need to continue to identify the emerging factors that promote obesity in T1D.

Insulin pumps

Automated or closed-loop delivery of insulin and glucagon has been the subject of intense research in the past decade. Feasibility of both insulin-only and bihormonal delivery systems have been established (67) and the first hybrid closed-loop system, the Medtronic 670G, was approved by the Food and Drug Administration in 2016. Multiple other systems are in development by academic and industry researchers (68). Current closed-loop systems are hybrid systems requiring users to manually deliver insulin bolus doses prior to meals/snacks to limit postprandial hyperglycemia, in part due to limitations of current insulin preparations (69). With closed-loop systems, improved overnight glucose control reduces hypoglycemia and increases time in target range by modulating insulin (and glucagon in bihormonal systems). This is in contrast to current open-loop systems that rely on preset insulin basal settings or user adjustments in real time.

It is uncertain what effect closed-loop systems that potentially allow even tighter control will have on obesity in people with T1D, although weight gain during 3 months was reported in the recent 670G trial [1.0 kg in adolescents (P = 0.065) and 1.4 kg in adults (P < 0.001)] (70). There may be different impacts on outcomes between subcutaneous insulin delivery and intraportal delivery via implantable pumps. The Veteran Affairs Implantable Insulin Pump Study tested the impact of 1 year of intensive therapy via multiple daily injections vs an implantable insulin pump in male patients with T2D. They found that weight gain was eliminated with the intraportal delivery method (71). There are insufficient data on bihormonal systems to determine whether weight gain is impacted differentially compared with other methods, as the studies to date have focused on safety and performance (72–74).

“Several studies indicate that the common sequelae of obesity in the general population also occur in individuals with T1D.”

It is reasonable to hypothesize that people with T1D with elevated HbA1c who attain better glucose control with closed-loop systems are likely to gain weight unless behavioral modifications of diet and exercise are implemented. In contrast, for individuals with T1D who have an HbA1c at target (i.e., <7.5%) but who achieve better glycemic control at the expense of recurrent hypoglycemia (65), closed-loop systems may reduce the need for unwanted carbohydrate intake to treat or prevent symptomatic hypoglycemia. If closed-loop systems become widely available clinically (75), then patient eating behaviors and glucose profiles may shift in a manner that promotes positive energy balance (76). This may require the implementation of obesity prevention strategies as a clinical priority.

Adjunctive therapies to insulin

Adjunctive therapies to insulin in T1D have the goal of improving glucose control while capitalizing on potentially beneficial effects on obesity and cardiovascular disease risk factors (77). Pramlintide, an amylin analog, is one such therapy, discussed earlier in this section (55, 57). Metformin has been studied most extensively with minimal improvement on glucose control as measured by HbA1c, but with benefits on weight, reduction of insulin doses, and improvement in some cardiovascular disease risk factors (78, 79). A recent meta-analysis of seven randomized controlled clinical trials indicated that addition of a glucagon-like peptide-1 (GLP-1) receptor agonist to insulin therapy modestly decreased HbA1c (−0.21%) and body weight (−3.5 kg). Most of these trials were small and of short duration, indicating the need for additional study (80). Limited data exist on sodium-glucose cotransporter (SGLT)2 inhibitors in addition to insulin therapy in T1D, but reports suggest improvement in HbA1c, weight, and waist circumference (81, 82). SGLT2 inhibitors have been associated with euglycemic diabetic ketoacidosis (83), yet a recent meta-analysis of four randomized clinical trials suggested no difference in adverse events as compared with placebo (84). SGLT2 inhibitors are not currently approved for use in T1D and additional studies are needed to understand this variability in reported safety data. A combination SGLT1/2 inhibitor (sotagliflozin) reduced body weight and increased time in glucose targets in a phase 3 study of adults with T1D and is the subject of ongoing research (85). Dipeptidyl peptidase-4 (DPP-4) inhibitors also have limited data in T1D with a meta-analysis reporting no benefit on HbA1c but reduction in insulin doses (86). In T2D, DPP-4 inhibitors are weight neutral (87) and do not have meaningful effects on cardiovascular disease (88–90). Data from the T1DX and the Prospective Diabetes Follow-up registries report that in 49,367 patients between the years 2015 and 2016, the use of any adjuvant medication to insulin was 5.4% in the T1DX registry and 2.0% in the Prospective Diabetes Follow-up registry, with metformin accounting for most (91). Further research is required to determine what medications may have dual beneficial effects on glucose control and weight in people with T1D.

Hypoglycemia

Another unique clinical factor potentially impacting both glycemic control and weight in T1D is hypoglycemia. In the DCCT, lower HbA1c levels were associated with increased severe hypoglycemia (92). More recent registry data indicate a weaker relationship between increased severe hypoglycemia and lower HbA1c; importantly, however, note that these studies did not quantify nonsevere hypoglycemia (93, 94). The relationship between rates of hypoglycemia based on treatment regimen has been extensively studied. A meta-analysis of 21 studies using modern pumps and insulins/insulin analogs showed that the rate of hypoglycemia was markedly reduced during continuous subcutaneous insulin infusion vs multiple daily injections with a rate ratio of 4.19 (95% CI, 2.86 to 6.13). Glycemic control was also improved on pump therapy with a mean improvement in HbA1C of 0.62% (95% CI, 0.47% to 0.78%) (95).

Although rates of severe hypoglycemia have decreased over time (96), they remain a concern as do clinically meaningful (but not severe) hypoglycemia of <70 mg/dL that require carbohydrate treatment (97). Acute hypoglycemia is associated with food cravings, particularly for foods that are rich in carbohydrates, which can lead to disinhibited eating behaviors (98). To compensate, excessive caloric intake and subsequent insulin omission may occur, which could lead to worsening glycemic control (99). For example, the recommended 15-g (~60 kcal) carbohydrate rescue for hypoglycemia taken once daily could translate into an excess of 6 pounds (2.7 kg) during the course of a year. Often, patients consume far more than this to treat hypoglycemic episodes.

Several studies have examined the relationship between hypoglycemia and weight. A study in 1168 DCCT participants aged ≥18 years at baseline (n = 586 on conventional treatment; n = 582 on intensive insulin therapy) who were followed for ~6 years found that there was no relationship between hypoglycemia and BMI (100). In a small cohort of DCCT participants (n = 29; age 13 to 39 years) on intensive insulin therapy followed for 1 year, those who experienced severe hypoglycemia gained more weight than those who did not (6.8 ± 4.8 kg vs 4.7 ± 6.3 kg, respectively) (61). In youth age 12 to 20 years (n = 75), the rate of hypoglycemic events requiring assistance or resulting in coma were reduced by ~50% and weight gain was blunted in adolescents on pumps vs multiple injections (101). In a randomized clinical trial that included both children and adults (n = 485) on pump vs multiple injections of insulin analogs, the rate of hypoglycemia was comparable and there was no significant weight gain (102). A double-blind, two-period crossover randomized clinical trial comparing two insulin analogs (glargine vs U100) showed a reduced rate of symptomatic hypoglycemia with the second generation U100 analog with equivalent impacts on weight (103).

The direct association between hypoglycemia, glycemic control, and weight gain has been difficult to prove, and thus additional studies are needed to understand these relationships. One reason for this gap is the lack of defined numerical thresholds for hypoglycemia due to variability in symptoms. Recent definitions have been proposed that align with clinical severity (97). Data are needed to understand how these newly proposed hypoglycemia thresholds and the subsequent compensation, if any, to temporary intake in food mediate unwanted weight gain with intensification of glucose management.

Adverse Health Outcomes of Obesity in T1D

An overwhelming amount of evidence has demonstrated that in the general population, obesity is a leading cause of death that is also associated with poorer mental health outcomes (e.g., depression), decreased quality of life, and health consequences, including cardiovascular disease, stroke, and cancer (e.g., breast, colon, kidney) (104–106). Historically, the treatment of T1D has focused on controlling hyperglycemia (and, to a lesser extent, hypertension and dyslipidemia) to reduce the risk of microvascular and macrovascular complications, including retinopathy, nephropathy, neuropathy cardiovascular disease, and peripheral vascular disease (107). The role that obesity per se plays in the onset and exacerbation of these deleterious health outcomes is poorly defined.

Several studies indicate that the common sequelae of obesity in the general population also occur in individuals with T1D (17, 108–110). Metabolic aberrations such as insulin resistance and metabolic syndrome are common, often concurrently, in obesity in the general population (111). These aberrations are now recognized to be prevalent phenotypes in patients with obesity and T1D. Insulin resistance is the lack of responsiveness to the effects of insulin in peripheral tissues, which leads to a chronic state of hyperinsulinemia (in patients with functional β-cells) and can predict development of T2D (1). Insulin resistance, assessed via the hyperinsulinemic-euglycemic clamp technique, was higher in normal-weight youth with T1D as compared with well-matched healthy controls. Interestingly, the insulin-resistant phenotype lacked the hallmark characteristics seen in individuals with obesity, that is, abnormal intramyocellular lipids, dyslipidemia, suppressed adiponectin levels, and excess adiposity. Of particular concern was the impairment of both exercise capacity and cardiac function seen in nonobese youth with T1D and insulin resistance (112). This highlights the crucial point that the obese phenotype in T1D cannot be assumed to mirror what is expected from the extensive literature in the general population and in people with T2D. A meta-analysis of 38 studies that assessed insulin sensitivity in comparison with heathy controls via hyperinsulinemic-euglycemic clamp demonstrated impaired insulin sensitivity in adults with T1D. BMI was similar in T1D and healthy controls and ranged from lean to overweight (113). These studies demonstrate that in both youth and adults, T1D is a state of insulin deficiency, where insulin resistance develops in the setting of exogenous insulin delivery, exhibits a unique phenotype, and correlates with aberrant physiological endpoints regardless of body weight. Therefore, targeting insulin resistance in T1D could be a viable therapeutic avenue to reduce complications. Indeed, in the Akita mouse model of T1D, reduced expression of myostatin, a muscle-specific protein that reduces lean muscle mass and promotes insulin resistance, is associated with preservation of lean body mass, improvements in insulin sensitivity, and improvements in glycemic control (114).

Metabolic syndrome is another health outcome associated with both obesity and T1D. It has various definitions, but includes parameters such as hyperglycemia, excess weight, central obesity, hyperlipidemia, and hypertension (115). In a study of the Pittsburgh EDC cohort (mean age 30.8 years), metabolic syndrome prevalence in T1D ranged from 8% to 21% depending on the definition used [defined based on Refs. (116–118)]. Despite this variability based on definition, major T1D outcomes such as cardiovascular disease, nephropathy, and death were higher in people with metabolic syndrome (119). Another study demonstrated the prevalence of metabolic syndrome [defined based on criteria in reference Ref. (116)] in T1D to be 31.9% (mean age 39.7 years), and these individuals had an impaired estimated glucose disposal rate that was indicative of insulin resistance. Importantly, individuals with retinopathy and neuropathy had the poorest estimated glucose disposal rate, suggesting a relationship between insulin sensitivity and microvascular complications (120). In comparison, the prevalence of metabolic syndrome in adults in the United States ≥20 years of age is 23.7% [based on criteria from Ref. (121)].

Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disorder that coexists in people with obesity and diabetes. The global estimates of NAFLD in adults indicate a prevalence of 24% (122) and it increases with age, metabolic disease, and BMI (123). For example, in adults with T2D the prevalence is estimated to be ~65% depending on population characteristics and the tool used to ascertain NAFLD (124). In children and adolescents, a meta-analysis showed a pooled mean prevalence of 7.6% in the general population and 34.2% in obesity clinics (125). The prevalence of NAFLD in T1D varies from study to study. In adults with T1D, the prevalence ranges from 12% to 52.4% (126–129). In children, it ranges from 0% in both cases with T1D and healthy controls (130) to 4.7% in children with T1D vs 13.4% in healthy controls of similar age and BMI (131). Two studies that used a similar method to assess hepatic fat (MRI) demonstrated a lower liver fat percentage in children (130) and adults (132) with T1D as compared with controls without T1D. Despite these variable rates, when NAFLD is present concurrently with T1D, it is accompanied by insulin resistance and higher rates of complications (126, 128, 129, 133–135). There have been very few studies in T1D that have applied advanced modalities that can more reliably ascertain steatosis burden and progression of disease, such as magnetic resonance elastography. To our knowledge, there have been no published studies that have employed advanced biomarker interrogations of NAFLD in T1D or that have compared the NAFLD phenotype in lean individuals vs those with obesity and T1D. Given that obesity is tightly linked with NAFLD, it is reasonable to postulate that as obesity increases in T1D, there will be a negative impact on liver health.

“The most severe defects in food intake regulation in T1D are due to impaired release of the pancreatic hormones insulin, glucagon, and amylin.”

In addition to showing an association between intensive insulin therapy and weight gain, the DCCT showed that in those with the most excessive weight gain, there were increases in both cardiometabolic risk factors (lipids, blood pressure) and more extensive atherosclerosis (17, 136). In an observational study in adults with T1D, obesity was associated with the presence and progression of coronary artery calcium, a marker of subclinical atherosclerosis (137). More recently, a study in the DCCT cohort through the 20-year follow-up period in EDIC showed that the groups on intensive insulin therapy and conventional therapy with the most weight gain did not have higher rates of cardiovascular disease or major adverse cardiovascular events at year 13 of EDIC. However, at year 14 of EDIC, the cardiovascular event curves began to diverge in the intensive insulin therapy group only through the 20-year follow-up. A Cox proportional hazard model showed that after year 14, the cardiovascular events in the intensive insulin therapy group with the most weight gain were significantly higher than the minimal weight gain group (P = 0.024; unadjusted hazard ratio 1.99; 95% CI, 1.12 to 3.63), with the event rate becoming indistinguishable from the conventional group (Fig. 1). This difference did not remain after full adjustment for cardiovascular risk factors and medications used to mitigate risk. There was no difference in the rate of major adverse cardiovascular events for the entire follow-up period in either group. Several possible reasons for these somewhat divergent findings were presented, the most compelling being that in the general population, despite an increase in obesity, the rates of cardiovascular disease have decreased, presumably due to more aggressive risk factor management (62). These studies demonstrate a strong possibility that obesity in T1D may have a detrimental impact on cardiovascular disease in at least a subset of individuals.

Weight gain impacts cardiovascular event rates after 14 years of follow-up in the EDIC trial. (a) Cumulative incidence during the EDIC study of the first occurrence of any cardiovascular disease (P = 0.21, log-rank test) and (b) nonfatal myocardial infarction, stroke, or death from cardiovascular disease (major adverse cardiovascular events), comparing the group with excessive weight gain group [fourth quartile of weight gain, n = 152 (red solid lines)] with the group with minimal weight gain [first through third quartiles combined, n = 457 (blue solid lines)] in the intensive insulin therapy group and with all participants in the conventional therapy group [n = 609 (green dotted lines)]. Using a Cox proportional hazards model, the unadjusted difference in hazards after year 14 of EDIC follow-up between the excessive and minimal weight gain groups receiving intensive insulin therapy was significant for the first occurrence of cardiovascular disease (P = 0.024). CONV, conventional therapy; INT, intensive insulin therapy. Reproduced from Purnell JQ, Braffett BH, Zinman B, et al. Impact of excessive weight gain on cardiovascular outcomes in type I diabetes: results from the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study. Diabetes Care. 2017;40(12):1756–1762.
Figure 1.

Weight gain impacts cardiovascular event rates after 14 years of follow-up in the EDIC trial. (a) Cumulative incidence during the EDIC study of the first occurrence of any cardiovascular disease (P = 0.21, log-rank test) and (b) nonfatal myocardial infarction, stroke, or death from cardiovascular disease (major adverse cardiovascular events), comparing the group with excessive weight gain group [fourth quartile of weight gain, n = 152 (red solid lines)] with the group with minimal weight gain [first through third quartiles combined, n = 457 (blue solid lines)] in the intensive insulin therapy group and with all participants in the conventional therapy group [n = 609 (green dotted lines)]. Using a Cox proportional hazards model, the unadjusted difference in hazards after year 14 of EDIC follow-up between the excessive and minimal weight gain groups receiving intensive insulin therapy was significant for the first occurrence of cardiovascular disease (P = 0.024). CONV, conventional therapy; INT, intensive insulin therapy. Reproduced from Purnell JQ, Braffett BH, Zinman B, et al. Impact of excessive weight gain on cardiovascular outcomes in type I diabetes: results from the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study. Diabetes Care. 2017;40(12):1756–1762.

More fundamentally, there is some evidence that obesity can influence the age at onset of both T1D and T2D. Increased visceral fat over time is correlated with increased risk of developing T2D (138). Evidence to support a similar relationship in T1D has been reported in both animal models and humans. For example, in male rodents only, a genetic background permissive of obesity in combination with a high-fat diet leads to the development of a T1D-like phenotype that is characterized by mononuclear cell infiltration and insulitis (139). Children who are obese with new-onset T1D display a pattern of cytokines and adipokines that suggest a proinflammatory state, making it plausible that this phenotype contributed to T1D onset (140). A meta-analysis of nine studies found evidence of an association between childhood obesity and subsequent risk of diabetes with an OR of 1.25 and 2.03 in studies reporting obesity as a continuous or categorical variable, respectively (141). BMI before and after diagnosis of T1D was found to be inversely associated with age at diagnosis in a cohort of 168 youth whose weights were recorded from birth. BMI was found to be higher than in the general population, although birth weights were similar to the general population (142). In adolescents, central obesity is correlated with earlier onset of T1D (143). Furthermore, the Diabetes Prevention Trial–Type 1 generated an algorithm that predicted T1D risk among relatives with autoimmune prediabetes. That score included BMI, along with age, C-peptide, and oral glucose tolerance test indexes (144). Obesity might also predict T1D progression. For example, a model that includes BMI, immunological markers, and age predicts worsening of T1D as defined by reduced residual C-peptide after 1 year of follow-up [area under the curve (AUC) of 0.936]. Furthermore, a combination of metabolic and immune markers could differentiate healthy controls, individuals at high-risk for T1D, and patients with T1D (145). These findings suggest that the metabolic and proinflammatory state that is characteristic of obesity and insulin resistance could be an important risk factor for long-term health outcomes or a biomarker for early detection.

The relationship between obesity and T1D onset is controversial. Contrary to the evidence presented above, a case-control study showed that birth weight was higher in cases, yet there was no excessive weight gain prior to diagnosis (146). In a study of 777 children from the BABYDIAB cohort, insulin sensitivity (assessed by homeostatic model assessment of insulin resistance) and BMI were not different in autoantibody-positive vs -negative children (147). In the Trial Net Pathway to Prevention study, although more children with obesity were found to be positive for a single autoantibody than were nonobese children, there was no difference in BMI in children who transitioned from single autoantibody to multiple autoantibodies or in those who eventually developed T1D vs those who did not (148). In a cross-sectional study of children with newly diagnosed T1D, levels of C-peptide were higher in children who were overweight and obese than with lean children (149). Within the SEARCH cohort there was an inverse association between BMI and age at diagnosis seen only in children with fasting C-peptide levels below the median (150). Given the multifactorial and complex etiology of T1D, it is not surprising that the data are controversial. More studies are needed to define the subset of individuals for whom obesity could be an accelerator so that weight management can be a prioritized in care plans.

Overall, it is clear that at least some of the relationships between obesity and the health of individuals with T1D are consistent with what is seen in the general population. The mechanisms mediating these relationships, assessment of causality, and the best therapeutic interventions to use within the context of intensive insulin therapy and progression of T1D are incompletely understood. Direct, longitudinal studies in lean and individuals with obesity and T1D, in comparison with well-matched healthy controls, are needed to understand the impact of the adverse health effects of obesity on T1D initiation and outcomes.

Physiology of Energy Balance in T1D

Weight gain occurs due to an imbalance between energy intake and expenditure. Under physiological conditions, homeostatic regulatory processes balance energy intake and expenditure to promote weight stability (151). Factors such as body composition (152, 153), appetitive behaviors (154), macronutrient balance (155), metabolic flexibility (156), energy losses in fecal matter and urine (157, 158), gut microbiota composition and function (157), gastric emptying, gut transit time (159), changes in size of metabolically active organs (160, 161), and multiorgan communication via the enteroendocrine system (162) differentially affect energy balance, and relationships are often bidirectional (163). The sections below summarize the current knowledge on the physiology and mechanisms that influence the energy balance equation, which have been more extensively reviewed elsewhere (151–153, 164–166), with an emphasis on how these mechanisms relate to obesity in T1D.

Energy intake and satiety pathways

The systems that control food choices and the physiological responses to food intake are complex and can contribute to energy imbalance when dysregulated (165). The relationship between T1D and energy intake depends on physiological, environmental, and psychological factors. In the SEARCH for Diabetes in Youth study, energy intake did not differ in youth with T1D vs T2D: mean calorie intake was 1925 kcal in those 10 to 14 years of age and 2057 kcal in those ≥15 years of age (167). These levels of intake are similar to those reported in the 2009 to 2010 NHANES, which revealed a trend for increased caloric intake in adolescents that could contribute to an increase in obesity in youth (168). The current ADA recommendations for energy intake for people with diabetes focus on achieving and maintaining a healthy weight and reaching glycemic goals (169). To adapt those guidelines to the current trends of obesity in T1D requires focused investigations to determine energy requirements as insulin therapy is modified, novel therapeutics are implemented into health care paradigms, glycemic control deteriorates/normalizes, or weight is lost/gained.

Energy is not the only component that is relevant in T1D with regard to dietary intake. Many studies demonstrate that overall, individuals with T1D do not meet dietary guidelines for macronutrient intake (167, 170). On average, people with T1D consume 45% of calories from carbohydrates (26, 171), which is less than the general US population intake of 48.7% in males and 49.9% in females (172). There are currently no specific recommendations for an optimal pattern of macronutrient distribution for people with diabetes. The expert consensus reported in the 2018 ADA Guidelines for Lifestyle Management states that macronutrient patterns need to be individualized (24). The International Society for Pediatric and Adolescent Diabetes guidelines reiterate the importance of individualized macronutrient distribution and suggest a reasonable range of intake to be: carbohydrate 50% to 55% of energy, fat <35% of energy (saturated fat <10%), and protein 15% to 20% of energy (173). Optimizing macronutrient intake to normalize fuel oxidation and enhance thermogenesis is an area that requires focused research attention, as it could be a means to modulate glycemic control and energy expenditure.

The dietary intake patterns described above can influence or be influenced by the biological homeostatic mechanisms that control satiety and appetite. When, why, what, and how much one chooses to eat are behaviors regulated by a multifaceted set of pathways within the gut/brain axis, with additional controls exerted by peripheral tissues and social cues. These pathways elicit a complex set of reactions, involving hormones, signaling molecules, neurotransmitters, enzymes, and receptors that influence the gastrointestinal system, metabolism, and behavior (165). Within the gastrointestinal system, gut distention, chemical effects of food on the intestine, and release of hormones and neurotransmitters occur in parallel and converge to lead to the desired outcome of satiation. Gut and systemically derived molecules such as insulin, glucagon, GLP-1, oxyntomodulin, cholecystokinin (CCK), peptide tyrosine-tyrosine (PYY), and ghrelin play key roles in regulating appetite and food intake. Of these, only ghrelin is orexigenic, which means there is considerable redundancy in mechanisms to reduce food intake (Table 1). Eating behaviors are also impacted by the interaction of gut hormones, adipokines such as leptin, and insulin with neuronal circuitry in the arcuate nucleus of the hypothalamus. Within this brain region, the neuropeptides proopiomelanocortin and neuropeptide Y interact with defined sets of neurons to inhibit or stimulate appetite, respectively (162, 165). Additionally, the insulinotropic effect of the incretin hormones GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) is important for glycemic control and satiety (174, 175). This “incretin effect,” defined as an increase in insulin secretion after an oral vs an intravenous glucose load, is defective in T2D (176), and perhaps to a lesser extent in T1D (177, 178). Given that insulin is an important satiety signal, it stands to reason that these pathways are relevant for both T1D and obesity. The impact of these complex interactions is an active area of investigation, and mechanisms are not fully elucidated (179).

Table 1.

Key Enteroendocrine Hormones That Regulate Food Intake Through Central Nervous System Interactions

Enteroendocrine HormoneTissue of OriginImpact on Appetite
GhrelinGastrointestinal tract
AmylinPancreas
CholecystokininGastrointestinal tract
GIPGastrointestinal tract
GLP-1Gastrointestinal tract
InsulinPancreas
LeptinAdipose
OxyntomodulinGastrointestinal tract
Pancreatic PeptidePancreas
PYYGastrointestinal tract
Enteroendocrine HormoneTissue of OriginImpact on Appetite
GhrelinGastrointestinal tract
AmylinPancreas
CholecystokininGastrointestinal tract
GIPGastrointestinal tract
GLP-1Gastrointestinal tract
InsulinPancreas
LeptinAdipose
OxyntomodulinGastrointestinal tract
Pancreatic PeptidePancreas
PYYGastrointestinal tract

Adapted from Begg DP, Woods SC. The endocrinology of food intake. Nat Rev Endocrinol. 2013; 9:584; and Cummings DE, Overduin J. Gastrointestinal regulation of food intake. J Clin Invest. 2007; 117:13–23.

Table 1.

Key Enteroendocrine Hormones That Regulate Food Intake Through Central Nervous System Interactions

Enteroendocrine HormoneTissue of OriginImpact on Appetite
GhrelinGastrointestinal tract
AmylinPancreas
CholecystokininGastrointestinal tract
GIPGastrointestinal tract
GLP-1Gastrointestinal tract
InsulinPancreas
LeptinAdipose
OxyntomodulinGastrointestinal tract
Pancreatic PeptidePancreas
PYYGastrointestinal tract
Enteroendocrine HormoneTissue of OriginImpact on Appetite
GhrelinGastrointestinal tract
AmylinPancreas
CholecystokininGastrointestinal tract
GIPGastrointestinal tract
GLP-1Gastrointestinal tract
InsulinPancreas
LeptinAdipose
OxyntomodulinGastrointestinal tract
Pancreatic PeptidePancreas
PYYGastrointestinal tract

Adapted from Begg DP, Woods SC. The endocrinology of food intake. Nat Rev Endocrinol. 2013; 9:584; and Cummings DE, Overduin J. Gastrointestinal regulation of food intake. J Clin Invest. 2007; 117:13–23.

The mechanisms controlling satiety and appetite in diabetes and obesity are dysfunctional and involved in the pathobiology of disease. Defective communication circuits between the gut and brain have been implicated in the pathogenesis of obesity and T2D (180). Although the data are limited in T1D, a comprehensive assessment of gut peptides in children with T1D (age at diagnosis 6.87 ± 2.47 years; duration of diagnosis 6.5 ± 2.22. years), levels of ghrelin and amylin were reduced, GIP was upregulated, and GLP-1 and PYY were unchanged as compared with healthy controls (181), indicating that there is dysregulation of enteroendocrine signaling in T1D. The sections below highlight the canonical functions of a subset of satiety hormones with an emphasis on how they are regulated in T1D.

Ghrelin and its isoforms acyl-ghrelin, des-acylghrelin, and obestatin are enteroendocrine hormones that also modulate food intake and insulin responsiveness. The ultimate actions of these hormones are to stimulate food intake (182). Plasma ghrelin concentrations are reduced in obesity (183) and insulin resistance (184, 185). Ghrelin levels are reduced in adults (186) and children (187) with T1D. Ghrelin levels have been shown to decrease (188) or have no response (187) to insulin treatment. There is an inverse relationship between ghrelin and BMI in T1D (187, 188), which is consistent with the reduced ghrelin seen in obese populations (183, 186).

Upon central nervous system activation in response to a meal stimulus, the incretin hormone GLP-1 regulates body weight by diminishing appetite and delaying gastric emptying (174) in concert with the actions of PYY and CCK on gastric emptying and satiety (162, 189, 190). In the proinflammatory, insulin-resistant state that is common in obesity and T2D, the incretin effect is decreased as a result of increased degradation of GLP-1 by the enzyme DPP-4 (191). GLP-1 is also a key mediator for the regulation of glucose homeostasis (192). This is exemplified in bariatric surgery patients. The weight loss and T2D resolution that can occur after bariatric surgery are accompanied by restoration of the function of several enteroendocrine molecules, including GLP-1, ghrelin, and bile acids (193, 194). The functional profile of GLP-1 in T1D is not well characterized, and levels of GLP-1 are similar to healthy controls (181). There is no difference between healthy controls and T1D cases with respect to the postmeal rise in GLP-1 (195).

The most severe defects in food intake regulation in T1D are due to impairments in release of the pancreatic hormones insulin, glucagon, and amylin (55, 196). Insulin regulates both appetite and body weight via central nervous system interactions in response to circulating nutrients and in proportion to level of adiposity (197, 198). Insulin, in conjunction with leptin, regulates energy balance by minimizing the impact of acute changes in the flux of energy. This occurs via a balancing of anabolic and catabolic hypothalamic controls [reviewed in Refs. (151) and (165)]. A small amount of circulating insulin reaches the brain. Because insulin is secreted in proportion to adiposity, its role in the brain is to activate a negative feedback loop to adjust food intake in the direction that will promote body weight homeostasis. Importantly, similar populations of neurons in the hypothalamus control both food intake and the impact of pancreatic hormones on glucose homeostasis (198). Exogenous insulin is given in response to anticipated food intake and the expected amount needed to normalize glucose, which bypasses the endogenous control of insulin release in response to adiposity and the signals generated upon a meal stimulus. Additionally, when exogenous insulin leads to hypoglycemia, there is an increased tendency to eat, which negates the expected reduction on food intake from a stronger insulin signal in the brain (198).

Under physiological conditions amylin is cosecreted with insulin from pancreatic β-cells in response to a meal stimulus and functions to decrease food intake, suppress glucagon secretion, regulate body weight, and increase energy expenditure (199). In obesity, insulin resistance, and T2D, there is a dampened postmeal release of amylin. In T1D, there is a dual defect: levels are markedly lower than in the general population and do not respond to a meal stimulus. Because of this, the homeostatic controls on food intake are dramatically impaired (200).

During fasting conditions, glucagon is released from α-cells to promote satiety. This satiety response appears to be preserved in T1D but not in obesity (186). Intramuscular glucagon administration decreases hunger in lean individuals and those with T1D, but not in individuals with obesity. This experimental hyperglucagonemia leads to a decrease in hunger, coincident with a reduction in ghrelin levels, in T1D and healthy controls only (186). Given this discordance between the effects of glucagon on hunger in T1D/lean individuals vs those with obesity, it is unclear how dysregulation of ghrelin secretion will impact hunger when obesity and T1D are concurrent phenotypes.

In addition to the more canonical pathways of food intake regulation, bile acids are well recognized for their important endocrine functions. They correlate positively with BMI, and evidence is emerging on their role in food intake via effects on eating behaviors (201) and interactions with gut hormones such ghrelin and GLP-1 (202, 203). The bile acid pathway is under active investigation as a mediator of the metabolic aberrations in obesity and T2D (194, 204, 205). The role of bile acids on obesity in T1D has not been investigated, particularly with respect to a dual role in glycemic control and energy homeostasis.

Beyond the role of the enteroendocrine system in controlling energy intake, fundamental defects in gut function have been linked to T1D. For example, PYY, which is released from the gut in response to food intake to suppress appetite, is also important for islet cell development and in regeneration of pancreatic cells (206). Suppression of the bactericidal function of intestinal Paneth cells was identified in the streptozotocin (STZ) mouse model of T1D, which could have deleterious effect on innate immunity (207). This dysfunction was reversed upon insulin treatment, suggesting a dual role for insulin in glycemic control in gut health. The importance of the gut and its released peptides as therapeutic modalities was elegantly demonstrated in both cell-based and mouse experiments where ablation of forkhead box O (Foxo)1 gave rise to insulin-producing cells in the gut that displayed some of the characteristics of β-cells (208). This concept of reprogramming the gut to restore insulin production has been demonstrated in several other studies (209–211), highlighting the unique role of a healthy gut for T1D therapeutics.

Given the aberrant functionality the gastrointestinal hormones in obesity, T1D and T2D could impact appetitive behavior, energy intake, and body weight. It is not known how the effects of obesity on the homeostatic controls that regulate food intake and satiety impact health and metabolism in individuals with T1D. Given the negative impact of obesity on gut health, preventing and treating obesity aggressively in people with T1D, or in those at risk for developing it, could be a critical determinant of long-term health outcomes.

Energy expenditure

The main component of energy expenditure is resting energy expenditure, which accounts for ~55% to 70% of total daily energy expenditure. Energy expended by daily activities (~30%) and the thermic effect of food (~10%) are the major remaining components of energy expenditure (212). Energy expenditure is influenced by a number of biological factors, including age, daily activity, and body composition, particularly fat-free mass (153). There is interindividual variability in energy expenditure, and the specific mechanisms contributing to this variability are the subject of intense study (152, 153). T1D is a catabolic state, particularly in insulin deficiency (213). Standard equations for estimating energy expenditure and dietary intake requirements in healthy individuals are not adequate for those with T1D. First, in patients with uncontrolled T1D, glycosuria can account for 300 to 400 kcal/d in obligate energy losses (158). Second, most studies demonstrate a modest increase in resting energy expenditure (range, 100 to 300 kcal/d) in people with T1D relative to healthy controls or predicted values (158, 214–217). A study that manipulated glucagon levels via insulin deprivation/replacement experiments revealed that the resting energy expenditure increase in people with T1D appears to be proportionate to the degree of hyperglycemia and attributed to increased hepatic gluconeogenesis driven by hyperglucagonemia (218). One possible explanation for this is that in the absence of insulin, excess glucagon leads to gluconeogenesis and increased protein turnover, resulting in a net negative energy balance (218–220). There are limited data available on the impact of systemic vs portal insulin delivery on energy expenditure. One study in a canine model of T1D demonstrated that 24-hour energy expenditure (assessed by ventilated hood) is higher with portal vs systemic insulin administration (221). Other factors, including increased sympathetic nervous system activity (222), increased protein turnover and substrate transport across cellular membranes (218), alterations in the gut microbiome (223), and increases in size of metabolically active organs (224), may contribute to alterations in energy expenditure in T1D.

Very few studies have been conducted to fully understand the energy balance equation in T1D. Most of the available literature dates back several decades. Initial studies showed conflicting evidence regarding energy balance status in T1D. In a controlled metabolic ward study, basal energy expenditure was higher than predicted in the absence of insulin and was restored to predicted values with insulin replenishment in poorly controlled individuals with T1D (n = 10) (217). In individuals with poorly controlled T1D, resting metabolic rate (assessed via ventilated hood) decreased, the thermic response to high-fat overfeeding and norepinephrine infusion was blunted, and weight increased by an average of 3.5 kg after changing from conventional therapy to intensive insulin therapy (225). In one of the few studies conducted in individuals with T1D using whole-room calorimetry, lean individuals with well-controlled T1D had significantly lower diet-induced thermogenesis compared with well-matched healthy controls. This dampened thermic effect of food was inversely correlated with 24-hour glycemia, suggesting that that the energy cost of thermogenesis may be reduced, at least in part, by hyperglycemia. There were no significant differences on other energy and substrate oxidation profiles including 24-hour energy expenditure and respiratory quotient (a measure of whole-body substrate oxidation) between groups (216). The energy expenditure finding corroborates what was seen in a previous study where the increased energy expenditure in poorly controlled T1D normalized after initiation of insulin therapy (217). The energy expenditure during exercise was also lower in the T1D group, but the net work efficiency was not, meaning that the response to exercise was similar (216). A recent crossover design study comparing the effects of insulin glargine with peglispro, a novel hepatoselective insulin, in 15 patients with T1D demonstrated a small (80 kcal/24 h) increment in 24-hour energy expenditure with peglispro, accompanied by increases in nocturnal lipid oxidation rates (226). Additional studies are needed to understand the energy expenditure profile in T1D across the spectrum of body weight and glycemic control to inform calorie prescriptions to achieve weight goals. A theoretical model of the impacts of glycemic control on the components of energy expenditure is shown in Figure 2.

Theoretical model of the currently known impacts of glycemic control on energy balance. The largest component of the energy balance equation is resting energy expenditure (REE), of which the largest constituent is sleep energy expenditure (SEE). It accounts for 50% to 70% of total daily energy expenditure. The remaining major components of energy expenditure are thermic effect of food (TEF) and activity energy expenditure (AEE). AEE includes exercise, activities of daily living (ADL), work, and fidgeting. There are multiple other components that can influence energy expenditure differentially, including the gut microbiome, energy losses in urine, stool, or volatile organic compounds, thermogenesis, changes in energy expenditure from metabolically active organs (brain, kidney, liver, muscle, heart), and sympathetic nervous system activity. Achievement of well-controlled T1D due to insulin intensification reduces urinary glucose losses, REE, and TEF but does not affect food intake, resulting in a net positive energy balance and weight gain. Uncontrolled T1D is associated with an increase in resting energy expenditure due to increases in metabolically active organ weights and substrate cycling, as well as glucosuria resulting in a net negative energy balance (due to an overall increase in 24-hour energy expenditure) and weight loss.
Figure 2.

Theoretical model of the currently known impacts of glycemic control on energy balance. The largest component of the energy balance equation is resting energy expenditure (REE), of which the largest constituent is sleep energy expenditure (SEE). It accounts for 50% to 70% of total daily energy expenditure. The remaining major components of energy expenditure are thermic effect of food (TEF) and activity energy expenditure (AEE). AEE includes exercise, activities of daily living (ADL), work, and fidgeting. There are multiple other components that can influence energy expenditure differentially, including the gut microbiome, energy losses in urine, stool, or volatile organic compounds, thermogenesis, changes in energy expenditure from metabolically active organs (brain, kidney, liver, muscle, heart), and sympathetic nervous system activity. Achievement of well-controlled T1D due to insulin intensification reduces urinary glucose losses, REE, and TEF but does not affect food intake, resulting in a net positive energy balance and weight gain. Uncontrolled T1D is associated with an increase in resting energy expenditure due to increases in metabolically active organ weights and substrate cycling, as well as glucosuria resulting in a net negative energy balance (due to an overall increase in 24-hour energy expenditure) and weight loss.

Metabolic flexibility

Metabolic flexibility is defined as the ability of an organism to rapidly shift substrate oxidation rates to accommodate changes in substrate availability due to dietary inputs, energy demands, environmental changes, and biological signals (227). Notably, an inability to switch fuel oxidation in response to dietary macronutrients and metabolic demands (such as fasting) is a central component of both optimal glycemic control and weight regulation (228, 229). Metabolic inflexibility has been demonstrated in individuals who are obese, with and without T2D (166, 230). In newly diagnosed individuals with well-controlled T2D, metabolic flexibility is one key determinant of insulin sensitivity (231). Although most research in diabetes has focused on the central role of dysregulated carbohydrate metabolism, lack of homeostatic control of lipid synthesis and oxidation are also critically important (232). The mechanisms that control metabolic flexibility in healthy and metabolically compromised individuals are the subject of intense study.

T1D is characterized by marked differences in substrate oxidation rates with an increase in lipid oxidation rates during basal conditions, a blunted ability to shift to carbohydrate oxidation during meals, and a decrease in the thermic effect of food compared with controls without T1D (158, 214–218). In the insulin-deficient STZ mouse model of T1D, treatment with bezafibrate, a peroxisome proliferator-activated receptor pan-agonist, results in an improvement in insulin sensitivity, metabolic flexibility, and liver physiology (233). A mouse model of maturity-onset diabetes of the young generated by ablation of the three Foxo genes displays metabolic inflexibility characterized by preferential oxidation of lipids. This inflexibility leads to impaired insulin secretion (234). Recently, the concept of β-cell dedifferentiation has revealed that there are windows of opportunity for preserving β-cell function. It is thought that an early event in the decline of β-cell action is metabolic inflexibility (235). It is not known whether whole-body metabolic flexibility interacts with β-cell metabolic flexibility. Importantly, the health implications of obesity plus T1D in this paradigm, including the potential connection between obesity, insulin resistance, and progression/age of onset of T1D, have not been studied.

In sharp contrast to the wealth of evidence about energy metabolism in obesity and T2D, there have been virtually no published randomized clinical trials to systematically, comprehensively, and precisely study energy balance and metabolic flexibility as insulin therapy is modified to achieve glycemic control. In one of the studies reviewed above, an increase in metabolic flexibility assessed in response to a high carbohydrate breakfast meal was noted in patients with T1D during treatment with peglispro compared with glargine (226). Collectively, this literature suggests that studies are needed to target various aspects of the energy balance equation and metabolic inflexibility in T1D through lifestyle or medical interventions to understand the impact on glycemic control, weight regulation, and overall health.

Potential Mechanisms Modulating the Obese Phenotype in T1D

Obesity is a complex phenotype that involves many pathophysiological pathways. The causes of obesity are both biological and environmental in nature (236). With the possible exception of genetics (237, 238), the mechanisms thought to be causal for obesity can also be modulated by the obese phenotype. The sections below provide a framework for how many of the key mechanisms thought to be important in obesity causality and pathophysiology are involved in non-T1D obesity, describe the literature related to these mechanisms in T1D (if any), and use the currently available knowledge to highlight the gaps that need to be addressed to prevent and treat obesity in people with T1D.

Genetics

The genetic underpinnings of obesity and T2D and how these two metabolic diseases could have shared genetic control points have been a subject of intense study (239–243). The combined effect of genetic variants associated with both diseases on pathogenesis is poorly understood and points toward few shared genetic features but many shared mechanistic pathways (244). Indeed, even the well-studied genetic variants in fat mass and obesity-associated protein (FTO) may not be the causal variants associated with obesity and T2D, but perhaps instead (or additionally) there are roles for neighboring genes (245). This suggests there is still much work that needs to be done to harness the power of genetics for predicting disease risk and informing on therapeutic options.

The genetics of T1D have also been intensively studied, particularly with respect to alleles that increase susceptibility (246). The data with respect the genetics of obesity and T1D are emerging. In the DCCT, it was demonstrated that individuals with T1D on intensive insulin therapy with a family history of T2D gained more weight than did those without a family history. Weight gain was similar on conventional therapy, regardless of family history (100). This implies that a genetic predisposition to T2D may interact with the environmental input of glycemic control to influence the phenotypic outcome of obesity in T1D.

Some of the genes that are relevant for obesity and T2D are connected to T1D. As discussed earlier, one key pathway in the regulation of energy balance is the central melanocortin system where a tightly orchestrated set of signals converges on the arcuate nucleus to regulate food intake and satiety [reviewed in Ref. (247)]. Three well-studied examples of genes within this pathway that are associated with obesity are FTO (248, 249), melanocortin 4 receptor (MC4R) (250, 251), and proopiomelanocortin (252). These genes are tied to neuronal controls of appetite and eating behaviors, which points to a biologically plausible connection to obesity (247). In a cohort of 1119 children with T1D, the association between BMI and known obesity susceptibility genes was studied. Polymorphisms in multiple genes were tested, including FTO and MC4R. Only the A allele of rs9939609 in the FTO gene was associated with higher BMI in T1D (253). The TCF7L2 gene has been associated with both obesity and T2D, but one study found no association with T1D incidence (254). However, in a cohort of 810 individuals with T1D, carriers of the T allele at the rs4506565 locus of TCF7L2 were more likely to have only a single autoantibody present, higher C-peptide AUC, and lower glucose AUC during an oral glucose tolerance test after adjustment of several parameters, including BMI z score, suggesting that this gene contributes to a distinct T1D phenotype with milder immune and metabolic characteristics (255). These studies suggest that genetic screening for obesity in T1D may allow for stratification of the individuals at the highest risk for developing obesity (FTO) or more advanced disease (TCF7L2).

In addition to the overlap between genetics of T1D, T2D, and obesity, unique relationships in T1D have been identified. Given the challenges in establishing the causality of genetic variants on disease phenotypes due to the effects of confounding factors or reverse causality, Mendelian randomization approaches have emerged to exploit the random distribution of alleles at meiosis to essentially implement randomization based on normal distributions of alleles in populations. The alleles serve as instrumental variables, and the success of these methods depends on the strength of the association between the genetic variants and the phenotype of interest (256, 257). This method was used to test the role of childhood adiposity on T1D etiology using 23 single nucleotide polymorphisms (SNPs) as instrumental variables. The SNPs were initially selected from a previous genome-wide association study on SNPs associated with childhood adiposity. A set of additional criteria and validation steps was implemented to select the final set of 23 SNPs. They found that genetic risk for higher childhood BMI increased the risk of T1D by 32% (OR 1.32; 95% CI, 1.06 to 1.64) per SD score of BMI (256). Additionally, Mendelian randomization analysis with a genetic risk score comprised of 30 validated BMI loci showed a U-shaped relationship between BMI over lifespan and diabetic kidney disease, microalbuminuria, and end-stage renal disease in a cohort of 6049 individuals with T1D (257). This suggests that being underweight or overweight impacts risk of complications.

Inflammation is a phenotype that is prominent in both obesity and T1D (258). Although the direct connection between these three phenotypes has not been studied, there are several lines of evidence that connect risk alleles for T1D and inflammation. The most fundamental genetic connection between T1D and inflammation is the variation in human leukocyte antigen (HLA) genes, which are part of the major histocompatibility complex and key regulators adaptive immunity. These alleles account for about half of the genetic risk in T1D (259). TNF-induced protein 3 (TNFAIP3) is a susceptibility locus for T1D, and variation in this gene (at the rs2327832 locus) is associated with lower stimulated C-peptide and higher HbA1c 12 months after diagnosis (260). Loss-of-function polymorphisms in tyrosine kinase 2 areassociated with an increased risk of T1D. Several cell-based experiments to dampen tyrosinekinase 2 function demonstrate antiapoptotic and anti-inflammatory effects in β-cells (261). Based on these data, it is possible that the proinflammatory phenotypes of obesity and T1D could synergize to negatively impact outcomes. The combined effects of susceptibility variants for obesity and T1D, within the context of inflammation, could provide valuable insight into disease risk and severity.

The genetic underpinnings of T1D risk are, with a few notable exceptions, distinct from T2D and obesity. Although it is reasonable to postulate that the genetic defects that promote obesity susceptibility might coexist with genetic variants known to be risk factors for T1D, how these concurrent genetic aberrations will impact the health of individuals with T1D is unclear. Studies are needed to compare the genetic risk factors for T1D in lean individuals vs those who are obese. Whether genetic susceptibility to obesity or T2D interacts with the genes known to be causal for T1D has not been reported in the literature. Therefore, there are many gaps in knowledge that prevent the application of genetics for disease prediction and therapeutics, including limited data in multiethnic groups. These foundational hurdles need to be overcome before the connection between genetics, obesity, and T1D can be elucidated.

Epigenetics

Genetics is an essentially static biological fingerprint that exerts substantial control over metabolic disease susceptibility and pathophysiology. Epigenetics, in contrast, is a dynamic set of modifications to DNA and histones that can have profound effects on gene expression. Epigenetic modifications integrate environmental cues throughout the lifespan and modify biological pathways in response to the actual or expected environment. These modifications impact obesity, T2D (262), and T1D (263) susceptibility and pathophysiology. A critical window of time where these modifications are established is in utero through the early postnatal period.

In utero exposures to obesity and diabetes have been shown to affect fetal development and risk for future metabolic disease, including obesity, metabolic syndrome, and T2D (264–266). For example, maternal nutritional status, both overnutrition and undernutrition, and maternal weight have been associated with increased risk for obesity in offspring (265, 267–269). Specific nutritional components can also influence offspring health. The agouti viable yellow mouse model displays a phenotype of increasing weight and yellowing coat color as the agouti viable yellow locus becomes increasingly demethylated. Hypermethylation represses the normal agouti signaling functions, which normally promotes a yellow color and suppresses satiety signaling via MC4R. When females with obesity are weaned onto a hypermethylating methyl-supplemented diet, the transgenerational amplification of the obese phenotype to subsequent generations of offspring does not occur as it does in females weaned to a control diet (270). Similar to T2D and obesity, there is a relationship between in utero exposure to maternal T1D and future health of the offspring, including increased risk of metabolic disease (271), increased adiposity and adipocyte dysfunction (272, 273), altered concentrations of metabolic hormones such as leptin (271, 272) and ghrelin (274), and future metabolic disease, including prediabetes and T2D (266, 275). The risk of T1D is higher in children born to fathers vs mothers with T1D due to differential inheritance of risk alleles, maternal vs paternal imprinting (an epigenetic phenomenon), and reduced risk of islet autoimmunity in the first year of life when the mother has T1D (276). The increased risk of T1D in children of mothers who are overweight or obese is confined to mothers that are nondiabetic (277, 278).

The early postnatal period is also a critical window of exposure that impacts metabolic disease. In rats, an overfeeding paradigm during the early postnatal window created by dividing offspring into small litters (overfed), normal litters (normal feeding), and large litters (underfeeding) led to obesity, hypertension, and hyperinsulinemia in adulthood. Furthermore, these overfed adult rats developed a T1D-like phenotype, characterized by hyperglycemia, glucosuria, and ketonuria on a low dose of STZ (279). Another study, also in rats, showed that postnatal overfeeding led to reduced glucose-stimulated insulin secretion (GSIS) at weaning that persisted into adulthood. This was recapitulated in isolated islets. Although glucose tolerance was normal, the work shows the importance of postnatal nutrition on metabolic responses that have relevance to T1D (280). In female mice, early postnatal overnutrition reduced energy expenditure, in part due to reduced physical activity. This was associated with higher body weight and fat mass that persisted into adulthood (281). Given the close connection between the brain and the gut in modulating eating behaviors and energy balance, these mechanisms could prove to be relevant for obesity in both T1D and T2D. Therefore, the window between in utero development and the early postnatal period is a critical time frame that connects nutritional and environmental factors with future health outcomes of offspring.

The mechanisms driving the early programming of metabolic disease are incompletely understood, but there is abundant evidence pointing toward epigenetics [reviewed in Ref. (282)]. The changes in GSIS due to postnatal overfeeding in rats described above were accompanied by persistent changes in gene expression that were likely due to epigenetic mechanisms (280). There are multiple types of epigenetic modifications, each with unique mechanistic consequences. Perhaps the most common type of epigenetic modification is DNA methylation, which is most often associated with gene silencing. The changes in energy expenditure, physical activity, body weight, and fat mass due to postnatal overnutrition in rats described in the previous paragraph were accompanied by modest alterations in hypothalamic methylation at genes that are relevant for neural development or function. These changes in methylation persisted into adulthood (281). It is plausible that these epigenetic marks were responsible for the subsequent metabolic phenotype given the neurobiological regulation of physical activity (283). Leptin (284) and adiponectin (285), adipokines that mediate both insulin action and adiposity, have been shown to be differentially methylated based on glycemic status of the mother. Leptin gene methylation was assessed in placental tissues in normal vs impaired glucose-tolerant females. In the impaired glucose-tolerant females, reduced leptin methylation on the maternal-facing side of the placenta was negatively correlated with maternal 2-hour glycemia during a glucose tolerance test (284). Lower methylation of the adiponectin gene promoter in both the fetal- and maternal-facing placental tissue was associated with compromised glycemic control in the mother (285). It is conceivable that aberrant methylation of leptin and adiponectin could predispose offspring to metabolic disease and that this will hold true in individuals with obesity and T1D.

“Metabolic flexibility is defined as the ability of an organism to rapidly shift substrate oxidation rates.”

As described earlier, genetic variation in the FTO locus is associated with obesity, T1D, and T2D. Another regulatory mechanism that modulates the impact of FTO on metabolic disease is epigenetics. In adults with T2D, methylation of the FTO obesity susceptibility haplotype (tagged by SNP rs8050136) was increased (286). FTO expression was not analyzed because there has not been a correlation between FTO SNPs and expression levels (287), making causality difficult to ascertain. FTO methylation changes in lean individuals vs those with obesity and T1D have not been reported. Despite the lack of data on the role of methylation on the expression of metabolic regulators in T1D, there have been unique methylation mechanisms uncovered in T1D. For instance, in nonobese diabetic (NOD) mice, progression of T1D correlated with changes in methylation of insulin DNA in β-cells that was inversely associated with gene transcription. This methylation was induced by proinflammatory cytokines (288).

Acetylation is another important epigenetic mechanism that impacts gene expression and influences health. The effects of acetylation and deacetylation on transcriptional activity depend on the site of the epigenetic modification. Sirtuins are energy sensors that are activated in conditions of energy restriction. They control gene expression through histone deacetylation of target genes with profound effects on metabolism (289). One mechanistic explanation for the beneficial effects of sirtuins was demonstrated in a sirtuin 1 (SirT1) gain-of-function transgenic mouse model. As compared with control mice, gain of function of SirT1 leads to decreased energy requirements (mirroring a calorie-restricted state), increased adiponectin, and improved insulin sensitivity in response to a high-fat diet despite equivalent weight gain. Part of the mechanism of action involved activation of Foxo1 via deacetylation (290). Pharmacological activation of SirT1 in mice leads to a similar plethora of metabolic benefits (291). These benefits of SitT1 on insulin sensitivity and obesity have made it an attractive target of investigation as a pharmacological option for T2D (292). The knowledge in T1D is limited, but suggests that this pathway could also be a target for therapeutics in T1D. In NOD mice, SirT1 activation via subcutaneous delivery of resveratrol led to an 80% reduction in the development of T1D. This was accompanied by reduced insulitis, higher number of islets, and alterations in immune mediators (e.g., reduction in interferon γ) (293). Similarly, in NOD mice, administration of lysine deacetylase inhibitors, which have shown promise in immunomodulation, at doses that are tolerated clinically led to reduced T1D cumulative incidence, delayed T1D onset, diminished insulitis, and normalized immune function. Lysine deacetylase inhibition also diminished human islet cell apoptosis by reducing inflammatory mediators (294). This points to a unique set of mechanisms by which acetylation could impact immune system function that could be potentially beneficial when obesity and T1D coexist.

Another epigenetic mechanism with a prominent effect on metabolism involves noncoding RNAs, such as microRNA, that regulate the expression of metabolic gene networks. In a study of 2317 individuals from the Framingham Heart Study Offspring Cohort, 16 microRNAs were found to be associated with insulin levels, and two additional microRNAs were also associated with homeostatic model assessment of insulin resistance after adjusting for age, sex, and BMI. Of those 18 microRNAs, miR-122 was also associated with greater BMI, waist circumference, and visceral fat (295). Multiple studies [reviewed in Ref. (296)] have identified microRNAs that are dysregulated in adipose tissue of individuals with obesity. MicroRNAs are also important in regulating physiological processes that are central to the pathogenesis of T1D. A systematic review identified 11 microRNAs that were consistently different in T1D vs healthy controls (297). A review of microRNAs associated with either T1D or T2D reveals a unique fingerprint of microRNAs in T1D (298). Additional studies have demonstrated the impact of microRNA on insulin secretion (299–302), insulin synthesis (303, 304), and pancreatic physiology (305–309) in T1D. Investigations on the differences in miRNA profiles across the spectrum of BMI and body composition could reveal important biomarkers to drive prevention and treatment strategies (310).

The translational relevance of epigenetic control points in T1D has been demonstrated in several studies. In cohorts from DCCT/EDIC with and without diabetes complications by study year 10, profiling of peripheral blood monocytes showed differential epigenetic marks in people on conventional therapy as compared with intensive therapy, and these patterns were associated with higher HbA1c and epigenetic alterations in an inflammatory pathway known to be associated with complications (nuclear factor κ-light-chain-enhancer of activated B cells) (311). In a follow-up study, it was uncovered that these biologically relevant epigenetic marks persisted at study years 16 to 17 in individuals with complications (312). The relationship between epigenetics and T1D complications is supported by another study where genome-wide changes in methylation were identified in individuals with retinopathy as compared with controls (313). Other studies support the role of differential epigenetic marks in individuals with T1D with and without complications (314, 315). These studies are intriguing, as they suggest a role for epigenetics in metabolic memory, which aside from its importance in glycemic control and complications, could modulate the physiological relationships between T1D and obesity (316–318).

Based on the current knowledge of the role of epigenetic mechanisms on energy balance and metabolism, it is likely that a unique set of epigenetic marks will be relevant in scenarios where T1D and obesity coexist. Importantly, these epigenetic fingerprints could serve as biomarkers for disease risk and future complications, making them viable therapeutic targets. Studies to specifically address the gaps in our understanding of the epigenetics of obesity in the context of T1D are essential for maximizing the modifiable influence of epigenetics on physiology.

Microbiome

The human gut microbiome has emerged as a modulator of health outcomes of the host, particularly metabolic diseases with shared pathogenic mechanisms such as obesity (319), NAFLD (320), and diabetes (321). One of the landmark studies that demonstrated the importance of the gut microbiome on metabolic disease showed that fat pads and adipocytes were smaller in germ-free mice vs conventionally raised mice. When gut microbes from conventionally raised mice were transferred to germ-free mice, the obese phenotype was recapitulated (322). Shortly after that discovery, the findings in mice were translated to humans, where it was demonstrated that there are shifts in the proportions of the bacterial phyla Firmicutes and Bacteroidetes in individuals with obesity. (323, 324). The plethora of evidence connecting the gut microbiome to obesity led to many studies showing associations with other metabolic diseases. One prominent example is the connection of the microbiome with NAFLD in both mice (325) and humans (326).

Based on these findings, it is not surprising that the alterations in gut microbiota composition and function are associated with diabetes. Individuals with T2D display moderate dysbiosis, reduced abundance of butyrate-producing bacteria, an increase in opportunistic pathogens, and increased sulfate reduction and drug resistance functions (327). A metagenome analysis in individuals with T2D, as compared with controls, confirmed dysbiotic conditions (decreased Firmicutes, Clostridia, and butyrate-producing bacteria) (328). An important connection between obesity, T2D, and the microbiome is illustrated in bariatric surgery patients. The improvements in T2D that are associated with bariatric surgery are attributable, at least in part, to weight-independent changes in the gut microbiome, incretins, and bile acid signaling (329). As obesity in T1D increases, the likelihood of implementation of bariatric surgery as a therapeutic modality may increase. Studies are needed to understand the unique metabolic outcomes and microbiome phenotypes of bariatric surgery in T1D.

Considerable advances have been made to our understanding of the microbiome in individuals with T1D. Although the microbiome will likely be a major mechanistic connector between obesity and T1D, a full review of the literature is outside the scope of the present review [see Refs. (330) and (331) for reviews]. A subset of key literature is discussed below, and a summary of the progression of the knowledge base in T1D with additional references is provided in Table 2 (332–347). Early rodent studies demonstrated the interaction between innate immunity and the microbiome in the development of T1D. A knockout mouse model of the Toll-like receptor adaptor protein myeloid differentiation primary response 88 on a NOD background was protected from T1D, but when the mice were raised in germ-free conditions, T1D ensued. When germ-free mice were colonized with a complement of microbes that mirror what is found in the human gut, there was a reduction in the development of T1D (332).

Table 2.

Summary of Major Developments in Microbiome Research in T1D

Rodent ModelHuman ModelKey FindingsReferences
XCorrelation between dysbiosis and glycemic control(445, 446)
XMicrobiome imparts protection from T1D(332, 447, 448)
XXMicrobiome differences based on T1D presence or susceptibility(333–336, 449–452)
XXDysbiosis prior to T1D onset(337–341, 453)
XXDysbiosis at or after T1D onset(333, 342, 452, 454)
XXInteractions between the microbiome and immunity in T1D(207, 343, 344, 447, 455–458)
XAntibiotics impact microbiome and T1D onset(459–461)
XMicrobial metabolites and peptides impact T1D pathophysiology(344–347)
XXLifestyle or environmental exposures and microbiome composition in T1D(446, 455, 462–464)
XXInteraction between genetics, microbiome, and T1D(465–467)
Rodent ModelHuman ModelKey FindingsReferences
XCorrelation between dysbiosis and glycemic control(445, 446)
XMicrobiome imparts protection from T1D(332, 447, 448)
XXMicrobiome differences based on T1D presence or susceptibility(333–336, 449–452)
XXDysbiosis prior to T1D onset(337–341, 453)
XXDysbiosis at or after T1D onset(333, 342, 452, 454)
XXInteractions between the microbiome and immunity in T1D(207, 343, 344, 447, 455–458)
XAntibiotics impact microbiome and T1D onset(459–461)
XMicrobial metabolites and peptides impact T1D pathophysiology(344–347)
XXLifestyle or environmental exposures and microbiome composition in T1D(446, 455, 462–464)
XXInteraction between genetics, microbiome, and T1D(465–467)
Table 2.

Summary of Major Developments in Microbiome Research in T1D

Rodent ModelHuman ModelKey FindingsReferences
XCorrelation between dysbiosis and glycemic control(445, 446)
XMicrobiome imparts protection from T1D(332, 447, 448)
XXMicrobiome differences based on T1D presence or susceptibility(333–336, 449–452)
XXDysbiosis prior to T1D onset(337–341, 453)
XXDysbiosis at or after T1D onset(333, 342, 452, 454)
XXInteractions between the microbiome and immunity in T1D(207, 343, 344, 447, 455–458)
XAntibiotics impact microbiome and T1D onset(459–461)
XMicrobial metabolites and peptides impact T1D pathophysiology(344–347)
XXLifestyle or environmental exposures and microbiome composition in T1D(446, 455, 462–464)
XXInteraction between genetics, microbiome, and T1D(465–467)
Rodent ModelHuman ModelKey FindingsReferences
XCorrelation between dysbiosis and glycemic control(445, 446)
XMicrobiome imparts protection from T1D(332, 447, 448)
XXMicrobiome differences based on T1D presence or susceptibility(333–336, 449–452)
XXDysbiosis prior to T1D onset(337–341, 453)
XXDysbiosis at or after T1D onset(333, 342, 452, 454)
XXInteractions between the microbiome and immunity in T1D(207, 343, 344, 447, 455–458)
XAntibiotics impact microbiome and T1D onset(459–461)
XMicrobial metabolites and peptides impact T1D pathophysiology(344–347)
XXLifestyle or environmental exposures and microbiome composition in T1D(446, 455, 462–464)
XXInteraction between genetics, microbiome, and T1D(465–467)

The potential mechanistic driver connecting immunity and the microbiome was recently uncovered, also in NOD mice, with associated translation to clinical populations. Mucosal-associated invariant T (MAIT) cells are a specialized type of innate-like T-cells that recognize microbially derived riboflavin derivatives and promote inflammation and cell death. MAIT cells were highly suppressed in germ-free NOD mice and were found to have protective effects in the development of T1D. Importantly, MAIT cells were highly suppressed in newly diagnosed children with T1D as compared with healthy controls. Modeling with MAIT cell parameters was sufficient to distinguish healthy controls, early onset T1D, and established T1D, suggesting that MAIT cells could be an early biomarker of T1D (343). The work in MAIT cells shows a strong connection between the microbiome and autoimmunity. It is not known whether obesity, in which MAIT cells are also reduced (348, 349), will further deteriorate MAIT cell function in T1D.

In humans, when comparing T1D cases to healthy controls, there is lower functional diversity, as well as a dampening of butyrate production capacity, both of which may contribute to gut permeability and autoimmunity (333). Children with β-cell autoimmunity have a distinct microbiome composition when compared with children matched for age, sex, HLA-DQB1 genotype, and breastfeeding history (337). Several other studies have confirmed the difference in gut microbes in healthy vs T1D cohorts (334, 335, 338, 342). In a study of 33 HLA-matched Estonian infants followed longitudinally from birth to age 3 years, the diversity of the gut microbiota flattens just prior to seroconversion, whereas it increases in nonconverters. The converter microbial profile is characterized by a spike in inflammatory pathways, despite consistent metabolic pathway activity (339). The gut mucosa of individuals with long-standing T1D has proinflammatory phenotype and microbiome composition that are distinct from healthy controls or those with celiac disease. Although causality could not be established, the findings highlight the duodenum as a therapeutic target for the inflammatory processes that accompany autoimmunity (336). In support of this notion, increased intestinal permeability is thought to be an important mediator of T1D onset (340, 350), and this permeability could be remedied by restoring a healthy gut flora (341, 351).

There are several mechanisms whereby the microbiome modulates human physiology. One such method is via the production of metabolites from diet, drugs, or bacterial metabolic activities. The most prominent and well-studied microbial metabolites are short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate, which are produced from the fermentation of indigestible carbohydrates. SCFAs reduce energy harvest, reduce adiposity, and improve insulin sensitivity, which makes them attractive therapeutic targets for obesity and T2D (352). The metabolic phenotypes thought to be attributable to the fermentation of SCFAs could prove beneficial in individuals with obesity and T1D, but there are no published reports on this. However, a metabolic connection in T1D is plausible based on the role of SCFAs on GSIS and immunity. One study found that acetate turnover and plasma/fecal concentrations are markedly increased in high-fat fed, obese, insulin-resistant rats. Additionally, there was a causal relationship between acetate turnover and stimulation of GSIS, which resulted in increased ghrelin secretion, hyperphagia, ectopic deposition, and insulin resistance (345). It is reasonable to hypothesize that individuals with obesity and T1D would also have increased acetate turnover and associated metabolic derangements. Furthermore, in NOD mice, dietary interventions that promote SCFA production protected against the development of T1D via reductions in autoimmunity and protection of gut integrity (346). In Kilham rat virus–induced T1D, provision of SFCAs in drinking water to mothers prior to pregnancy reduced the incidence of virus-induced T1D and insulitis. This was due, at least in part, to SCFA-mediated alterations in gut microbial taxa, restoration of microbial diversity, and a reduction in inflammation. These benefits of SCFAs were not seen when provided after weaning, suggesting the potential for an epigenetic mechanism (347).

Nutrients are life-long modulators of the gut microbiome, and it is not surprising that there is a constantly growing body of literature in this area, including the impact of sugars, resistant starch, fiber, fats, proteins, and micronutrients (e.g., antioxidants, choline, carnitine, betaine) on microbial diversity and metabolism. Recent and comprehensive reviews on this topic are available elsewhere (353, 354), and only a few examples are discussed in this review. Glycemic responses to foods with equivalent glycemic load are highly individualized, and one of the modulators of this individuality is the gut microbiome (355). Artificial sweeteners, which are commonly consumed by individuals with diabetes, alter the microbiome and negatively impact glycemia (356). Dietary and endogenous branched-chain amino acids (BCAAs) are associated with increased risk of developing T2D (357). The plasma metabolome of individuals with insulin resistance is characterized by an increase in BCAAs and a concomitant enrichment of gut microbes that synthesize BCAAs (358). This could be relevant in T1D given the significant positive association between dietary BCAAs and fasting C-peptide in individuals with preserved β-cell function at baseline (fasting C-peptide ≥0.23ng/mL) that was identified in a prospective analysis of the SEARCH for Diabetes in Youth Study (359). The efficacy of customized nutritional interventions in T1D for amelioration of hyperglycemia, promoting insulin sensitivity, modulating energy balance, and preventing autoimmunity via interactions with the microbiome is an area that requires intense research attention.

Much like nutrients that reach the gut microbiota are subject to metabolism, drugs can be subject to alterations in efficacy and bioavailability due to microbial interactions. Drugs used to treat T2D have unique microbial interactions (360). For example, metformin, a commonly used medication for T2D and the most intensively studied adjunctive therapy in T1D, impacts gut microbiome composition (77, 361). It is thought that at least part of the therapeutic efficacy of metformin is due to its effect on the microbiome, particularly on the species Akkermansia muciniphila (362, 363), which improves insulin sensitivity, reduces adipocyte size, and increases fecal energy losses (364). The gut microbes of individuals with T2D differ based on metformin treatment status (328). Emerging evidence is delineating a central role of the microbiome in drug metabolism (365), and this will impact our current understanding of the variability in response to diabetes therapeutics. Studies are needed to uncover the potential intersections between the microbiome, obesity, and T1D therapeutics.

Despite the many advances in the field of the gut microbiome in T1D, many questions remain unanswered, including ascertainment of causality, the functional connection between the autoimmune process and the gut microbiome, and the therapeutic efficacy of microbiome modulators (366). Importantly, there are no published studies characterizing the microbiome in lean individuals vs those who are obese and have T1D or deciphering how differences in the microbiome could affect T1D and obesity-related complications.

Adipose tissue dysfunction

In contrast to the historical characterization of adipose tissue as an inactive organ with a primary role of storing excess energy, it is now undeniable that it is an endocrine organ with far-reaching effects on physiology. The beneficial actions of adipose tissue become dysregulated in obesity [reviewed in Refs. (367) and (368)]. Total adiposity only reveals a fraction of the physiological impact of obesity (369). Adipose distribution, particularly higher central localization in the abdominal region and accumulation in muscle, is associated with insulin resistance, T2D (370–372), and myocardial infarction (373). Skeletal muscle infiltration of adipose tissue (intramuscular lipid content) correlates with insulin resistance in obesity and T2D (374). In lean individuals (BMI 25.1 ± 2.5 kg/m2) with well-controlled T1D compared with people with T2D and obesity, deep thigh fat was similar and the ratio of mid-thigh deep fat/total fat was higher. This phenotype might be associated with insulin resistance (372). In support of this notion, in lean individuals (BMI 22.5 ± 0.7 kg/m2) with poorly controlled T1D, higher intramuscular lipid content was a marker of insulin resistance (375). Adipose tissue that surrounds the coronary arteries, that is, pericardial adipose tissue, is associated with heart disease and T2D (376, 377). In people with and without T1D, the ratio of pericardial adipose tissue to subcutaneous adipose tissue is associated with insulin resistance irrespective of total adipose volume (378). These studies highlight the importance of adipose tissue distribution in the metabolic phenotype of obesity, T1D, and T2D. The mechanisms mediating the role of specific adipose depots on health are active areas of investigation, and epigenetic control points seem to explain some of the functional differences (379, 380). Additional research is needed to define the relationships between adipose distribution and in-depth metabolic phenotypes in lean individuals vs those with obesity and T1D compared with matched healthy controls.

Adipose tissue is modulated during obesity progression and its dysfunction, which involves inflammation, fibrosis, disruptions in angiogenesis, and alterations in release of adipokines, influences insulin sensitivity, energy balance, and other key metabolic control points, creating a perpetuating cycle of disease advancement (368). The functional importance of adipose tissue for obesity, T1D, and T2D, described elsewhere in this review, includes its roles in metabolic flexibility, β-cell physiology, and mitochondrial function. A few additional examples of the metabolic consequences of unhealthy adipose tissue are discussed below.

Release of adequate amounts of functional adipokines, such as leptin and adiponectin, is necessary for glucose metabolism (381, 382), and one of the target tissues of insulin-sensitizing drugs is adipose tissue (383). Adiponectin has a unique regulatory paradigm in T1D. Adiponectin correlates with insulin sensitivity in T1D similarly to controls but at slightly higher adiponectin concentrations for a similar degree of insulin resistance (384). Leptin function is also altered in T1D; leptin levels are lower at onset of T1D as compared with healthy controls, but are rapidly restored with insulin treatment (385). Additionally, leptin levels are higher in individuals treated with intensive insulin therapy vs conventional therapy (386). The mechanistic impact of leptin in T1D was demonstrated in the Akita mouse, a nonobese model of T1D that displays β-cell failure, hyperglycemia, and a shortened lifespan (387). In these mice, expression of leptin in the hypothalamus restored euglycemia (388). Transgenic Akita mice with hyperleptinemia are normoglycemic, have improved renal function, and have a longer lifespan (389). Several other studies have highlighted the role of restoring adipokine function as a means to treat T1D in rodent models (390–394). The potential benefits of restoring the hormonal output of adipose tissue in T1D was demonstrated via the use of recombinant methionyl human leptin (metreleptin) in individuals with T1D. After 20 weeks of therapy, there were modest reductions in weight (6.6%), adipose (8%), and insulin dose (15%), without an improvement in glycemic control (395). Despite the lack of efficacy with respect to glycemic control in this pilot study, the benefits on weight and reduction in insulin dose support the idea that healthy adipose function is important for T1D.

“Studies are needed to compare the genetic risk factors for T1D in lean individuals vs those who are obese.”

Adipose tissue fibrosis is also a hallmark of obesity and T2D. It is characterized by excessive extracellular matrix deposition and is a pathological consequence of long-term adipose dysfunction (368). In obesity, adipose tissue fibrosis is associated with accumulation of the adipokine endotrophin, inflammation, and insulin resistance (396). In db/db mice, there is an upregulation of most adipose tissue collagens, and this is abrogated in wild-type mice treated with peroxisome proliferator-activated receptor γ agonists or in mice overexpressing adiponectin. One of the most enriched collagens in adipose tissue is collagen VI, and it is overexpressed in both ob/ob and db/db mice (397). Given the relevance of adipose tissue fibrosis on obesity, insulin resistance, and T2D, the impact of T1D on the metabolic aberrations that accompany adipose tissue fibrosis is an area that could reveal novel mechanistic pathways and therapeutic options.

The role of adipose tissue function, and not merely changes in adipose tissue mass or distribution, was demonstrated in a study where transplantation of healthy adipose into C57BL6/J mice led to an improvement in glucose tolerance and insulin sensitivity (398). Metabolic benefits of adipose tissue transplantation have also been demonstrated in T1D models. The BioBreeding/Ottawa Karlsburg rat develops T1D spontaneously. One research group found that transplantation of visceral white adipose tissue led to a reduction in the development of T1D in females more than males (399). This dichotomy parallels the findings related to greater metabolic flexibility in females, which is due, at least in part, to adipose tissue function (400). These results are supported by a study showing that transplantation of embryonic brown adipose tissue (BAT) leads to weight gain and euglycemia in STZ-treated mice. This euglycemia persists for >6 months. Interestingly, this improvement in glucose metabolism is not dependent on insulin. Restoration of healthy adipose tissue seemed to be the main driver of these benefits as evidenced by reduced adipose tissue inflammation, increased leptin, adiponectin, and IGF-1, glucagon suppression, and insulin receptor activation (401). The importance of BAT for T1D is supported by the finding that STZ mice have both BAT dysfunction and fibroblast growth factor 21 (FGF21) deficiency. Treatment with an FGF21 analog (LY2405319) reduced blood glucose levels and improved BAT function (402). These findings are particularly intriguing given that these are the first preclinical data in T1D of an FGF21 analog that is under active investigation in clinical trials for its glycemic and other metabolic benefits in T2D and obesity (403). Once sufficient preclinical data are available, clinical trials will be needed to understand whether LY2405319 will be beneficial in people with T1D, particularly in the context of obesity. Although the studies presented related to restoration of adipose tissue function are restricted to preclinical models, they highlight a potential therapeutic avenue for treatment of human disease that could be beneficial in individuals with T1D. There are ongoing clinical trials of transplantation of adipose-derived stem cells for the treatment of various diseases. Therefore, the potential benefits of restoration of adipose tissue function on metabolic disease merit additional investigation into the long-term safety and efficacy of transplantation of adipose tissue and/or cell derivatives in humans (404).

Overall, the studies reviewed above highlight the potential importance of adipose tissue burden, distribution, and function for glycemic control in T1D and suggests that adipose-targeted therapeutics that are independent of insulin could be highly beneficial in T1D. This makes the adipose dysfunction seen in obesity a major concern for people with T1D that warrants targeted investigation.

β-Cell dysfunction

T1D is characterized by a total or near lack of insulin secretion due to the destruction of β-cells by autoimmune or (rarely) other processes. Although T1D often presents as an acute illness due to marked hyperglycemia and metabolic decompensation, studies have demonstrated that the β-cell phenotype develops gradually over time, although with a more rapid course than in T2D (405). Before overt β-cell failure occurs, there is evidence of β-cell dysfunction and insulin resistance, and this sometimes correlates with the presence of excess adiposity (406).

Considerable evidence links obesity-mediated pathways to β-cell dysfunction in T2D, including a chronic state of elevated free fatty acids, which leads to lipotoxicity in β-cells (407) along with CD95 receptor–mediated inflammation and apoptosis in human islets (408). Additionally, weight loss improves β-cell function (409), and multiple studies on populations around the globe have shown the benefits of weight loss for long-term diabetes prevention (410–412). Therefore, achieving or maintaining a healthy weight is an important therapeutic mechanism to prevent or ameliorate T2D via preservation of β-cell function.

There are few studies addressing the β-cell phenotype in individuals with obesity and T1D. One study in an ethnically diverse cohort of children with newly diagnosed T1D found that children with obesity had higher C-peptide levels than did overweight and lean counterparts (149), and this relationship was confirmed by other studies (143, 413). This may reflect obesity-related insulin resistance and resulting β-cell compensation preceding the onset of the autoimmune process in T1D. This apparent paradox may be due to the fact that healthy adipose tissue is essential for β-cell function [discussed above and reviewed in Ref. (414)]. Additional studies are needed to understand this association between BMI, C-peptide, and β-cell decline over time.

As discussed previously, the microbiome is relevant to both T1D and obesity. Recent evidence suggests that the microbiome impacts β-cell function. Cathelicidin antimicrobial peptide (CAMP) is a secreted gut peptide that modulates immune function and interacts with the microbiome. CAMP is expressed in β-cells and δ-cells, but not α-cells, of rats, mice, and humans. In a rat model of T1D, the BioBreeding diabetes-prone rat, the secretion of this peptide was diminished prior to the onset of T1D as compared with the BioBreeding control rat. Treatment with CAMP restored islet function as evidenced by insulin and glucagon secretion. It also induced a potentially beneficial gut microbiome profile (344).

Evidence exists that in T1D, a large proportion of individuals display residual β-cell function (415, 416) and, importantly, that insulin secretion can be partially restored (417, 418). Preserving or restoring this function is a central focus of novel T1D therapeutic efforts (419). This is of key importance when considering the potential impact of obesity on the long-term outcomes of people with T1D, as the β-cell hostile environment that accompanies the obese phenotype may exacerbate the loss of this residual functionality. The effects that obesity plus T1D have on β-cell longevity are not well understood, and maximizing the efficacy of novel therapeutics requires this knowledge.

Mitochondrial dysfunction

GSIS from β-cells depends on mitochondrial generation of ATP. Because mitochondrial function is essential for maintaining glucose homeostasis, it is not surprising that mitochondrial dysfunction is a common underlying mechanism in metabolic diseases (420, 421). A major component of one’s ability to metabolically adapt to fuel availability and demand is thought to be driven primarily by mitochondria in skeletal muscle and other tissues (227).

In twins discordant for obesity, the obese phenotype is characterized by downregulation of mitochondrial oxidative phosphorylation and reduced mitochondrial DNA. These defects are present before the onset of metabolic disease, suggesting a role in pathogenesis (422). In T2D and insulin-resistant states, there is a reduction in the functionality of the mitochondrial electron transport chain (423, 424) via multiple mechanisms, including changes in gene expression (425, 426), reduced enzyme activity (427), and shifts in lipid metabolism in skeletal muscle (428, 429), liver, and adipose tissue (430). It is likely that mitochondrial dysfunction precedes the onset of T2D because lean, insulin-resistant offspring of individuals with T2D demonstrate reduced mitochondrial ATP production (431). More support for this early causative role of mitochondrial dysfunction on metabolic disease comes from studies that demonstrated that elevated free fatty acids after a lipid infusion are associated with diminished skeletal muscle mitochondrial membrane potential, insulin resistance (432), and altered nuclear-encoded mitochondrial genes (433).

In agreement with what is known in T2D and obesity, there have been documented mitochondrial deficiencies associated with T1D. In the Akita+/Ins2 model of monogenic T1D, mitochondrial dysfunction was evidenced by defects in mitochondrial bioenergetics (reduced ATP respiration, maximal respiration, and reserve capacity), dysregulation of mitochondrial respiratory chain proteins, dysmorphology, increased oxidative stress (427), and remodeling of the mitochondrial proteome (434). There is a relationship between PDX1, a gene that is essential for islet development, β-cell maintenance, GSIS (435), and mitochondrial function (436). This gene has been implicated in both T1D and T2D pathogenesis and may shed light onto mitochondrial therapeutics to preserve or reactivate β-cells. Insulin itself has profound effects on mitochondrial function in multiple tissues by enhancing production of ATP in skeletal muscle mitochondria, increasing activity of skeletal muscle mitochondrial oxidative enzymes (437), signaling through Foxo1 in liver (438), and reducing mitochondrial uncoupling in BAT (439). The ability to restore insulin modulation of mitochondrial function fully with exogenous insulin is likely to be suboptimal.

In clinical studies of T1D, the impact of mitochondrial function on diabetes complications has been revealed. Mitochondrial DNA haplogroups are patterns of point mutations in mitochondrial DNA that are thought to impact mitochondrial function. In comparison with people with T1D free from retinopathy, mitochondrial DNA haplogroups have been associated with higher severity of diabetic retinopathy (440). Diabetic kidney disease is associated with genetic risk factors for mitochondrial function (441), and this mirrors findings in mice where glomerular endothelial mitochondrial function is a central control point for kidney disease progression (442). The epigenetic patterns of mitochondrial function genes in circulating DNA differ in individuals with T1D with and without renal disease (443), suggesting that mitochondrial dysfunction is indeed an early event that impacts kidney health.

“It is likely that a unique set of epigenetic marks will be relevant in scenarios where T1D and obesity coexist.”

There is robust evidence that unhealthy mitochondria are a hallmark of obesity, T1D, and T2D. It is unclear how the potential additive effects of obesity and T1D will impact mitochondrial dysfunction and T1D progression. It is also plausible that T1D-mediated mitochondrial dysfunction could promote obesity and insulin resistance. Understanding both paradigms is necessary to effectively ameliorate or prevent obesity in T1D and to develop therapies that address the unique mitochondrial characteristics when obesity is concurrent with T1D.

Future Directions and Perspectives

The collective knowledge presented in this review strongly suggests that there is a pressing need to generate the scientific evidence base to develop weight management guidelines that are specific to T1D. Several major gaps in our understanding need to be addressed so that weight management strategies specific for people with T1D can be developed, including:

  • What are the unique behavioral, psychological, and social characteristics that underlie obesity in T1D?

  • What are the biological and behavioral mechanisms that contribute to the obese phenotype in T1D?

  • Does obesity impact the natural history of T1D, from seroconversion through the appearance of complications?

    • When obesity occurs before or after T1D diagnosis, how is the health of a person with T1D modified?

  • Could obesity-related pathways serve as therapeutic or prevention targets that are insulin-independent or act synergistically with intensive insulin therapy?

  • How do energy and macronutrient balance shift in response to advanced medical therapies (pharmacologic and technologic), dietary challenges, exercise, and glycemic control in the context of obesity?

    • How do these shifts influence therapeutic requirements and the subsequent response to those therapies?

  • What lifestyle approaches will be most efficacious for preventing and addressing obesity in people with T1D while concurrently achieving glycemic control?

  • What is the role for medications that promote weight loss?

    • Can these be used as adjunctive therapy in patients with T1D to counterbalance the effects of intensive insulin therapy to improve glycemic control?

A proposed framework to traverse the continuum from the lack of scientific evidence to the implementation of efficacious health care paradigms that address the maintenance or achievement of a healthy weight while optimizing glycemic control is presented in Figure 3. A key first step for building this framework is the collaboration between synergistic research experts with a comprehensive understanding of existing knowledge to design and execute research approaches that will define individual metabolic, biological, and behavioral characteristics of obesity in T1D. Identification of subgroups of individuals with T1D who respond differentially to interventions will allow for the design and testing of innovative individualized approaches. Accomplishing this will require the use of novel trial designs that are more efficient, especially relative to subgroups who may respond differently to various approaches. These designs need to consider, at minimum, sex, glycemic control, and therapeutic regimen (including use of insulin pump therapy, multiple daily injections, and automated insulin delivery systems). This can be accomplished through adaptive interventions and sequential multiple assignment randomized trial designs (444). Studies are needed to uncover medical approaches to promote glycemic control while maintaining energy balance, barriers and facilitators of behavior change, unique aspects of ingestive behavior, and the phenotype of energy and macronutrient balance. Finally, efforts will be needed to ensure effective implementation and dissemination of proven interventions. In concert, such efforts will advance capacity to improve outcomes for individuals living with T1D and obesity.

A framework for defining transdisciplinary research paradigms aimed at the development of T1D-specific weight management recommendations. This framework requires the merger of complementary expertise and existing knowledge to design and implement transdisciplinary research approaches that will generate the data needed to develop T1D-specific weight management recommendations.
Figure 3.

A framework for defining transdisciplinary research paradigms aimed at the development of T1D-specific weight management recommendations. This framework requires the merger of complementary expertise and existing knowledge to design and implement transdisciplinary research approaches that will generate the data needed to develop T1D-specific weight management recommendations.

Abbreviations

    Abbreviations
     
  • ADA

    American Diabetes Association

  •  
  • AUC

    area under the curve

  •  
  • BAT

    brown adipose tissue

  •  
  • BCAA

    branched-chain amino acid

  •  
  • BMI

    body mass index

  •  
  • CAMP

    cathelicidin antimicrobial peptide

  •  
  • CCK

    cholecystokinin

  •  
  • DCCT

    Diabetes Control and Complications Trial

  •  
  • DPP-4

    dipeptidyl peptidase-4

  •  
  • EDIC

    Epidemiology of Diabetes Interventions and Complications

  •  
  • FGF21

    fibroblast growth factor 21

  •  
  • Foxo

    forkhead box O

  •  
  • FTO

    fat mass and obesity-associated protein

  •  
  • GIP

    glucose-dependent insulinotropic polypeptide

  •  
  • GLP-1

    glucagon-like peptide-1

  •  
  • GSIS

    glucose-stimulated insulin secretion

  •  
  • HLA

    human leukocyte antigen

  •  
  • MAIT

    mucosal-associated invariant T

  •  
  • MC4R

    melanocortin 4 receptor

  •  
  • NAFLD

    nonalcoholic fatty liver disease

  •  
  • NHANES

    National Health and Nutrition Examination Survey

  •  
  • NOD

    nonobese diabetic

  •  
  • Pittsburgh EDC

    Pittsburgh Epidemiology of Diabetes Complications

  •  
  • PYY

    peptide tyrosine-tyrosine

  •  
  • SCFA

    short-chain fatty acid

  •  
  • SGLT

    sodium-glucose cotransporter

  •  
  • SirT1

    sirtuin 1

  •  
  • SNP

    single nucleotide polymorphism

  •  
  • STZ

    streptozotocin

  •  
  • T1D

    type 1 diabetes

  •  
  • T2D

    type 2 diabetes

  •  
  • TIDX

    T1D Exchange Registry

Acknowledgments

We thank Anna Kahkoska for assistance with creating the graphics for Figure 3 that summarize the key message in this review.

Advancing Care for Type 1 Diabetes and Obesity Network (ACT1ON) is a consortium of clinical, behavioral, and basic scientists comprising expertise in endocrinology, epidemiology, nutrition, exercise physiology, clinical psychology, and biostatistics from multiple institutions led by the University of North Carolina, Stanford University, the Florida Hospital Translational Medicine Research Institute for Metabolism and Diabetes, and the Barbara Davis Center. ACT1ON aims to elucidate drivers of overweight and obesity in youth and adults with T1D. The overarching goal of ACT1ON is to develop evidence-based guidelines for individuals with T1D and their diabetes care team regarding weight management and weight loss. Members of the consortium for ACT1ON include the following: University of Colorado Denver Barbara Davis Center: Franziska K. Bishop, MS, CPHS CDE, and Kimberly A. Driscoll, PhD; Stanford University Division of Endocrinology: David M. Maahs, MD, PhD; Ananta Addala, DO, MPH; Laya Ekhlaspour, MD; Korey K. Hood, PhD; and Marissa Town, RN, BSN, CDE; Florida Hospital Translational Research Institute for Metabolism and Diabetes: Richard Pratley, MD; Karen D. Corbin, PhD, RD, Steven R. Smith, MD; and Keri Whitaker, RN, CCRP; North Carolina State University: Eric Laber, PhD; University of North Carolina at Chapel Hill, Glillings School of Global Public Health, Departments of Nutrition and Medicine: Elizabeth Mayer-Davis, PhD; Kyle Burger, PhD; Ian Carroll, PhD; Daria Igudesman; Anna Kahkoska; Michael R. Kosorok, PhD; Grace Shearrer, PhD; and Joan Thomas, MS, RD; Boston Children’s Hospital: Garry Steil, PhD; York University School of Kinesiology and Health Science: Michael Riddell, PhD.

Disclosure Summary: D.M.M. has received institutional research funding from Dexcom, Medtronic, Bigfoot Biomedical, Insulet, and Roche; has served on the advisory board for Insulet; and has acted as a consultant for Abbott Diabetes Care, Sanofi, and Eli Lilly. R.E.P. received research grants, honoraria, speaker’s bureau fees, and consulting fees paid directly to Florida Hospital, a nonprofit organization, from AstraZeneca, Boehringer Ingelheim, Gilead Sciences, GlaxoSmithKline, Hanmi Pharmaceutical, Eli Lilly and Company, ICON Clinical Research, Janssen Pharmaceuticals, Lexicon Pharmaceuticals, Ligand Pharmaceuticals, Merck & Company, Novo Nordisk, Pfizer, Sanofi-Aventis, Takeda, and Eisai. S.R.S. serves on scientific advisory boards or consults for Takeda, Arena Pharmaceuticals, AstraZeneca, Boehringer Ingelheim Pharma, Bristol-Myers Squibb, Eisai, Elcelyx, Eli Lilly and Company, Five Prime Therapeutics, GlaxoSmithKline US Processing/Genpact AP, NGM Biopharmaceuticals, Novo Nordisk, Orexigen Therapeutics, Piramal Life Sciences, Takeda Global Research and Development, and Zafgen; receives research support from Amylin Pharmaceuticals, BirdRock Bio, Eli Lilly and Company, and Takeda; and has equity in Jenrin Discovery and Zafgen. The remaining authors have nothing to disclose.

References

1.

Kahn
SE
,
Hull
RL
,
Utzschneider
KM
.
Mechanisms linking obesity to insulin resistance and type 2 diabetes
.
Nature
.
2006
;
444
(
7121
):
840
846
.

2.

American Diabetes Association
.
7. Obesity management for the treatment of type 2 diabetes: Standards of Medical Care in Diabetes—2018.
Diabetes Care
.
2018
;
41
(
Suppl 1
):
S65
S72
.

3.

Conway
B
,
Miller
RG
,
Costacou
T
,
Fried
L
,
Kelsey
S
,
Evans
RW
,
Orchard
TJ
.
Temporal patterns in overweight and obesity in type 1 diabetes
.
Diabet Med
.
2010
;
27
(
4
):
398
404
.

4.

Baskaran
C
,
Volkening
LK
,
Diaz
M
,
Laffel
LM
.
A decade of temporal trends in overweight/obesity in youth with type 1 diabetes after the Diabetes Control and Complications Trial
.
Pediatr Diabetes
.
2015
;
16
(
4
):
263
270
.

5.

Minges
KE
,
Whittemore
R
,
Weinzimer
SA
,
Irwin
ML
,
Redeker
NS
,
Grey
M
.
Correlates of overweight and obesity in 5529 adolescents with type 1 diabetes: the T1D Exchange Clinic Registry
.
Diabetes Res Clin Pract
.
2017
;
126
:
68
78
.

6.

Ogden
CL
,
Carroll
MD
,
Kit
BK
,
Flegal
KM
.
Prevalence of childhood and adult obesity in the United States, 2011–2012
.
JAMA
.
2014
;
311
(
8
):
806
814
.

7.

Sharma
A
,
Lavie
CJ
,
Borer
JS
,
Vallakati
A
,
Goel
S
,
Lopez-Jimenez
F
,
Arbab-Zadeh
A
,
Mukherjee
D
,
Lazar
JM
.
Meta-analysis of the relation of body mass index to all-cause and cardiovascular mortality and hospitalization in patients with chronic heart failure
.
Am J Cardiol
.
2015
;
115
(
10
):
1428
1434
.

8.

Anderson
LN
,
Lebovic
G
,
Hamilton
J
,
Hanley
AJ
,
McCrindle
BW
,
Maguire
JL
,
Parkin
PC
,
Birken
CS
;
TARGet Kids Collaboration
.
Body mass index, waist circumference, and the clustering of cardiometabolic risk factors in early childhood
.
Paediatr Perinat Epidemiol
.
2016
;
30
(
2
):
160
170
.

9.

Masmiquel
L
,
Leiter
LA
,
Vidal
J
,
Bain
S
,
Petrie
J
,
Franek
E
,
Raz
I
,
Comlekci
A
,
Jacob
S
,
van Gaal
L
,
Baeres
FM
,
Marso
SP
,
Eriksson
M
;
LEADER investigators
.
LEADER 5: prevalence and cardiometabolic impact of obesity in cardiovascular high-risk patients with type 2 diabetes mellitus: baseline global data from the LEADER trial
.
Cardiovasc Diabetol
.
2016
;
15
(
1
):
29
.

10.

Guo
F
,
Garvey
WT
.
Cardiometabolic disease risk in metabolically healthy and unhealthy obesity: stability of metabolic health status in adults
.
Obesity (Silver Spring)
.
2016
;
24
(
2
):
516
525
.

11.

Renehan
AG
,
Tyson
M
,
Egger
M
,
Heller
RF
,
Zwahlen
M
.
Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies
.
Lancet
.
2008
;
371
(
9612
):
569
578
.

12.

Arnold
M
,
Pandeya
N
,
Byrnes
G
,
Renehan
PAG
,
Stevens
GA
,
Ezzati
PM
,
Ferlay
J
,
Miranda
JJ
,
Romieu
I
,
Dikshit
R
,
Forman
D
,
Soerjomataram
I
.
Global burden of cancer attributable to high body-mass index in 2012: a population-based study
.
Lancet Oncol
.
2015
;
16
(
1
):
36
46
.

13.

Dabelea
D
,
Mayer-Davis
EJ
,
Saydah
S
,
Imperatore
G
,
Linder
B
,
Divers
J
,
Bell
R
,
Badaru
A
,
Talton
JW
,
Crume
T
,
Liese
AD
,
Merchant
AT
,
Lawrence
JM
,
Reynolds
K
,
Dolan
L
,
Liu
LL
,
Hamman
RF
;
SEARCH for Diabetes in Youth Study
.
Prevalence of type 1 and type 2 diabetes among children and adolescents from 2001 to 2009
.
JAMA
.
2014
;
311
(
17
):
1778
1786
.

14.

Mayer-Davis
EJ
,
Lawrence
JM
,
Dabelea
D
,
Divers
J
,
Isom
S
,
Dolan
L
,
Imperatore
G
,
Linder
B
,
Marcovina
S
,
Pettitt
DJ
,
Pihoker
C
,
Saydah
S
,
Wagenknecht
L
;
SEARCH for Diabetes in Youth Study
.
Incidence trends of type 1 and type 2 diabetes among youths, 2002–2012
.
N Engl J Med
.
2017
;
376
(
15
):
1419
1429
.

15.

Imperatore
G
,
Boyle
JP
,
Thompson
TJ
,
Case
D
,
Dabelea
D
,
Hamman
RF
,
Lawrence
JM
,
Liese
AD
,
Liu
LL
,
Mayer-Davis
EJ
,
Rodriguez
BL
,
Standiford
D
;
SEARCH for Diabetes in Youth Study Group
.
Projections of type 1 and type 2 diabetes burden in the U.S. population aged <20 years through 2050: dynamic modeling of incidence, mortality, and population growth
.
Diabetes Care
.
2012
;
35
(
12
):
2515
2520
.

16.

Maahs
DM
,
West
NA
,
Lawrence
JM
,
Mayer-Davis
EJ
.
Epidemiology of type 1 diabetes
.
Endocrinol Metab Clin North Am
.
2010
;
39
(
3
):
481
497
.

17.

Purnell
JQ
,
Zinman
B
,
Brunzell
JD
;
DCCT/EDIC Research Group
.
The effect of excess weight gain with intensive diabetes mellitus treatment on cardiovascular disease risk factors and atherosclerosis in type 1 diabetes mellitus: results from the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Study (DCCT/EDIC) study
.
Circulation
.
2013
;
127
(
2
):
180
187
.

18.

Pi-Sunyer
X
,
Blackburn
G
,
Brancati
FL
,
Bray
GA
,
Bright
R
,
Clark
JM
,
Curtis
JM
,
Espeland
MA
,
Foreyt
JP
,
Graves
K
,
Haffner
SM
,
Harrison
B
,
Hill
JO
,
Horton
ES
,
Jakicic
J
,
Jeffery
RW
,
Johnson
KC
,
Kahn
S
,
Kelley
DE
,
Kitabchi
AE
,
Knowler
WC
,
Lewis
CE
,
Maschak-Carey
BJ
,
Montgomery
B
,
Nathan
DM
,
Patricio
J
,
Peters
A
,
Redmon
JB
,
Reeves
RS
,
Ryan
DH
,
Safford
M
,
Van Dorsten
B
,
Wadden
TA
,
Wagenknecht
L
,
Wesche-Thobaben
J
,
Wing
RR
,
Yanovski
SZ
;
Look AHEAD Research Group
.
Reduction in weight and cardiovascular disease risk factors in individuals with type 2 diabetes: one-year results of the look AHEAD trial
.
Diabetes Care
.
2007
;
30
(
6
):
1374
1383
.

19.

Liu
LL
,
Lawrence
JM
,
Davis
C
,
Liese
AD
,
Pettitt
DJ
,
Pihoker
C
,
Dabelea
D
,
Hamman
R
,
Waitzfelder
B
,
Kahn
HS
;
SEARCH for Diabetes in Youth Study Group
.
Prevalence of overweight and obesity in youth with diabetes in USA: the SEARCH for Diabetes in Youth study
.
Pediatr Diabetes
.
2010
;
11
(
1
):
4
11
.

20.

DuBose
SN
,
Hermann
JM
,
Tamborlane
WV
,
Beck
RW
,
Dost
A
,
DiMeglio
LA
,
Schwab
KO
,
Holl
RW
,
Hofer
SE
,
Maahs
DM
;
Type 1 Diabetes Exchange Clinic Network and Diabetes Prospective Follow-up Registry
.
Obesity in youth with type 1 diabetes in Germany, Austria, and the United States
.
J Pediatr
.
2015
;
167
(
3
):
627
632.e1
–4.

21.

Ogden
CL
,
Carroll
MD
,
Lawman
HG
,
Fryar
CD
,
Kruszon-Moran
D
,
Kit
BK
,
Flegal
KM
.
Trends in obesity prevalence among children and adolescents in the United States, 1988–1994 through 2013–2014
.
JAMA
.
2016
;
315
(
21
):
2292
2299
.

22.

Flegal
KM
,
Carroll
MD
,
Ogden
CL
,
Curtin
LR
.
Prevalence and trends in obesity among US adults, 1999–2008
.
JAMA
.
2010
;
303
(
3
):
235
241
.

23.

Nathan
DM
,
Zinman
B
,
Cleary
PA
,
Backlund
JY
,
Genuth
S
,
Miller
R
,
Orchard
TJ
;
Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Research Group
.
Modern-day clinical course of type 1 diabetes mellitus after 30 years’ duration: the diabetes control and complications trial/epidemiology of diabetes interventions and complications and Pittsburgh epidemiology of diabetes complications experience (1983–2005)
.
Arch Intern Med
.
2009
;
169
(
14
):
1307
1316
.

24.

American Diabetes Association
.
4. Lifestyle management: Standards of Medical Care in Diabetes—2018.
Diabetes Care
.
2018
;
41
(
Suppl 1
):
S38
S50
.

25.

Wheeler
ML
,
Dunbar
SA
,
Jaacks
LM
,
Karmally
W
,
Mayer-Davis
EJ
,
Wylie-Rosett
J
,
Yancy
WS
Jr
.
Macronutrients, food groups, and eating patterns in the management of diabetes: a systematic review of the literature, 2010
.
Diabetes Care
.
2012
;
35
(
2
):
434
445
.

26.

Delahanty
LM
,
Nathan
DM
,
Lachin
JM
,
Hu
FB
,
Cleary
PA
,
Ziegler
GK
,
Wylie-Rosett
J
,
Wexler
DJ
;
Diabetes Control and Complications Trial/Epidemiology of Diabetes
.
Association of diet with glycated hemoglobin during intensive treatment of type 1 diabetes in the Diabetes Control and Complications Trial
.
Am J Clin Nutr
.
2009
;
89
(
2
):
518
524
.

27.

Jakicic
JM
,
Winters
C
,
Lang
W
,
Wing
RR
.
Effects of intermittent exercise and use of home exercise equipment on adherence, weight loss, and fitness in overweight women: a randomized trial
.
JAMA
.
1999
;
282
(
16
):
1554
1560
.

28.

Colberg
SR
,
Sigal
RJ
,
Yardley
JE
,
Riddell
MC
,
Dunstan
DW
,
Dempsey
PC
,
Horton
ES
,
Castorino
K
,
Tate
DF
.
Physical activity/exercise and diabetes: a position statement of the American Diabetes Association
.
Diabetes Care
.
2016
;
39
(
11
):
2065
2079
.

29.

McCarthy
MM
,
Funk
M
,
Grey
M
.
Cardiovascular health in adults with type 1 diabetes
.
Prev Med
.
2016
;
91
:
138
143
.

30.

Robertson
K
,
Riddell
MC
,
Guinhouya
BC
,
Adolfsson
P
,
Hanas
R
;
International Society for Pediatric and Adolescent Diabetes
.
ISPAD Clinical Practice Consensus Guidelines 2014. Exercise in children and adolescents with diabetes
.
Pediatr Diabetes
.
2014
;
15
(
Suppl 20
):
203
223
.

31.

Krause
MP
,
Riddell
MC
,
Hawke
TJ
.
Effects of type 1 diabetes mellitus on skeletal muscle: clinical observations and physiological mechanisms
.
Pediatr Diabetes
.
2011
;
12
(
4 Pt 1
):
345
364
.

32.

Bohn
B
,
Herbst
A
,
Pfeifer
M
,
Krakow
D
,
Zimny
S
,
Kopp
F
,
Melmer
A
,
Steinacker
JM
,
Holl
RW
;
DPV Initiative
.
Impact of physical activity on glycemic control and prevalence of cardiovascular risk factors in adults with type 1 diabetes: a cross-sectional multicenter study of 18,028 patients
.
Diabetes Care
.
2015
;
38
(
8
):
1536
1543
.

33.

Riddell
MC
,
Gallen
IW
,
Smart
CE
,
Taplin
CE
,
Adolfsson
P
,
Lumb
AN
,
Kowalski
A
,
Rabasa-Lhoret
R
,
McCrimmon
RJ
,
Hume
C
,
Annan
F
,
Fournier
PA
,
Graham
C
,
Bode
B
,
Galassetti
P
,
Jones
TW
,
Millán
IS
,
Heise
T
,
Peters
AL
,
Petz
A
,
Laffel
LM
.
Exercise management in type 1 diabetes: a consensus statement
.
Lancet Diabetes Endocrinol
.
2017
;
5
(
5
):
377
390
.

34.

Baughman
K
,
Logue
E
,
Sutton
K
,
Capers
C
,
Jarjoura
D
,
Smucker
W
.
Biopsychosocial characteristics of overweight and obese primary care patients: do psychosocial and behavior factors mediate sociodemographic effects
?
Prev Med
.
2003
;
37
(
2
):
129
137
.

35.

Herzer
M
,
Zeller
MH
,
Rausch
JR
,
Modi
AC
.
Perceived social support and its association with obesity-specific health-related quality of life
.
J Dev Behav Pediatr
.
2011
;
32
(
3
):
188
195
.

36.

Munt
AE
,
Partridge
SR
,
Allman-Farinelli
M
.
The barriers and enablers of healthy eating among young adults: a missing piece of the obesity puzzle: a scoping review
.
Obes Rev
.
2017
;
18
(
1
):
1
17
.

37.

Jaser
SS
.
Psychological problems in adolescents with diabetes
.
Adolesc Med State Art Rev
.
2010
;
21
(
1
):
138
151
.

38.

Crow
S
,
Kendall
D
,
Praus
B
,
Thuras
P
.
Binge eating and other psychopathology in patients with type II diabetes mellitus
.
Int J Eat Disord
.
2001
;
30
(
2
):
222
226
.

39.

Wing
RR
,
Goldstein
MG
,
Acton
KJ
,
Birch
LL
,
Jakicic
JM
,
Sallis
JF
Jr
,
Smith-West
D
,
Jeffery
RW
,
Surwit
RS
.
Behavioral science research in diabetes: lifestyle changes related to obesity, eating behavior, and physical activity
.
Diabetes Care
.
2001
;
24
(
1
):
117
123
.

40.

Malik
VS
,
Popkin
BM
,
Bray
GA
,
Després
JP
,
Hu
FB
.
Sugar-sweetened beverages, obesity, type 2 diabetes mellitus, and cardiovascular disease risk
.
Circulation
.
2010
;
121
(
11
):
1356
1364
.

41.

Driscoll
KA
,
Corbin
KD
,
Maahs
DM
,
Pratley
R
,
Bishop
FK
,
Kahkoska
A
,
Hood
KK
,
Mayer-Davis
E
;
Advancing Care for Type 1 Diabetes and Obesity Network (ACT1ON)
.
Biopsychosocial aspects of weight management in type 1 diabetes: a review and next steps
.
Curr Diab Rep
.
2017
;
17
(
8
):
58
.

42.

Goebel-Fabbri
AE
.
Disturbed eating behaviors and eating disorders in type 1 diabetes: clinical significance and treatment recommendations
.
Curr Diab Rep
.
2009
;
9
(
2
):
133
139
.

43.

Hanlan
ME
,
Griffith
J
,
Patel
N
,
Jaser
SS
.
Eating disorders and disordered eating in type 1 diabetes: prevalence, screening, and treatment options
.
Curr Diab Rep
.
2013
;
13
(
6
):
909
916
.

44.

Neumark-Sztainer
D
,
Patterson
J
,
Mellin
A
,
Ackard
DM
,
Utter
J
,
Story
M
,
Sockalosky
J
.
Weight control practices and disordered eating behaviors among adolescent females and males with type 1 diabetes: associations with sociodemographics, weight concerns, familial factors, and metabolic outcomes
.
Diabetes Care
.
2002
;
25
(
8
):
1289
1296
.

45.

Peveler
RC
,
Bryden
KS
,
Neil
HA
,
Fairburn
CG
,
Mayou
RA
,
Dunger
DB
,
Turner
HM
.
The relationship of disordered eating habits and attitudes to clinical outcomes in young adult females with type 1 diabetes
.
Diabetes Care
.
2005
;
28
(
1
):
84
88
.

46.

Polonsky
WH
,
Anderson
BJ
,
Lohrer
PA
,
Aponte
JE
,
Jacobson
AM
,
Cole
CF
.
Insulin omission in women with IDDM
.
Diabetes Care
.
1994
;
17
(
10
):
1178
1185
.

47.

Peterson
CM
,
Fischer
S
,
Young-Hyman
D
.
Topical review: a comprehensive risk model for disordered eating in youth with type 1 diabetes
.
J Pediatr Psychol
.
2015
;
40
(
4
):
385
390
.

48.

Pinhas-Hamiel
O
,
Levy-Shraga
Y
.
Eating disorders in adolescents with type 2 and type 1 diabetes
.
Curr Diab Rep
.
2013
;
13
(
2
):
289
297
.

49.

Goebel-Fabbri
AE
,
Fikkan
J
,
Franko
DL
,
Pearson
K
,
Anderson
BJ
,
Weinger
K
.
Insulin restriction and associated morbidity and mortality in women with type 1 diabetes
.
Diabetes Care
.
2008
;
31
(
3
):
415
419
.

50.

Dinneen
S
,
Alzaid
A
,
Turk
D
,
Rizza
R
.
Failure of glucagon suppression contributes to postprandial hyperglycaemia in IDDM
.
Diabetologia
.
1995
;
38
(
3
):
337
343
.

51.

McCrimmon
RJ
,
Sherwin
RS
.
Hypoglycemia in type 1 diabetes
.
Diabetes
.
2010
;
59
(
10
):
2333
2339
.

52.

Matteucci
E
,
Giampietro
O
,
Covolan
V
,
Giustarini
D
,
Fanti
P
,
Rossi
R
.
Insulin administration: present strategies and future directions for a noninvasive (possibly more physiological) delivery
.
Drug Des Devel Ther
.
2015
;
9
:
3109
3118
.

53.

Edgerton
DS
,
Lautz
M
,
Scott
M
,
Everett
CA
,
Stettler
KM
,
Neal
DW
,
Chu
CA
,
Cherrington
AD
.
Insulin’s direct effects on the liver dominate the control of hepatic glucose production
.
J Clin Invest
.
2006
;
116
(
2
):
521
527
.

54.

Martin
C
.
The physiology of amylin and insulin: maintaining the balance between glucose secretion and glucose uptake
.
Diabetes Educ
.
2006
;
32
(
Suppl 3
):
101S
104S
.

55.

Heptulla
RA
,
Rodriguez
LM
,
Mason
KJ
,
Haymond
MW
.
Twenty-four-hour simultaneous subcutaneous basal-bolus administration of insulin and amylin in adolescents with type 1 diabetes decreases postprandial hyperglycemia
.
J Clin Endocrinol Metab
.
2009
;
94
(
5
):
1608
1611
.

56.

Rodriguez
LM
,
Mason
KJ
,
Haymond
MW
,
Heptulla
RA
.
The role of prandial pramlintide in the treatment of adolescents with type 1 diabetes
.
Pediatr Res
.
2007
;
62
(
6
):
746
749
.

57.

Edelman
S
,
Garg
S
,
Frias
J
,
Maggs
D
,
Wang
Y
,
Zhang
B
,
Strobel
S
,
Lutz
K
,
Kolterman
O
.
A double-blind, placebo-controlled trial assessing pramlintide treatment in the setting of intensive insulin therapy in type 1 diabetes
.
Diabetes Care
.
2006
;
29
(
10
):
2189
2195
.

58.

Hoogwerf
BJ
,
Doshi
KB
,
Diab
D
.
Pramlintide, the synthetic analogue of amylin: physiology, pathophysiology, and effects on glycemic control, body weight, and selected biomarkers of vascular risk
.
Vasc Health Risk Manag
.
2008
;
4
(
2
):
355
362
.

59.

Chase
HP
,
Lutz
K
,
Pencek
R
,
Zhang
B
,
Porter
L
.
Pramlintide lowered glucose excursions and was well-tolerated in adolescents with type 1 diabetes: results from a randomized, single-blind, placebo-controlled, crossover study
.
J Pediatr
.
2009
;
155
(
3
):
369
373
.

60.

Weinzimer
SA
,
Sherr
JL
,
Cengiz
E
,
Kim
G
,
Ruiz
JL
,
Carria
L
,
Voskanyan
G
,
Roy
A
,
Tamborlane
WV
.
Effect of pramlintide on prandial glycemic excursions during closed-loop control in adolescents and young adults with type 1 diabetes
.
Diabetes Care
.
2012
;
35
(
10
):
1994
1999
.

61.

The DCCT Research Group
.
Weight gain associated with intensive therapy in the diabetes control and complications trial
.
Diabetes Care
.
1988
;
11
(
7
):
567
573
.

62.

Purnell
JQ
,
Braffett
BH
,
Zinman
B
,
Gubitosi-Klug
RA
,
Sivitz
W
,
Bantle
JP
,
Ziegler
G
,
Cleary
PA
,
Brunzell
JD
.
Impact of excessive weight gain on cardiovascular outcomes in type 1 diabetes: results from the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) study
.
Diabetes Care
.
2017
;
40
(
12
):
1756
1762
.

63.

Nansel
TR
,
Lipsky
LM
,
Iannotti
RJ
.
Cross-sectional and longitudinal relationships of body mass index with glycemic control in children and adolescents with type 1 diabetes mellitus
.
Diabetes Res Clin Pract
.
2013
;
100
(
1
):
126
132
.

64.

Ogden
CL
,
Carroll
MD
,
Kit
BK
,
Flegal
KM
.
Prevalence of obesity and trends in body mass index among US children and adolescents, 1999–2010
.
JAMA
.
2012
;
307
(
5
):
483
490
.

65.

Professional Practice Committee for the Standards of Medical Care in Diabetes—2016. Diabetes Care. 2016;39(Suppl 1):S107–S108
.

66.

Flegal
KM
,
Kruszon-Moran
D
,
Carroll
MD
,
Fryar
CD
,
Ogden
CL
.
Trends in obesity among adults in the United States, 2005 to 2014
.
JAMA
.
2016
;
315
(
21
):
2284
2291
.

67.

Kowalski
A
.
Pathway to artificial pancreas systems revisited: moving downstream
.
Diabetes Care
.
2015
;
38
(
6
):
1036
1043
.

68.

Kropff
J
,
DeVries
JH
.
Continuous glucose monitoring, future products, and update on worldwide artificial pancreas projects
.
Diabetes Technol Ther
.
2016
;
18
(
Suppl 2
):
S253
S263
.

69.

Forlenza
GP
,
Buckingham
B
,
Maahs
DM
.
Progress in diabetes technology: developments in insulin pumps, continuous glucose monitors, and progress towards the artificial pancreas
.
J Pediatr
.
2016
;
169
:
13
20
.

70.

Garg
SK
,
Weinzimer
SA
,
Tamborlane
WV
,
Buckingham
BA
,
Bode
BW
,
Bailey
TS
,
Brazg
RL
,
Ilany
J
,
Slover
RH
,
Anderson
SM
,
Bergenstal
RM
,
Grosman
B
,
Roy
A
,
Cordero
TL
,
Shin
J
,
Lee
SW
,
Kaufman
FR
.
Glucose outcomes with the in-home use of a hybrid closed-loop insulin delivery system in adolescents and adults with type 1 diabetes
.
Diabetes Technol Ther
.
2017
;
19
(
3
):
155
163
.

71.

Saudek
CD
,
Duckworth
WC
,
Giobbie-Hurder
A
,
Henderson
WG
,
Henry
RR
,
Kelley
DE
,
Edelman
SV
,
Zieve
FJ
,
Adler
RA
,
Anderson
JW
,
Anderson
RJ
,
Hamilton
BP
,
Donner
TW
,
Kirkman
MS
,
Morgan
NA
;
Department of Veterans Affairs Implantable Insulin Pump Study Group
.
Implantable insulin pump vs multiple-dose insulin for non-insulin-dependent diabetes mellitus: a randomized clinical trial
.
JAMA
.
1996
;
276
(
16
):
1322
1327
.

72.

Trevitt
S
,
Simpson
S
,
Wood
A
.
Artificial pancreas device systems for the closed-loop control of type 1 diabetes: what systems are in development
?
J Diabetes Sci Technol
.
2016
;
10
(
3
):
714
723
.

73.

Blauw
H
,
van Bon
AC
,
Koops
R
,
DeVries
JH
; on behalf of the PCDIAB Consortium.
Performance and safety of an integrated bihormonal artificial pancreas for fully automated glucose control at home
.
Diabetes Obes Metab
.
2016
;
18
(
7
):
671
677
.

74.

El-Khatib
FH
,
Balliro
C
,
Hillard
MA
,
Magyar
KL
,
Ekhlaspour
L
,
Sinha
M
,
Mondesir
D
,
Esmaeili
A
,
Hartigan
C
,
Thompson
MJ
,
Malkani
S
,
Lock
JP
,
Harlan
DM
,
Clinton
P
,
Frank
E
,
Wilson
DM
,
DeSalvo
D
,
Norlander
L
,
Ly
T
,
Buckingham
BA
,
Diner
J
,
Dezube
M
,
Young
LA
,
Goley
A
,
Kirkman
MS
,
Buse
JB
,
Zheng
H
,
Selagamsetty
RR
,
Damiano
ER
,
Russell
SJ
.
Home use of a bihormonal bionic pancreas versus insulin pump therapy in adults with type 1 diabetes: a multicentre randomised crossover trial
.
Lancet
.
2017
;
389
(
10067
):
369
380
.

75.

Maahs
DM
,
Buckingham
BA
,
Castle
JR
,
Cinar
A
,
Damiano
ER
,
Dassau
E
,
DeVries
JH
,
Doyle
FJ
III
,
Griffen
SC
,
Haidar
A
,
Heinemann
L
,
Hovorka
R
,
Jones
TW
,
Kollman
C
,
Kovatchev
B
,
Levy
BL
,
Nimri
R
,
O’Neal
DN
,
Philip
M
,
Renard
E
,
Russell
SJ
,
Weinzimer
SA
,
Zisser
H
,
Lum
JW
.
Outcome measures for artificial pancreas clinical trials: a consensus report
.
Diabetes Care
.
2016
;
39
(
7
):
1175
1179
.

76.

Kahkoska
AR
,
Mayer-Davis
EJ
,
Hood
KK
,
Maahs
DM
,
Burger
KS
.
Behavioural implications of traditional treatment and closed-loop automated insulin delivery systems in type 1 diabetes: applying a cognitive restraint theory framework
.
Diabet Med
.
2017
;
34
(
11
):
1500
1507
.

77.

Ang
KH
,
Sherr
JL
.
Moving beyond subcutaneous insulin: the application of adjunctive therapies to the treatment of type 1 diabetes
.
Expert Opin Drug Deliv
.
2017
;
14
(
9
):
1113
1131
.

78.

Libman
IM
,
Miller
KM
,
DiMeglio
LA
,
Bethin
KE
,
Katz
ML
,
Shah
A
,
Simmons
JH
,
Haller
MJ
,
Raman
S
,
Tamborlane
WV
,
Coffey
JK
,
Saenz
AM
,
Beck
RW
,
Nadeau
KJ
;
T1D Exchange Clinic Network Metformin RCT Study Group
.
Effect of metformin added to insulin on glycemic control among overweight/obese adolescents with type 1 diabetes: a randomized clinical trial
.
JAMA
.
2015
;
314
(
21
):
2241
2250
.

79.

Vella
S
,
Buetow
L
,
Royle
P
,
Livingstone
S
,
Colhoun
HM
,
Petrie
JR
.
The use of metformin in type 1 diabetes: a systematic review of efficacy
.
Diabetologia
.
2010
;
53
(
5
):
809
820
.

80.

Wang
W
,
Liu
H
,
Xiao
S
,
Liu
S
,
Li
X
,
Yu
P
.
Effects of insulin plus glucagon-like peptide-1 receptor agonists (GLP-1RAs) in treating type 1 diabetes mellitus: a systematic review and meta-analysis
.
Diabetes Ther
.
2017
;
8
(
4
):
727
738
.

81.

Perkins
BA
,
Cherney
DZ
,
Partridge
H
,
Soleymanlou
N
,
Tschirhart
H
,
Zinman
B
,
Fagan
NM
,
Kaspers
S
,
Woerle
HJ
,
Broedl
UC
,
Johansen
OE
.
Sodium-glucose cotransporter 2 inhibition and glycemic control in type 1 diabetes: results of an 8-week open-label proof-of-concept trial
.
Diabetes Care
.
2014
;
37
(
5
):
1480
1483
.

82.

Henry
RR
,
Thakkar
P
,
Tong
C
,
Polidori
D
,
Alba
M
.
Efficacy and safety of canagliflozin, a sodium-glucose cotransporter 2 inhibitor, as add-on to insulin in patients with type 1 diabetes
.
Diabetes Care
.
2015
;
38
(
12
):
2258
2265
.

83.

Peters
AL
,
Buschur
EO
,
Buse
JB
,
Cohan
P
,
Diner
JC
,
Hirsch
IB
.
Euglycemic diabetic ketoacidosis: a potential complication of treatment with sodium-glucose cotransporter 2 inhibition
.
Diabetes Care
.
2015
;
38
(
9
):
1687
1693
.

84.

El Masri
D
,
Ghosh
S
,
Jaber
LA
.
Safety and efficacy of sodium-glucose cotransporter 2 (SGLT2) inhibitors in type 1 diabetes: a systematic review and meta-analysis
.
Diabetes Res Clin Pract
.
2018
;
137
:
83
92
.

85.

Garg
SK
,
Henry
RR
,
Banks
P
,
Buse
JB
,
Davies
MJ
,
Fulcher
GR
,
Pozzilli
P
,
Gesty-Palmer
D
,
Lapuerta
P
,
Simó
R
,
Danne
T
,
McGuire
DK
,
Kushner
JA
,
Peters
A
,
Strumph
P
.
Effects of sotagliflozin added to insulin in patients with type 1 diabetes
.
N Engl J Med
.
2017
;
377
(
24
):
2337
2348
.

86.

Guo
H
,
Fang
C
,
Huang
Y
,
Pei
Y
,
Chen
L
,
Hu
J
.
The efficacy and safety of DPP4 inhibitors in patients with type 1 diabetes: a systematic review and meta-analysis
.
Diabetes Res Clin Pract
.
2016
;
121
:
184
191
.

87.

Karagiannis
T
,
Paschos
P
,
Paletas
K
,
Matthews
DR
,
Tsapas
A
.
Dipeptidyl peptidase-4 inhibitors for treatment of type 2 diabetes mellitus in the clinical setting: systematic review and meta-analysis
.
BMJ
.
2012
;
344
:
e1369
.

88.

White
WB
,
Cannon
CP
,
Heller
SR
,
Nissen
SE
,
Bergenstal
RM
,
Bakris
GL
,
Perez
AT
,
Fleck
PR
,
Mehta
CR
,
Kupfer
S
,
Wilson
C
,
Cushman
WC
,
Zannad
F
;
EXAMINE Investigators
.
Alogliptin after acute coronary syndrome in patients with type 2 diabetes
.
N Engl J Med
.
2013
;
369
(
14
):
1327
1335
.

89.

Scirica
BM
,
Bhatt
DL
,
Braunwald
E
,
Steg
PG
,
Davidson
J
,
Hirshberg
B
,
Ohman
P
,
Frederich
R
,
Wiviott
SD
,
Hoffman
EB
,
Cavender
MA
,
Udell
JA
,
Desai
NR
,
Mosenzon
O
,
McGuire
DK
,
Ray
KK
,
Leiter
LA
,
Raz
I
;
SAVOR-TIMI 53 Steering Committee and Investigators
.
Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus
.
N Engl J Med
.
2013
;
369
(
14
):
1317
1326
.

90.

Green
JB
,
Bethel
MA
,
Armstrong
PW
,
Buse
JB
,
Engel
SS
,
Garg
J
,
Josse
R
,
Kaufman
KD
,
Koglin
J
,
Korn
S
,
Lachin
JM
,
McGuire
DK
,
Pencina
MJ
,
Standl
E
,
Stein
PP
,
Suryawanshi
S
,
Van de Werf
F
,
Peterson
ED
,
Holman
RR
;
TECOS Study Group
.
Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
.
2015
;
373
(
3
):
232
242
.

91.

Lyons
S
,
Herman
J
,
Miller
K
,
Hofer
S
,
Foster
N
,
Rami-Merhar
B
,
Aleppo
G
,
Seufert
J
,
DiMeglio
L
,
Danne
T
,
Maahs
D
,
Holl
R
.
Use of adjuvant pharmacotherapy in type 1 diabetes: international comparison of 49,367 individuals in the DPV and T1D exchange registries
.
Diabetes Care
.
2017
;
40
(
10
):
e139
e140
.

92.

Nathan
DM
,
Genuth
S
,
Lachin
J
,
Cleary
P
,
Crofford
O
,
Davis
M
,
Rand
L
,
Siebert
C
;
Diabetes Control and Complications Trial Research Group
.
The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus
.
N Engl J Med
.
1993
;
329
(
14
):
977
986
.

93.

Haynes
A
,
Hermann
JM
,
Miller
KM
,
Hofer
SE
,
Jones
TW
,
Beck
RW
,
Maahs
DM
,
Davis
EA
,
Holl
RW
;
T1D Exchange, WACDD and DPV registries
.
Severe hypoglycemia rates are not associated with HbA1c: a cross-sectional analysis of 3 contemporary pediatric diabetes registry databases
.
Pediatr Diabetes
.
2017
;
18
(
7
):
643
650
.

94.

Weinstock
RS
,
Xing
D
,
Maahs
DM
,
Michels
A
,
Rickels
MR
,
Peters
AL
,
Bergenstal
RM
,
Harris
B
,
Dubose
SN
,
Miller
KM
,
Beck
RW
;
T1D Exchange Clinic Network
.
Severe hypoglycemia and diabetic ketoacidosis in adults with type 1 diabetes: results from the T1D Exchange clinic registry
.
J Clin Endocrinol Metab
.
2013
;
98
(
8
):
3411
3419
.

95.

Pickup
JC
,
Sutton
AJ
.
Severe hypoglycaemia and glycaemic control in type 1 diabetes: meta-analysis of multiple daily insulin injections compared with continuous subcutaneous insulin infusion
.
Diabet Med
.
2008
;
25
(
7
):
765
774
.

96.

Karges
B
,
Rosenbauer
J
,
Kapellen
T
,
Wagner
VM
,
Schober
E
,
Karges
W
,
Holl
RW
.
Hemoglobin A1c levels and risk of severe hypoglycemia in children and young adults with type 1 diabetes from Germany and Austria: a trend analysis in a cohort of 37,539 patients between 1995 and 2012
.
PLoS Med
.
2014
;
11
(
10
):
e1001742
.

97.

Jones
TW
;
ISPAD Hypoglycemia Guidelines Writing Group
.
Defining relevant hypoglycemia measures in children and adolescents with type 1 diabetes
.
Pediatr Diabetes
.
2018
;
19
(
3
):
354
355
.

98.

Strachan
MW
,
Ewing
FM
,
Frier
BM
,
Harper
A
,
Deary
IJ
.
Food cravings during acute hypoglycaemia in adults with type 1 diabetes
.
Physiol Behav
.
2004
;
80
(
5
):
675
682
.

99.

Merwin
RM
,
Moskovich
AA
,
Dmitrieva
NO
,
Pieper
CF
,
Honeycutt
LK
,
Zucker
NL
,
Surwit
RS
,
Buhi
L
.
Disinhibited eating and weight-related insulin mismanagement among individuals with type 1 diabetes
.
Appetite
.
2014
;
81
:
123
130
.

100.

Purnell
JQ
,
Dev
RK
,
Steffes
MW
,
Cleary
PA
,
Palmer
JP
,
Hirsch
IB
,
Hokanson
JE
,
Brunzell
JD
.
Relationship of family history of type 2 diabetes, hypoglycemia, and autoantibodies to weight gain and lipids with intensive and conventional therapy in the Diabetes Control and Complications Trial
.
Diabetes
.
2003
;
52
(
10
):
2623
2629
.

101.

Boland
EA
,
Grey
M
,
Oesterle
A
,
Fredrickson
L
,
Tamborlane
WV
.
Continuous subcutaneous insulin infusion. A new way to lower risk of severe hypoglycemia, improve metabolic control, and enhance coping in adolescents with type 1 diabetes
.
Diabetes Care
.
1999
;
22
(
11
):
1779
1784
.

102.

Bergenstal
RM
,
Tamborlane
WV
,
Ahmann
A
,
Buse
JB
,
Dailey
G
,
Davis
SN
,
Joyce
C
,
Peoples
T
,
Perkins
BA
,
Welsh
JB
,
Willi
SM
,
Wood
MA
;
STAR 3 Study Group
.
Effectiveness of sensor-augmented insulin-pump therapy in type 1 diabetes
.
N Engl J Med
.
2010
;
363
(
4
):
311
320
.

103.

Lane
W
,
Bailey
TS
,
Gerety
G
,
Gumprecht
J
,
Philis-Tsimikas
A
,
Hansen
CT
,
Nielsen
TSS
,
Warren
M
;
Group Information; SWITCH 1
.
Effect of insulin degludec vs insulin glargine U100 on hypoglycemia in patients with type 1 diabetes: the SWITCH 1 randomized clinical trial
.
JAMA
.
2017
;
318
(
1
):
33
44
.

104.

Hruby
A
,
Manson
JE
,
Qi
L
,
Malik
VS
,
Rimm
EB
,
Sun
Q
,
Willett
WC
,
Hu
FB
.
Determinants and consequences of obesity
.
Am J Public Health
.
2016
;
106
(
9
):
1656
1662
.

105.

Gregg
EW
,
Jakicic
JM
,
Blackburn
G
,
Bloomquist
P
,
Bray
GA
,
Clark
JM
,
Coday
M
,
Curtis
JM
,
Egan
C
,
Evans
M
,
Foreyt
J
,
Foster
G
,
Hazuda
HP
,
Hill
JO
,
Horton
ES
,
Hubbard
VS
,
Jeffery
RW
,
Johnson
KC
,
Kitabchi
AE
,
Knowler
WC
,
Kriska
A
,
Lang
W
,
Lewis
CE
,
Montez
MG
,
Nathan
DM
,
Neiberg
RH
,
Patricio
J
,
Peters
A
,
Pi-Sunyer
X
,
Pownall
H
,
Redmon
B
,
Regensteiner
J
,
Rejeski
J
,
Ribisl
PM
,
Safford
M
,
Stewart
K
,
Trence
D
,
Wadden
TA
,
Wing
RR
,
Yanovski
SZ
;
Look AHEAD Research Group
.
Association of the magnitude of weight loss and changes in physical fitness with long-term cardiovascular disease outcomes in overweight or obese people with type 2 diabetes: a post-hoc analysis of the Look AHEAD randomised clinical trial
.
Lancet Diabetes Endocrinol
.
2016
;
4
(
11
):
913
921
.

106.

Rubin
RR
,
Wadden
TA
,
Bahnson
JL
,
Blackburn
GL
,
Brancati
FL
,
Bray
GA
,
Coday
M
,
Crow
SJ
,
Curtis
JM
,
Dutton
G
,
Egan
C
,
Evans
M
,
Ewing
L
,
Faulconbridge
L
,
Foreyt
J
,
Gaussoin
SA
,
Gregg
EW
,
Hazuda
HP
,
Hill
JO
,
Horton
ES
,
Hubbard
VS
,
Jakicic
JM
,
Jeffery
RW
,
Johnson
KC
,
Kahn
SE
,
Knowler
WC
,
Lang
W
,
Lewis
CE
,
Montez
MG
,
Murillo
A
,
Nathan
DM
,
Patricio
J
,
Peters
A
,
Pi-Sunyer
X
,
Pownall
H
,
Rejeski
WJ
,
Rosenthal
RH
,
Ruelas
V
,
Toledo
K
,
Van Dorsten
B
,
Vitolins
M
,
Williamson
D
,
Wing
RR
,
Yanovski
SZ
,
Zhang
P
;
Look AHEAD Research Group
.
Impact of intensive lifestyle intervention on depression and health-related quality of life in type 2 diabetes: the Look AHEAD Trial
.
Diabetes Care
.
2014
;
37
(
6
):
1544
1553
.

107.

Atkinson
MA
,
Eisenbarth
GS
,
Michels
AW
.
Type 1 diabetes
.
Lancet
.
2014
;
383
(
9911
):
69
82
.

108.

Conway
B
,
Miller
RG
,
Costacou
T
,
Fried
L
,
Kelsey
S
,
Evans
RW
,
Orchard
TJ
.
Adiposity and mortality in type 1 diabetes
.
Int J Obes
.
2009
;
33
(
7
):
796
805
.

109.

Silverstein
J
,
Cheng
P
,
Ruedy
KJ
,
Kollman
C
,
Beck
RW
,
Klingensmith
GJ
,
Wood
JR
,
Willi
S
,
Bacha
F
,
Lee
J
,
Cengiz
E
,
Redondo
MJ
,
Tamborlane
WV
;
Pediatric Diabetes Consortium
.
Depressive symptoms in youth with type 1 or type 2 diabetes: results of the Pediatric Diabetes Consortium Screening Assessment of Depression in Diabetes Study
.
Diabetes Care
.
2015
;
38
(
12
):
2341
2343
.

110.

Harding
JL
,
Shaw
JE
,
Peeters
A
,
Cartensen
B
,
Magliano
DJ
.
Cancer risk among people with type 1 and type 2 diabetes: disentangling true associations, detection bias, and reverse causation
.
Diabetes Care
.
2015
;
38
(
2
):
264
270
.

111.

Ford
ES
,
Giles
WH
,
Dietz
WH
.
Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey
.
JAMA
.
2002
;
287
(
3
):
356
359
.

112.

Nadeau
KJ
,
Regensteiner
JG
,
Bauer
TA
,
Brown
MS
,
Dorosz
JL
,
Hull
A
,
Zeitler
P
,
Draznin
B
,
Reusch
JE
.
Insulin resistance in adolescents with type 1 diabetes and its relationship to cardiovascular function
.
J Clin Endocrinol Metab
.
2010
;
95
(
2
):
513
521
.

113.

Donga
E
,
Dekkers
OM
,
Corssmit
EP
,
Romijn
JA
.
Insulin resistance in patients with type 1 diabetes assessed by glucose clamp studies: systematic review and meta-analysis
.
Eur J Endocrinol
.
2015
;
173
(
1
):
101
109
.

114.

Coleman
SK
,
Rebalka
IA
,
D’Souza
DM
,
Deodhare
N
,
Desjardins
EM
,
Hawke
TJ
.
Myostatin inhibition therapy for insulin-deficient type 1 diabetes
.
Sci Rep
.
2016
;
6
(
1
):
32495
.

115.

Alberti
KG
,
Zimmet
P
,
Shaw
J
;
IDF Epidemiology Task Force Consensus Group
.
The metabolic syndrome—a new worldwide definition
.
Lancet
.
2005
;
366
(
9491
):
1059
1062
.

116.

Grundy
SM
,
Cleeman
JI
,
Daniels
SR
,
Donato
KA
,
Eckel
RH
,
Franklin
BA
,
Gordon
DJ
,
Krauss
RM
,
Savage
PJ
,
Smith
SC
Jr
,
Spertus
JA
,
Costa
F
;
American Heart Association; National Heart, Lung, and Blood Institute
.
Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute scientific statement
.
Circulation
.
2005
;
112
(
17
):
2735
2752
.

117.

Alberti
KG
,
Zimmet
P
,
Shaw
J
.
Metabolic syndrome–a new worldwide definition. A Consensus Statement from the International Diabetes Federation
.
Diabet Med
.
2006
;
23
:
469
480
.

118.

World Health Organization. Definition, diagnosis, and classification of diabetes mellitus and its complications: report of a WHO consultation. Part 1, Diagnosis and classification of diabetes mellitus. Geneva, Switzerland: World Health Organization; 1999
.

119.

Pambianco
G
,
Costacou
T
,
Orchard
TJ
.
The prediction of major outcomes of type 1 diabetes: a 12-year prospective evaluation of three separate definitions of the metabolic syndrome and their components and estimated glucose disposal rate: the Pittsburgh Epidemiology of Diabetes Complications Study experience
.
Diabetes Care
.
2007
;
30
(
5
):
1248
1254
.

120.

Chillarón
JJ
,
Goday
A
,
Flores-Le-Roux
JA
,
Benaiges
D
,
Carrera
MJ
,
Puig
J
,
Cano-Pérez
JF
,
Pedro-Botet
J
.
Estimated glucose disposal rate in assessment of the metabolic syndrome and microvascular complications in patients with type 1 diabetes
.
J Clin Endocrinol Metab
.
2009
;
94
(
9
):
3530
3534
.

121.

National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III)
.
Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report
.
Circulation
.
2002
;
106
(
25
):
3143
3421
.

122.

Younossi
Z
,
Anstee
QM
,
Marietti
M
,
Hardy
T
,
Henry
L
,
Eslam
M
,
George
J
,
Bugianesi
E
.
Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention
.
Nat Rev Gastroenterol Hepatol
.
2018
;
15
(
1
):
11
20
.

123.

Loomba
R
,
Sanyal
AJ
.
The global NAFLD epidemic
.
Nat Rev Gastroenterol Hepatol
.
2013
;
10
(
11
):
686
690
.

124.

Bril
F
,
Cusi
K
.
Management of nonalcoholic fatty liver disease in patients with type 2 diabetes: a call to action
.
Diabetes Care
.
2017
;
40
(
3
):
419
430
.

125.

Anderson
EL
,
Howe
LD
,
Jones
HE
,
Higgins
JP
,
Lawlor
DA
,
Fraser
A
.
The prevalence of non-alcoholic fatty liver disease in children and adolescents: a systematic review and meta-analysis
.
PLoS One
.
2015
;
10
(
10
):
e0140908
.

126.

Serra-Planas
E
,
Aguilera
E
,
Castro
L
,
Rodríguez
R
,
Salinas
I
,
Lucas
A
,
Joaquín
C
,
Puig
R
,
Mauricio
D
,
Puig-Domingo
M
.
Low prevalence of non-alcoholic fatty liver disease in patients with type 1 diabetes is associated with decreased subclinical cardiovascular disease
.
J Diabetes
.
2017
;
9
(
12
):
1065
1072
.

127.

Targher
G
,
Bertolini
L
,
Padovani
R
,
Rodella
S
,
Zoppini
G
,
Pichiri
I
,
Sorgato
C
,
Zenari
L
,
Bonora
E
.
Prevalence of non-alcoholic fatty liver disease and its association with cardiovascular disease in patients with type 1 diabetes
.
J Hepatol
.
2010
;
53
(
4
):
713
718
.

128.

Targher
G
,
Mantovani
A
,
Pichiri
I
,
Mingolla
L
,
Cavalieri
V
,
Mantovani
W
,
Pancheri
S
,
Trombetta
M
,
Zoppini
G
,
Chonchol
M
,
Byrne
CD
,
Bonora
E
.
Nonalcoholic fatty liver disease is independently associated with an increased incidence of chronic kidney disease in patients with type 1 diabetes
.
Diabetes Care
.
2014
;
37
(
6
):
1729
1736
.

129.

Mantovani
A
,
Rigolon
R
,
Mingolla
L
,
Pichiri
I
,
Cavalieri
V
,
Salvotelli
L
,
Stoico
V
,
Zoppini
G
,
Bonora
E
,
Targher
G
.
Nonalcoholic fatty liver disease is associated with an increased prevalence of distal symmetric polyneuropathy in adult patients with type 1 diabetes
.
J Diabetes Complications
.
2017
;
31
(
6
):
1021
1026
.

130.

Regnell
SE
,
Peterson
P
,
Trinh
L
,
Broberg
P
,
Leander
P
,
Lernmark
Å
,
Månsson
S
,
Elding Larsson
H
.
Magnetic resonance imaging reveals altered distribution of hepatic fat in children with type 1 diabetes compared to controls
.
Metabolism
.
2015
;
64
(
8
):
872
878
.

131.

Petit
JM
,
Pedro
L
,
Guiu
B
,
Duvillard
L
,
Bouillet
B
,
Jooste
V
,
Habchi
M
,
Crevisy
E
,
Fourmont
C
,
Buffier
P
,
Hillon
P
,
Cercueil
JP
,
Verges
B
.
Type 1 diabetes is not associated with an increased prevalence of hepatic steatosis
.
Diabet Med
.
2015
;
32
(
12
):
1648
1651
.

132.

Llauradó
G
,
Sevastianova
K
,
Sädevirta
S
,
Hakkarainen
A
,
Lundbom
N
,
Orho-Melander
M
,
Groop
PH
,
Forsblom
C
,
Yki-Järvinen
H
.
Liver fat content and hepatic insulin sensitivity in overweight patients with type 1 diabetes
.
J Clin Endocrinol Metab
.
2015
;
100
(
2
):
607
616
.

133.

Mantovani
A
,
Mingolla
L
,
Rigolon
R
,
Pichiri
I
,
Cavalieri
V
,
Zoppini
G
,
Lippi
G
,
Bonora
E
,
Targher
G
.
Nonalcoholic fatty liver disease is independently associated with an increased incidence of cardiovascular disease in adult patients with type 1 diabetes
.
Int J Cardiol
.
2016
;
225
:
387
391
.

134.

Targher
G
,
Pichiri
I
,
Zoppini
G
,
Trombetta
M
,
Bonora
E
.
Increased prevalence of cardiovascular disease in type 1 diabetic patients with non-alcoholic fatty liver disease
.
J Endocrinol Invest
.
2012
;
35
(
5
):
535
540
.

135.

Bulum
T
,
Kolarić
B
,
Duvnjak
L
,
Duvnjak
M
.
Nonalcoholic fatty liver disease markers are associated with insulin resistance in type 1 diabetes
.
Dig Dis Sci
.
2011
;
56
(
12
):
3655
3663
.

136.

Purnell
JQ
,
Hokanson
JE
,
Marcovina
SM
,
Steffes
MW
,
Cleary
PA
,
Brunzell
JD
.
Effect of excessive weight gain with intensive therapy of type 1 diabetes on lipid levels and blood pressure: results from the DCCT. Diabetes Control and Complications Trial
.
JAMA
.
1998
;
280
(
2
):
140
146
.

137.

Rodrigues
TC
,
Veyna
AM
,
Haarhues
MD
,
Kinney
GL
,
Rewers
M
,
Snell-Bergeon
JK
.
Obesity and coronary artery calcium in diabetes: the Coronary Artery Calcification in Type 1 Diabetes (CACTI) study
.
Diabetes Technol Ther
.
2011
;
13
(
10
):
991
996
.

138.

Kuwahara
K
,
Honda
T
,
Nakagawa
T
,
Yamamoto
S
,
Hayashi
T
,
Mizoue
T
.
Body mass index trajectory patterns and changes in visceral fat and glucose metabolism before the onset of type 2 diabetes
.
Sci Rep
.
2017
;
7
(
1
):
43521
.

139.

Nguyen
KH
,
Ande
SR
,
Mishra
S
.
Obesity-related abnormalities couple environmental triggers with genetic susceptibility in adult-onset T1D
.
Biochem Biophys Res Commun
.
2016
;
470
(
1
):
94
100
.

140.

Redondo
MJ
,
Rodriguez
LM
,
Haymond
MW
,
Hampe
CS
,
Smith
EO
,
Balasubramanyam
A
,
Devaraj
S
.
Serum adiposity-induced biomarkers in obese and lean children with recently diagnosed autoimmune type 1 diabetes
.
Pediatr Diabetes
.
2014
;
15
(
8
):
543
549
.

141.

Verbeeten
KC
,
Elks
CE
,
Daneman
D
,
Ong
KK
.
Association between childhood obesity and subsequent type 1 diabetes: a systematic review and meta-analysis
.
Diabet Med
.
2011
;
28
(
1
):
10
18
.

142.

Betts
P
,
Mulligan
J
,
Ward
P
,
Smith
B
,
Wilkin
T
.
Increasing body weight predicts the earlier onset of insulin-dependant diabetes in childhood: testing the “accelerator hypothesis” (2)
.
Diabet Med
.
2005
;
22
(
2
):
144
151
.

143.

Cedillo
M
,
Libman
IM
,
Arena
VC
,
Zhou
L
,
Trucco
M
,
Ize-Ludlow
D
,
Pietropaolo
M
,
Becker
DJ
.
Obesity, islet cell autoimmunity, and cardiovascular risk factors in youth at onset of type 1 autoimmune diabetes
.
J Clin Endocrinol Metab
.
2015
;
100
(
1
):
E82
E86
.

144.

Sosenko
JM
,
Krischer
JP
,
Palmer
JP
,
Mahon
J
,
Cowie
C
,
Greenbaum
CJ
,
Cuthbertson
D
,
Lachin
JM
,
Skyler
JS
;
Diabetes Prevention Trial-Type 1 Study Group
.
A risk score for type 1 diabetes derived from autoantibody-positive participants in the diabetes prevention trial-type 1
.
Diabetes Care
.
2008
;
31
(
3
):
528
533
.

145.

Galgani
M
,
Nugnes
R
,
Bruzzese
D
,
Perna
F
,
De Rosa
V
,
Procaccini
C
,
Mozzillo
E
,
Cilio
CM
,
Elding Larsson
H
,
Lernmark
A
,
La Cava
A
,
Franzese
A
,
Matarese
G
.
Meta-immunological profiling of children with type 1 diabetes identifies new biomarkers to monitor disease progression
.
Diabetes
.
2013
;
62
(
7
):
2481
2491
.

146.

Kuchlbauer
V
,
Vogel
M
,
Gausche
R
,
Kapellen
T
,
Rothe
U
,
Vogel
C
,
Pfäffle
R
,
Kiess
W
.
High birth weights but not excessive weight gain prior to manifestation are related to earlier onset of diabetes in childhood: “accelerator hypothesis” revisited
.
Pediatr Diabetes
.
2014
;
15
(
6
):
428
435
.

147.

Winkler
C
,
Marienfeld
S
,
Zwilling
M
,
Bonifacio
E
,
Ziegler
AG
.
Is islet autoimmunity related to insulin sensitivity or body weight in children of parents with type 1 diabetes
?
Diabetologia
.
2009
;
52
(
10
):
2072
2078
.

148.

Meah
FA
,
DiMeglio
LA
,
Greenbaum
CJ
,
Blum
JS
,
Sosenko
JM
,
Pugliese
A
,
Geyer
S
,
Xu
P
,
Evans-Molina
C
;
Type 1 Diabetes Trial/Net Study Group
.
The relationship between BMI and insulin resistance and progression from single to multiple autoantibody positivity and type 1 diabetes among TrialNet Pathway to Prevention participants
.
Diabetologia
.
2016
;
59
(
6
):
1186
1195
.

149.

Redondo
MJ
,
Rodriguez
LM
,
Escalante
M
,
O’Brian Smith
E
,
Balasubramanyam
A
,
Haymond
MW
.
Beta cell function and BMI in ethnically diverse children with newly diagnosed autoimmune type 1 diabetes
.
Pediatr Diabetes
.
2012
;
13
(
7
):
564
571
.

150.

Dabelea
D
,
D’Agostino
RB
Jr
,
Mayer-Davis
EJ
,
Pettitt
DJ
,
Imperatore
G
,
Dolan
LM
,
Pihoker
C
,
Hillier
TA
,
Marcovina
SM
,
Linder
B
,
Ruggiero
AM
,
Hamman
RF
;
SEARCH for Diabetes in Youth Study Group
.
Testing the accelerator hypothesis: body size, β-cell function, and age at onset of type 1 (autoimmune) diabetes
.
Diabetes Care
.
2006
;
29
(
2
):
290
294
.

151.

Woods
SC
,
Seeley
RJ
,
Porte
D
Jr
,
Schwartz
MW
.
Signals that regulate food intake and energy homeostasis
.
Science
.
1998
;
280
(
5368
):
1378
1383
.

152.

Donahoo
WT
,
Levine
JA
,
Melanson
EL
.
Variability in energy expenditure and its components
.
Curr Opin Clin Nutr Metab Care
.
2004
;
7
(
6
):
599
605
.

153.

Ravussin
E
,
Lillioja
S
,
Anderson
TE
,
Christin
L
,
Bogardus
C
.
Determinants of 24-hour energy expenditure in man. Methods and results using a respiratory chamber
.
J Clin Invest
.
1986
;
78
(
6
):
1568
1578
.

154.

Burger
KS
,
Sanders
AJ
,
Gilbert
JR
.
Hedonic hunger is related to increased neural and perceptual responses to cues of palatable food and motivation to consume: evidence from 3 independent investigations
.
J Nutr
.
2016
;
146
(
9
):
1807
1812
.

155.

Schrauwen
P
,
van Marken Lichtenbelt
WD
,
Westerterp
KR
.
Fat and carbohydrate balances during adaptation to a high-fat diet
.
Am J Clin Nutr
.
2000
;
72
(
5
):
1239
1240
.

156.

Sparks
LM
,
Ukropcova
B
,
Smith
J
,
Pasarica
M
,
Hymel
D
,
Xie
H
,
Bray
GA
,
Miles
JM
,
Smith
SR
.
Relation of adipose tissue to metabolic flexibility
.
Diabetes Res Clin Pract
.
2009
;
83
(
1
):
32
43
.

157.

Jumpertz
R
,
Le
DS
,
Turnbaugh
PJ
,
Trinidad
C
,
Bogardus
C
,
Gordon
JI
,
Krakoff
J
.
Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans
.
Am J Clin Nutr
.
2011
;
94
(
1
):
58
65
.

158.

Jacob
AN
,
Salinas
K
,
Adams-Huet
B
,
Raskin
P
.
Potential causes of weight gain in type 1 diabetes mellitus
.
Diabetes Obes Metab
.
2006
;
8
(
4
):
404
411
.

159.

Cunningham
KM
,
Daly
J
,
Horowitz
M
,
Read
NW
.
Gastrointestinal adaptation to diets of differing fat composition in human volunteers
.
Gut
.
1991
;
32
(
5
):
483
486
.

160.

Gallagher
D
,
Albu
J
,
He
Q
,
Heshka
S
,
Boxt
L
,
Krasnow
N
,
Elia
M
.
Small organs with a high metabolic rate explain lower resting energy expenditure in African American than in white adults
.
Am J Clin Nutr
.
2006
;
83
(
5
):
1062
1067
.

161.

Bosy-Westphal
A
,
Reinecke
U
,
Schlörke
T
,
Illner
K
,
Kutzner
D
,
Heller
M
,
Müller
MJ
.
Effect of organ and tissue masses on resting energy expenditure in underweight, normal weight and obese adults
.
Int J Obes Relat Metab Disord
.
2004
;
28
(
1
):
72
79
.

162.

Cummings
DE
,
Overduin
J
.
Gastrointestinal regulation of food intake
.
J Clin Invest
.
2007
;
117
(
1
):
13
23
.

163.

Karl
JP
,
Young
AJ
,
Rood
JC
,
Montain
SJ
.
Independent and combined effects of eating rate and energy density on energy intake, appetite, and gut hormones
.
Obesity (Silver Spring)
.
2013
;
21
(
3
):
E244
E252
.

164.

Schwartz
MW
.
Central nervous system regulation of food intake
.
Obesity (Silver Spring)
.
2006
;
14
(
Suppl 1
):
1S
8S
.

165.

Begg
DP
,
Woods
SC
.
The endocrinology of food intake
.
Nat Rev Endocrinol
.
2013
;
9
(
10
):
584
597
.

166.

Goodpaster
BH
,
Sparks
LM
.
Metabolic flexibility in health and disease
.
Cell Metab
.
2017
;
25
(
5
):
1027
1036
.

167.

Mayer-Davis
EJ
,
Nichols
M
,
Liese
AD
,
Bell
RA
,
Dabelea
DM
,
Johansen
JM
,
Pihoker
C
,
Rodriguez
BL
,
Thomas
J
,
Williams
D
;
SEARCH for Diabetes in Youth Study Group
.
Dietary intake among youth with diabetes: the SEARCH for Diabetes in Youth Study
.
J Am Diet Assoc
.
2006
;
106
(
5
):
689
697
.

168.

Mendez
MA
,
Sotres-Alvarez
D
,
Miles
DR
,
Slining
MM
,
Popkin
BM
.
Shifts in the recent distribution of energy intake among U.S. children aged 2–18 years reflect potential abatement of earlier declining trends
.
J Nutr
.
2014
;
144
(
8
):
1291
1297
.

169.

American Diabetes Association
.
4. Lifestyle Management
.
Diabetes Care
.
2017
;
40
(
Suppl 1
):
S33
S43
.

170.

Patton
SR
.
Adherence to diet in youth with type 1 diabetes
.
J Am Diet Assoc
.
2011
;
111
(
4
):
550
555
.

171.

Vitolins
MZ
,
Anderson
AM
,
Delahanty
L
,
Raynor
H
,
Miller
GD
,
Mobley
C
,
Reeves
R
,
Yamamoto
M
,
Champagne
C
,
Wing
RR
,
Mayer-Davis
E
;
Look AHEAD Research Group
.
Action for Health in Diabetes (Look AHEAD) trial: baseline evaluation of selected nutrients and food group intake
.
J Am Diet Assoc
.
2009
;
109
(
8
):
1367
1375
.

172.

An
R
,
Burd
NA
.
Change in daily energy intake associated with pairwise compositional change in carbohydrate, fat and protein intake among US adults, 1999–2010
.
Public Health Nutr
.
2015
;
18
(
8
):
1343
1352
.

173.

Smart
CE
,
Annan
F
,
Bruno
LP
,
Higgins
LA
,
Acerini
CL
;
International Society for Pediatric and Adolescent Diabetes
.
ISPAD Clinical Practice Consensus Guidelines 2014. Nutritional management in children and adolescents with diabetes
.
Pediatr Diabetes
.
2014
;
15
(
Suppl 20
):
135
153
.

174.

Deane
AM
,
Nguyen
NQ
,
Stevens
JE
,
Fraser
RJ
,
Holloway
RH
,
Besanko
LK
,
Burgstad
C
,
Jones
KL
,
Chapman
MJ
,
Rayner
CK
,
Horowitz
M
.
Endogenous glucagon-like peptide-1 slows gastric emptying in healthy subjects, attenuating postprandial glycemia
.
J Clin Endocrinol Metab
.
2010
;
95
(
1
):
215
221
.

175.

Vilsbøll
T
,
Krarup
T
,
Madsbad
S
,
Holst
JJ
.
Both GLP-1 and GIP are insulinotropic at basal and postprandial glucose levels and contribute nearly equally to the incretin effect of a meal in healthy subjects
.
Regul Pept
.
2003
;
114
(
2–3
):
115
121
.

176.

Bagger
JI
,
Knop
FK
,
Lund
A
,
Vestergaard
H
,
Holst
JJ
,
Vilsbøll
T
.
Impaired regulation of the incretin effect in patients with type 2 diabetes
.
J Clin Endocrinol Metab
.
2011
;
96
(
3
):
737
745
.

177.

Greenbaum
CJ
,
Prigeon
RL
,
D’Alessio
DA
.
Impaired β-cell function, incretin effect, and glucagon suppression in patients with type 1 diabetes who have normal fasting glucose
.
Diabetes
.
2002
;
51
(
4
):
951
957
.

178.

Vilsbøll
T
,
Krarup
T
,
Sonne
J
,
Madsbad
S
,
Vølund
A
,
Juul
AG
,
Holst
JJ
.
Incretin secretion in relation to meal size and body weight in healthy subjects and people with type 1 and type 2 diabetes mellitus
.
J Clin Endocrinol Metab
.
2003
;
88
(
6
):
2706
2713
.

179.

Dupre
J
.
Glycaemic effects of incretins in Type 1 diabetes mellitus: a concise review, with emphasis on studies in humans
.
Regul Pept
.
2005
;
128
(
2
):
149
157
.

180.

Clemmensen
C
,
Müller
TD
,
Woods
SC
,
Berthoud
HR
,
Seeley
RJ
,
Tschöp
MH
.
Gut-brain cross-talk in metabolic control
.
Cell
.
2017
;
168
(
5
):
758
774
.

181.

Huml
M
,
Kobr
J
,
Siala
K
,
Varvařovská
J
,
Pomahačová
R
,
Karlíková
M
,
Sýkora
J
.
Gut peptide hormones and pediatric type 1 diabetes mellitus
.
Physiol Res
.
2011
;
60
(
4
):
647
658
.

182.

Kiewiet
RM
,
van Aken
MO
,
van der Weerd
K
,
Uitterlinden
P
,
Themmen
AP
,
Hofland
LJ
,
de Rijke
YB
,
Delhanty
PJ
,
Ghigo
E
,
Abribat
T
,
van der Lely
AJ
.
Effects of acute administration of acylated and unacylated ghrelin on glucose and insulin concentrations in morbidly obese subjects without overt diabetes
.
Eur J Endocrinol
.
2009
;
161
(
4
):
567
573
.

183.

Tschöp
M
,
Weyer
C
,
Tataranni
PA
,
Devanarayan
V
,
Ravussin
E
,
Heiman
ML
.
Circulating ghrelin levels are decreased in human obesity
.
Diabetes
.
2001
;
50
(
4
):
707
709
.

184.

Ikezaki
A
,
Hosoda
H
,
Ito
K
,
Iwama
S
,
Miura
N
,
Matsuoka
H
,
Kondo
C
,
Kojima
M
,
Kangawa
K
,
Sugihara
S
.
Fasting plasma ghrelin levels are negatively correlated with insulin resistance and PAI-1, but not with leptin, in obese children and adolescents
.
Diabetes
.
2002
;
51
(
12
):
3408
3411
.

185.

McLaughlin
T
,
Abbasi
F
,
Lamendola
C
,
Frayo
RS
,
Cummings
DE
.
Plasma ghrelin concentrations are decreased in insulin-resistant obese adults relative to equally obese insulin-sensitive controls
.
J Clin Endocrinol Metab
.
2004
;
89
(
4
):
1630
1635
.

186.

Arafat
AM
,
Weickert
MO
,
Adamidou
A
,
Otto
B
,
Perschel
FH
,
Spranger
J
,
Möhlig
M
,
Pfeiffer
AF
.
The impact of insulin-independent, glucagon-induced suppression of total ghrelin on satiety in obesity and type 1 diabetes mellitus
.
J Clin Endocrinol Metab
.
2013
;
98
(
10
):
4133
4142
.

187.

Soriano-Guillén
L
,
Barrios
V
,
Lechuga-Sancho
A
,
Chowen
JA
,
Argente
J
.
Response of circulating ghrelin levels to insulin therapy in children with newly diagnosed type 1 diabetes mellitus
.
Pediatr Res
.
2004
;
55
(
5
):
830
835
.

188.

Ashraf
A
,
Mick
G
,
Meleth
S
,
Wang
X
,
McCormick
K
.
Insulin treatment reduces pre-prandial plasma ghrelin concentrations in children with type 1 diabetes
.
Med Sci Monit
.
2007
;
13
(
12
):
CR533
CR537
.

189.

Chelikani
PK
,
Haver
AC
,
Reidelberger
RD
.
Ghrelin attenuates the inhibitory effects of glucagon-like peptide-1 and peptide YY(3-36) on food intake and gastric emptying in rats
.
Diabetes
.
2006
;
55
(
11
):
3038
3046
.

190.

Enç
FY
,
Imeryüz
N
,
Akin
L
,
Turoğlu
T
,
Dede
F
,
Haklar
G
,
Tekeşin
N
,
Bekiroğlu
N
,
Yeğen
BC
,
Rehfeld
JF
,
Holst
JJ
,
Ulusoy
NB
.
Inhibition of gastric emptying by acarbose is correlated with GLP-1 response and accompanied by CCK release
.
Am J Physiol Gastrointest Liver Physiol
.
2001
;
281
(
3
):
G752
G763
.

191.

Migoya
EM
,
Bergeron
R
,
Miller
JL
,
Snyder
RN
,
Tanen
M
,
Hilliard
D
,
Weiss
B
,
Larson
P
,
Gutierrez
M
,
Jiang
G
,
Liu
F
,
Pryor
KA
,
Yao
J
,
Zhu
L
,
Holst
JJ
,
Deacon
C
,
Herman
G
,
Thornberry
N
,
Amatruda
J
,
Williams-Herman
D
,
Wagner
JA
,
SinhaRoy
R
.
Dipeptidyl peptidase-4 inhibitors administered in combination with metformin result in an additive increase in the plasma concentration of active GLP-1
.
Clin Pharmacol Ther
.
2010
;
88
(
6
):
801
808
.

192.

Edwards
CM
,
Todd
JF
,
Mahmoudi
M
,
Wang
Z
,
Wang
RM
,
Ghatei
MA
,
Bloom
SR
.
Glucagon-like peptide 1 has a physiological role in the control of postprandial glucose in humans: studies with the antagonist exendin 9-39
.
Diabetes
.
1999
;
48
(
1
):
86
93
.

193.

Karamanakos
SN
,
Vagenas
K
,
Kalfarentzos
F
,
Alexandrides
TK
.
Weight loss, appetite suppression, and changes in fasting and postprandial ghrelin and peptide-YY levels after Roux-en-Y gastric bypass and sleeve gastrectomy: a prospective, double blind study
.
Ann Surg
.
2008
;
247
(
3
):
401
407
.

194.

Pournaras
DJ
,
Glicksman
C
,
Vincent
RP
,
Kuganolipava
S
,
Alaghband-Zadeh
J
,
Mahon
D
,
Bekker
JH
,
Ghatei
MA
,
Bloom
SR
,
Walters
JR
,
Welbourn
R
,
le Roux
CW
.
The role of bile after Roux-en-Y gastric bypass in promoting weight loss and improving glycaemic control
.
Endocrinology
.
2012
;
153
(
8
):
3613
3619
.

195.

Kielgast
U
,
Holst
JJ
,
Madsbad
S
.
Antidiabetic actions of endogenous and exogenous GLP-1 in type 1 diabetic patients with and without residual β-cell function
.
Diabetes
.
2011
;
60
(
5
):
1599
1607
.

196.

Arafat
MA
,
Otto
B
,
Rochlitz
H
,
Tschöp
M
,
Bähr
V
,
Möhlig
M
,
Diederich
S
,
Spranger
J
,
Pfeiffer
AF
.
Glucagon inhibits ghrelin secretion in humans
.
Eur J Endocrinol
.
2005
;
153
(
3
):
397
402
.

197.

Woods
SC
,
Porte
D
Jr
,
Bobbioni
E
,
Ionescu
E
,
Sauter
JF
,
Rohner-Jeanrenaud
F
,
Jeanrenaud
B
.
Insulin: its relationship to the central nervous system and to the control of food intake and body weight
.
Am J Clin Nutr
.
1985
;
42
(
5
,
Suppl
):
1063
1071
.

198.

Woods
SC
,
Lutz
TA
,
Geary
N
,
Langhans
W
.
Pancreatic signals controlling food intake; insulin, glucagon and amylin
.
Philos Trans R Soc Lond B Biol Sci
.
2006
;
361
(
1471
):
1219
1235
.

199.

Lutz
TA
.
Control of food intake and energy expenditure by amylin-therapeutic implications
.
Int J Obes
.
2009
;
33
(
Suppl 1
):
S24
S27
.

200.

Hieronymus
L
,
Griffin
S
.
Role of amylin in type 1 and type 2 diabetes
.
Diabetes Educ
.
2015
;
41
(
1
,
Suppl
):
47S
56S
.

201.

Prinz
P
,
Hofmann
T
,
Ahnis
A
,
Elbelt
U
,
Goebel-Stengel
M
,
Klapp
BF
,
Rose
M
,
Stengel
A
.
Plasma bile acids show a positive correlation with body mass index and are negatively associated with cognitive restraint of eating in obese patients
.
Front Neurosci
.
2015
;
9
:
199
.

202.

Wu
T
,
Bound
MJ
,
Standfield
SD
,
Gedulin
B
,
Jones
KL
,
Horowitz
M
,
Rayner
CK
.
Effects of rectal administration of taurocholic acid on glucagon-like peptide-1 and peptide YY secretion in healthy humans
.
Diabetes Obes Metab
.
2013
;
15
(
5
):
474
477
.

203.

Roberts
RE
,
Glicksman
C
,
Alaghband-Zadeh
J
,
Sherwood
RA
,
Akuji
N
,
le Roux
CW
.
The relationship between postprandial bile acid concentration, GLP-1, PYY and ghrelin
.
Clin Endocrinol (Oxf)
.
2011
;
74
(
1
):
67
72
.

204.

Roberts
RE
,
Alaghband-Zadeh
J
,
Le Roux
CW
. The role of bile acids in gut-hormone-induced weight loss after bariatric surgery: implications for appetite control and diabetes. In:
Preedy
VR
,
Watson
RR
,
Martin
CR
, eds.
Handbook of Behavior, Food and Nutrition
.
New York, NY
:
Springer New York
;
2011
:
1317
1330
.

205.

Brufau
G
,
Stellaard
F
,
Prado
K
,
Bloks
VW
,
Jonkers
E
,
Boverhof
R
,
Kuipers
F
,
Murphy
EJ
.
Improved glycemic control with colesevelam treatment in patients with type 2 diabetes is not directly associated with changes in bile acid metabolism
.
Hepatology
.
2010
;
52
(
4
):
1455
1464
.

206.

Liu
G
,
Arnaud-Dabernat
S
,
Kritzik
MR
,
Kayali
AG
,
Zhang
YQ
,
Sarvetnick
N
.
PYY in the expanding pancreatic epithelium
.
Endocrine
.
2006
;
30
(
1
):
103
112
.

207.

Yu
T
,
Yang
HS
,
Lu
XJ
,
Xia
ZS
,
Ouyang
H
,
Shan
TD
,
Huang
CZ
,
Chen
QK
.
Association of bactericidal dysfunction of Paneth cells in streptozocin-induced diabetic mice with insulin deficiency
.
Med Sci Monit
.
2016
;
22
:
3062
3072
.

208.

Talchai
C
,
Xuan
S
,
Kitamura
T
,
DePinho
RA
,
Accili
D
.
Generation of functional insulin-producing cells in the gut by Foxo1 ablation
.
Nat Genet
.
2012
;
44
:
406
412
.

209.

Cheung
AT
,
Dayanandan
B
,
Lewis
JT
,
Korbutt
GS
,
Rajotte
RV
,
Bryer-Ash
M
,
Boylan
MO
,
Wolfe
MM
,
Kieffer
TJ
.
Glucose-dependent insulin release from genetically engineered K cells
.
Science
.
2000
;
290
(
5498
):
1959
1962
.

210.

Unniappan
S
,
Wideman
RD
,
Donald
C
,
Gunn
V
,
Wall
JL
,
Zhang
QX
,
Webber
TD
,
Cheung
AT
,
Kieffer
TJ
.
Treatment of diabetes by transplantation of drug-inducible insulin-producing gut cells
.
J Mol Med (Berl)
.
2009
;
87
(
7
):
703
712
.

211.

Han
J
,
Lee
HH
,
Kwon
H
,
Shin
S
,
Yoon
JW
,
Jun
HS
.
Engineered enteroendocrine cells secrete insulin in response to glucose and reverse hyperglycemia in diabetic mice
.
Mol Ther
.
2007
;
15
(
6
):
1195
1202
.

212.

Lam
YY
,
Ravussin
E
.
Analysis of energy metabolism in humans: a review of methodologies
.
Mol Metab
.
2016
;
5
(
11
):
1057
1071
.

213.

Hebert
SL
,
Nair
KS
.
Protein and energy metabolism in type 1 diabetes
.
Clin Nutr
.
2010
;
29
(
1
):
13
17
.

214.

Rigalleau
V
,
Lasseur
C
,
Pécheur
S
,
Chauveau
P
,
Combe
C
,
Perlemoine
C
,
Baillet
L
,
Gin
H
.
Resting energy expenditure in uremic, diabetic, and uremic diabetic subjects
.
J Diabetes Complications
.
2004
;
18
(
4
):
237
241
.

215.

Carlson
MG
,
Campbell
PJ
.
Intensive insulin therapy and weight gain in IDDM
.
Diabetes
.
1993
;
42
(
12
):
1700
1707
.

216.

Greco
AV
,
Tataranni
PA
,
Mingrone
G
,
De Gaetano
A
,
Manto
A
,
Cotroneo
P
,
Ghirlanda
G
.
Daily energy metabolism in patients with type 1 diabetes mellitus
.
J Am Coll Nutr
.
1995
;
14
(
3
):
286
291
.

217.

Nair
KS
,
Halliday
D
,
Garrow
JS
.
Increased energy expenditure in poorly controlled type 1 (insulin-dependent) diabetic patients
.
Diabetologia
.
1984
;
27
(
1
):
13
16
.

218.

Charlton
MR
,
Nair
KS
.
Role of hyperglucagonemia in catabolism associated with type 1 diabetes: effects on leucine metabolism and the resting metabolic rate
.
Diabetes
.
1998
;
47
(
11
):
1748
1756
.

219.

Nair
KS
.
Hyperglucagonemia increases resting metabolic rate in man during insulin deficiency
.
J Clin Endocrinol Metab
.
1987
;
64
(
5
):
896
901
.

220.

Tessari
P
,
Inchiostro
S
,
Barazzoni
R
,
Zanetti
M
,
Vettore
M
,
Biolo
G
,
Iori
E
,
Kiwanuka
E
,
Tiengo
A
.
Hyperglucagonemia stimulates phenylalanine oxidation in humans
.
Diabetes
.
1996
;
45
(
4
):
463
470
.

221.

Freyse
E-J
,
Fischer
U
,
Knospe
S
,
Ford
GC
,
Nair
KS
.
Differences in protein and energy metabolism following portal versus systemic administration of insulin in diabetic dogs
.
Diabetologia
.
2006
;
49
(
3
):
543
551
.

222.

Pop-Busui
R
,
Kirkwood
I
,
Schmid
H
,
Marinescu
V
,
Schroeder
J
,
Larkin
D
,
Yamada
E
,
Raffel
DM
,
Stevens
MJ
.
Sympathetic dysfunction in type 1 diabetes: association with impaired myocardial blood flow reserve and diastolic dysfunction
.
J Am Coll Cardiol
.
2004
;
44
(
12
):
2368
2374
.

223.

Tai
N
,
Wong
FS
,
Wen
L
.
The role of gut microbiota in the development of type 1, type 2 diabetes mellitus and obesity
.
Rev Endocr Metab Disord
.
2015
;
16
(
1
):
55
65
.

224.

Davidson
LE
,
Kelley
DE
,
Heshka
S
,
Thornton
J
,
Pi-Sunyer
FX
,
Boxt
L
,
Balasubramanyam
A
,
Gallagher
D
;
MRI Ancillary Study Subgroup of the Look AHEAD Research Group
.
Skeletal muscle and organ masses differ in overweight adults with type 2 diabetes
.
J Appl Physiol
.
2014
;
117
(
4
):
377
382
.

225.

Leslie
P
,
Jung
RT
,
Isles
TE
,
Baty
J
,
Newton
RW
,
Illingworth
P
.
Effect of optimal glycaemic control with continuous subcutaneous insulin infusion on energy expenditure in type I diabetes mellitus
.
Br Med J (Clin Res Ed)
.
1986
;
293
(
6555
):
1121
1126
.

226.

Porksen
NK
,
Linnebjerg
H
,
Lam
ECQ
,
Garhyan
P
,
Pachori
A
,
Pratley
RE
,
Smith
SR
.
Basal insulin peglispro increases lipid oxidation, metabolic flexibility, thermogenesis and ketone bodies compared to insulin glargine in subjects with type 1 diabetes mellitus
.
Diabetes Obes Metab
.
2018
;
20
(
5
):
1193
1201
.

227.

Galgani
JE
,
Moro
C
,
Ravussin
E
.
Metabolic flexibility and insulin resistance
.
Am J Physiol Endocrinol Metab
.
2008
;
295
(
5
):
E1009
E1017
.

228.

DeFronzo
RA
,
Tripathy
D
.
Skeletal muscle insulin resistance is the primary defect in type 2 diabetes
.
Diabetes Care
.
2009
;
32
(
Suppl 2
):
S157
S163
.

229.

Blaak
EE
,
Hul
G
,
Verdich
C
,
Stich
V
,
Martinez
A
,
Petersen
M
,
Feskens
EF
,
Patel
K
,
Oppert
JM
,
Barbe
P
,
Toubro
S
,
Anderson
I
,
Polak
J
,
Astrup
A
,
Macdonald
IA
,
Langin
D
,
Holst
C
,
Sørensen
TI
,
Saris
WH
.
Fat oxidation before and after a high fat load in the obese insulin-resistant state
.
J Clin Endocrinol Metab
.
2006
;
91
(
4
):
1462
1469
.

230.

Chomentowski
P
,
Coen
PM
,
Radiková
Z
,
Goodpaster
BH
,
Toledo
FG
.
Skeletal muscle mitochondria in insulin resistance: differences in intermyofibrillar versus subsarcolemmal subpopulations and relationship to metabolic flexibility
.
J Clin Endocrinol Metab
.
2011
;
96
(
2
):
494
503
.

231.

Apostolopoulou
M
,
Strassburger
K
,
Herder
C
,
Knebel
B
,
Kotzka
J
,
Szendroedi
J
,
Roden
M
;
GDS Group
.
Metabolic flexibility and oxidative capacity independently associate with insulin sensitivity in individuals with newly diagnosed type 2 diabetes
.
Diabetologia
.
2016
;
59
(
10
):
2203
2207
.

232.

Kelley
DE
,
Mandarino
LJ
.
Fuel selection in human skeletal muscle in insulin resistance: a reexamination
.
Diabetes
.
2000
;
49
(
5
):
677
683
.

233.

Franko
A
,
Huypens
P
,
Neschen
S
,
Irmler
M
,
Rozman
J
,
Rathkolb
B
,
Neff
F
,
Prehn
C
,
Dubois
G
,
Baumann
M
,
Massinger
R
,
Gradinger
D
,
Przemeck
GK
,
Repp
B
,
Aichler
M
,
Feuchtinger
A
,
Schommers
P
,
Stöhr
O
,
Sanchez-Lasheras
C
,
Adamski
J
,
Peter
A
,
Prokisch
H
,
Beckers
J
,
Walch
AK
,
Fuchs
H
,
Wolf
E
,
Schubert
M
,
Wiesner
RJ
,
Hrabě de Angelis
M
.
Bezafibrate improves insulin sensitivity and metabolic flexibility in STZ-induced diabetic mice
.
Diabetes
.
2016
;
65
(
9
):
2540
2552
.

234.

Kim-Muller
JY
,
Zhao
S
,
Srivastava
S
,
Mugabo
Y
,
Noh
HL
,
Kim
YR
,
Madiraju
SR
,
Ferrante
AW
,
Skolnik
EY
,
Prentki
M
,
Accili
D
.
Metabolic inflexibility impairs insulin secretion and results in MODY-like diabetes in triple FoxO-deficient mice
.
Cell Metab
.
2014
;
20
(
4
):
593
602
.

235.

Accili
D
,
Talchai
SC
,
Kim-Muller
JY
,
Cinti
F
,
Ishida
E
,
Ordelheide
AM
,
Kuo
T
,
Fan
J
,
Son
J
.
When β-cells fail: lessons from dedifferentiation
.
Diabetes Obes Metab
.
2016
;
18
(
Suppl 1
):
117
122
.

236.

Skelton
JA
,
Irby
MB
,
Grzywacz
JG
,
Miller
G
.
Etiologies of obesity in children: nature and nurture
.
Pediatr Clin North Am
.
2011
;
58
(
6
):
1333
1354, ix
.

237.

Chung
WK
,
Leibel
RL
.
Considerations regarding the genetics of obesity
.
Obesity (Silver Spring)
.
2008
;
16
(
Suppl 3
):
S33
S39
.

238.

Naukkarinen
J
,
Rissanen
A
,
Kaprio
J
,
Pietiläinen
KH
.
Causes and consequences of obesity: the contribution of recent twin studies
.
Int J Obes
.
2012
;
36
(
8
):
1017
1024
.

239.

Corella
D
,
Coltell
O
,
Sorlí
JV
,
Estruch
R
,
Quiles
L
,
Martínez-González
MA
,
Salas-Salvadó
J
,
Castañer
O
,
Arós
F
,
Ortega-Calvo
M
,
Serra-Majem
L
,
Gómez-Gracia
E
,
Portolés
O
,
Fiol
M
,
Díez Espino
J
,
Basora
J
,
Fitó
M
,
Ros
E
,
Ordovás
JM
.
Polymorphism of the transcription factor 7-like 2 gene (TCF7L2) interacts with obesity on type-2 diabetes in the PREDIMED study emphasizing the heterogeneity of genetic variants in type-2 diabetes risk prediction: time for obesity-specific genetic risk scores
.
Nutrients
.
2016
;
8
(
12
):
E793
.

240.

Locke
AE
,
Kahali
B
,
Berndt
SI
,
Justice
AE
,
Pers
TH
,
Day
FR
,
Powell
C
,
Vedantam
S
,
Buchkovich
ML
,
Yang
J
,
Croteau-Chonka
DC
,
Esko
T
,
Fall
T
,
Ferreira
T
,
Gustafsson
S
,
Kutalik
Z
,
Luan
J
,
Mägi
R
,
Randall
JC
,
Winkler
TW
,
Wood
AR
,
Workalemahu
T
,
Faul
JD
,
Smith
JA
,
Zhao
JH
,
Zhao
W
,
Chen
J
,
Fehrmann
R
,
Hedman
ÅK
,
Karjalainen
J
,
Schmidt
EM
,
Absher
D
,
Amin
N
,
Anderson
D
,
Beekman
M
,
Bolton
JL
,
Bragg-Gresham
JL
,
Buyske
S
,
Demirkan
A
,
Deng
G
,
Ehret
GB
,
Feenstra
B
,
Feitosa
MF
,
Fischer
K
,
Goel
A
,
Gong
J
,
Jackson
AU
,
Kanoni
S
,
Kleber
ME
,
Kristiansson
K
,
Lim
U
,
Lotay
V
,
Mangino
M
,
Leach
IM
,
Medina-Gomez
C
,
Medland
SE
,
Nalls
MA
,
Palmer
CD
,
Pasko
D
,
Pechlivanis
S
,
Peters
MJ
,
Prokopenko
I
,
Shungin
D
,
Stančáková
A
,
Strawbridge
RJ
,
Sung
YJ
,
Tanaka
T
,
Teumer
A
,
Trompet
S
,
van der Laan
SW
,
van Setten
J
,
Van Vliet-Ostaptchouk
JV
,
Wang
Z
,
Yengo
L
,
Zhang
W
,
Isaacs
A
,
Albrecht
E
,
Ärnlöv
J
,
Arscott
GM
,
Attwood
AP
,
Bandinelli
S
,
Barrett
A
,
Bas
IN
,
Bellis
C
,
Bennett
AJ
,
Berne
C
,
Blagieva
R
,
Blüher
M
,
Böhringer
S
,
Bonnycastle
LL
,
Böttcher
Y
,
Boyd
HA
,
Bruinenberg
M
,
Caspersen
IH
,
Chen
YI
,
Clarke
R
,
Daw
EW
,
de Craen
AJM
,
Delgado
G
,
Dimitriou
M
,
Doney
ASF
,
Eklund
N
,
Estrada
K
,
Eury
E
,
Folkersen
L
,
Fraser
RM
,
Garcia
ME
,
Geller
F
,
Giedraitis
V
,
Gigante
B
,
Go
AS
,
Golay
A
,
Goodall
AH
,
Gordon
SD
,
Gorski
M
,
Grabe
HJ
,
Grallert
H
,
Grammer
TB
,
Gräßler
J
,
Grönberg
H
,
Groves
CJ
,
Gusto
G
,
Haessler
J
,
Hall
P
,
Haller
T
,
Hallmans
G
,
Hartman
CA
,
Hassinen
M
,
Hayward
C
,
Heard-Costa
NL
,
Helmer
Q
,
Hengstenberg
C
,
Holmen
O
,
Hottenga
JJ
,
James
AL
,
Jeff
JM
,
Johansson
Å
,
Jolley
J
,
Juliusdottir
T
,
Kinnunen
L
,
Koenig
W
,
Koskenvuo
M
,
Kratzer
W
,
Laitinen
J
,
Lamina
C
,
Leander
K
,
Lee
NR
,
Lichtner
P
,
Lind
L
,
Lindström
J
,
Lo
KS
,
Lobbens
S
,
Lorbeer
R
,
Lu
Y
,
Mach
F
,
Magnusson
PKE
,
Mahajan
A
,
McArdle
WL
,
McLachlan
S
,
Menni
C
,
Merger
S
,
Mihailov
E
,
Milani
L
,
Moayyeri
A
,
Monda
KL
,
Morken
MA
,
Mulas
A
,
Müller
G
,
Müller-Nurasyid
M
,
Musk
AW
,
Nagaraja
R
,
Nöthen
MM
,
Nolte
IM
,
Pilz
S
,
Rayner
NW
,
Renstrom
F
,
Rettig
R
,
Ried
JS
,
Ripke
S
,
Robertson
NR
,
Rose
LM
,
Sanna
S
,
Scharnagl
H
,
Scholtens
S
,
Schumacher
FR
,
Scott
WR
,
Seufferlein
T
,
Shi
J
,
Smith
AV
,
Smolonska
J
,
Stanton
AV
,
Steinthorsdottir
V
,
Stirrups
K
,
Stringham
HM
,
Sundström
J
,
Swertz
MA
,
Swift
AJ
,
Syvänen
AC
,
Tan
ST
,
Tayo
BO
,
Thorand
B
,
Thorleifsson
G
,
Tyrer
JP
,
Uh
HW
,
Vandenput
L
,
Verhulst
FC
,
Vermeulen
SH
,
Verweij
N
,
Vonk
JM
,
Waite
LL
,
Warren
HR
,
Waterworth
D
,
Weedon
MN
,
Wilkens
LR
,
Willenborg
C
,
Wilsgaard
T
,
Wojczynski
MK
,
Wong
A
,
Wright
AF
,
Zhang
Q
,
Brennan
EP
,
Choi
M
,
Dastani
Z
,
Drong
AW
,
Eriksson
P
,
Franco-Cereceda
A
,
Gådin
JR
,
Gharavi
AG
,
Goddard
ME
,
Handsaker
RE
,
Huang
J
,
Karpe
F
,
Kathiresan
S
,
Keildson
S
,
Kiryluk
K
,
Kubo
M
,
Lee
JY
,
Liang
L
,
Lifton
RP
,
Ma
B
,
McCarroll
SA
,
McKnight
AJ
,
Min
JL
,
Moffatt
MF
,
Montgomery
GW
,
Murabito
JM
,
Nicholson
G
,
Nyholt
DR
,
Okada
Y
,
Perry
JRB
,
Dorajoo
R
,
Reinmaa
E
,
Salem
RM
,
Sandholm
N
,
Scott
RA
,
Stolk
L
,
Takahashi
A
,
Tanaka
T
,
van ’t Hooft
FM
,
Vinkhuyzen
AAE
,
Westra
HJ
,
Zheng
W
,
Zondervan
KT
,
Heath
AC
,
Arveiler
D
,
Bakker
SJL
,
Beilby
J
,
Bergman
RN
,
Blangero
J
,
Bovet
P
,
Campbell
H
,
Caulfield
MJ
,
Cesana
G
,
Chakravarti
A
,
Chasman
DI
,
Chines
PS
,
Collins
FS
,
Crawford
DC
,
Cupples
LA
,
Cusi
D
,
Danesh
J
,
de Faire
U
,
den Ruijter
HM
,
Dominiczak
AF
,
Erbel
R
,
Erdmann
J
,
Eriksson
JG
,
Farrall
M
,
Felix
SB
,
Ferrannini
E
,
Ferrières
J
,
Ford
I
,
Forouhi
NG
,
Forrester
T
,
Franco
OH
,
Gansevoort
RT
,
Gejman
PV
,
Gieger
C
,
Gottesman
O
,
Gudnason
V
,
Gyllensten
U
,
Hall
AS
,
Harris
TB
,
Hattersley
AT
,
Hicks
AA
,
Hindorff
LA
,
Hingorani
AD
,
Hofman
A
,
Homuth
G
,
Hovingh
GK
,
Humphries
SE
,
Hunt
SC
,
Hyppönen
E
,
Illig
T
,
Jacobs
KB
,
Jarvelin
MR
,
Jöckel
KH
,
Johansen
B
,
Jousilahti
P
,
Jukema
JW
,
Jula
AM
,
Kaprio
J
,
Kastelein
JJP
,
Keinanen-Kiukaanniemi
SM
,
Kiemeney
LA
,
Knekt
P
,
Kooner
JS
,
Kooperberg
C
,
Kovacs
P
,
Kraja
AT
,
Kumari
M
,
Kuusisto
J
,
Lakka
TA
,
Langenberg
C
,
Marchand
LL
,
Lehtimäki
T
,
Lyssenko
V
,
Männistö
S
,
Marette
A
,
Matise
TC
,
McKenzie
CA
,
McKnight
B
,
Moll
FL
,
Morris
AD
,
Morris
AP
,
Murray
JC
,
Nelis
M
,
Ohlsson
C
,
Oldehinkel
AJ
,
Ong
KK
,
Madden
PAF
,
Pasterkamp
G
,
Peden
JF
,
Peters
A
,
Postma
DS
,
Pramstaller
PP
,
Price
JF
,
Qi
L
,
Raitakari
OT
,
Rankinen
T
,
Rao
DC
,
Rice
TK
,
Ridker
PM
,
Rioux
JD
,
Ritchie
MD
,
Rudan
I
,
Salomaa
V
,
Samani
NJ
,
Saramies
J
,
Sarzynski
MA
,
Schunkert
H
,
Schwarz
PEH
,
Sever
P
,
Shuldiner
AR
,
Sinisalo
J
,
Stolk
RP
,
Strauch
K
,
Tönjes
A
,
Trégouët
DA
,
Tremblay
A
,
Tremoli
E
,
Virtamo
J
,
Vohl
MC
,
Völker
U
,
Waeber
G
,
Willemsen
G
,
Witteman
JC
,
Zillikens
MC
,
Adair
LS
,
Amouyel
P
,
Asselbergs
FW
,
Assimes
TL
,
Bochud
M
,
Boehm
BO
,
Boerwinkle
E
,
Bornstein
SR
,
Bottinger
EP
,
Bouchard
C
,
Cauchi
S
,
Chambers
JC
,
Chanock
SJ
,
Cooper
RS
,
de Bakker
PIW
,
Dedoussis
G
,
Ferrucci
L
,
Franks
PW
,
Froguel
P
,
Groop
LC
,
Haiman
CA
,
Hamsten
A
,
Hui
J
,
Hunter
DJ
,
Hveem
K
,
Kaplan
RC
,
Kivimaki
M
,
Kuh
D
,
Laakso
M
,
Liu
Y
,
Martin
NG
,
März
W
,
Melbye
M
,
Metspalu
A
,
Moebus
S
,
Munroe
PB
,
Njølstad
I
,
Oostra
BA
,
Palmer
CNA
,
Pedersen
NL
,
Perola
M
,
Pérusse
L
,
Peters
U
,
Power
C
,
Quertermous
T
,
Rauramaa
R
,
Rivadeneira
F
,
Saaristo
TE
,
Saleheen
D
,
Sattar
N
,
Schadt
EE
,
Schlessinger
D
,
Slagboom
PE
,
Snieder
H
,
Spector
TD
,
Thorsteinsdottir
U
,
Stumvoll
M
,
Tuomilehto
J
,
Uitterlinden
AG
,
Uusitupa
M
,
van der Harst
P
,
Walker
M
,
Wallaschofski
H
,
Wareham
NJ
,
Watkins
H
,
Weir
DR
,
Wichmann
HE
,
Wilson
JF
,
Zanen
P
,
Borecki
IB
,
Deloukas
P
,
Fox
CS
,
Heid
IM
,
O’Connell
JR
,
Strachan
DP
,
Stefansson
K
,
van Duijn
CM
,
Abecasis
GR
,
Franke
L
,
Frayling
TM
,
McCarthy
MI
,
Visscher
PM
,
Scherag
A
,
Willer
CJ
,
Boehnke
M
,
Mohlke
KL
,
Lindgren
CM
,
Beckmann
JS
,
Barroso
I
,
North
KE
,
Ingelsson
E
,
Hirschhorn
JN
,
Loos
RJF
,
Speliotes
EK
;
LifeLines Cohort Study; ADIPOGen Consortium; AGEN-BMI Working Group; CARDIOGRAMplusC4D Consortium; CKDGen Consortium; GLGC; ICBP; MAGIC Investigators; MuTHER Consortium; MIGen Consortium; PAGE Consortium; ReproGen Consortium; GENIE Consortium; International Endogene Consortium
.
Genetic studies of body mass index yield new insights for obesity biology
.
Nature
.
2015
;
518
(
7538
):
197
206
.

241.

Mahajan
A
,
Go
MJ
,
Zhang
W
,
Below
JE
,
Gaulton
KJ
,
Ferreira
T
,
Horikoshi
M
,
Johnson
AD
,
Ng
MC
,
Prokopenko
I
,
Saleheen
D
,
Wang
X
,
Zeggini
E
,
Abecasis
GR
,
Adair
LS
,
Almgren
P
,
Atalay
M
,
Aung
T
,
Baldassarre
D
,
Balkau
B
,
Bao
Y
,
Barnett
AH
,
Barroso
I
,
Basit
A
,
Been
LF
,
Beilby
J
,
Bell
GI
,
Benediktsson
R
,
Bergman
RN
,
Boehm
BO
,
Boerwinkle
E
,
Bonnycastle
LL
,
Burtt
N
,
Cai
Q
,
Campbell
H
,
Carey
J
,
Cauchi
S
,
Caulfield
M
,
Chan
JC
,
Chang
LC
,
Chang
TJ
,
Chang
YC
,
Charpentier
G
,
Chen
CH
,
Chen
H
,
Chen
YT
,
Chia
KS
,
Chidambaram
M
,
Chines
PS
,
Cho
NH
,
Cho
YM
,
Chuang
LM
,
Collins
FS
,
Cornelis
MC
,
Couper
DJ
,
Crenshaw
AT
,
van Dam
RM
,
Danesh
J
,
Das
D
,
de Faire
U
,
Dedoussis
G
,
Deloukas
P
,
Dimas
AS
,
Dina
C
,
Doney
AS
,
Donnelly
PJ
,
Dorkhan
M
,
van Duijn
C
,
Dupuis
J
,
Edkins
S
,
Elliott
P
,
Emilsson
V
,
Erbel
R
,
Eriksson
JG
,
Escobedo
J
,
Esko
T
,
Eury
E
,
Florez
JC
,
Fontanillas
P
,
Forouhi
NG
,
Forsen
T
,
Fox
C
,
Fraser
RM
,
Frayling
TM
,
Froguel
P
,
Frossard
P
,
Gao
Y
,
Gertow
K
,
Gieger
C
,
Gigante
B
,
Grallert
H
,
Grant
GB
,
Grrop
LC
,
Groves
CJ
,
Grundberg
E
,
Guiducci
C
,
Hamsten
A
,
Han
BG
,
Hara
K
,
Hassanali
N
,
Hattersley
AT
,
Hayward
C
,
Hedman
AK
,
Herder
C
,
Hofman
A
,
Holmen
OL
,
Hovingh
K
,
Hreidarsson
AB
,
Hu
C
,
Hu
FB
,
Hui
J
,
Humphries
SE
,
Hunt
SE
,
Hunter
DJ
,
Hveem
K
,
Hydrie
ZI
,
Ikegami
H
,
Illig
T
,
Ingelsson
E
,
Islam
M
,
Isomaa
B
,
Jackson
AU
,
Jafar
T
,
James
A
,
Jia
W
,
Jöckel
KH
,
Jonsson
A
,
Jowett
JB
,
Kadowaki
T
,
Kang
HM
,
Kanoni
S
,
Kao
WH
,
Kathiresan
S
,
Kato
N
,
Katulanda
P
,
Keinanen-Kiukaanniemi
KM
,
Kelly
AM
,
Khan
H
,
Khaw
KT
,
Khor
CC
,
Kim
HL
,
Kim
S
,
Kim
YJ
,
Kinnunen
L
,
Klopp
N
,
Kong
A
,
Korpi-Hyövälti
E
,
Kowlessur
S
,
Kraft
P
,
Kravic
J
,
Kristensen
MM
,
Krithika
S
,
Kumar
A
,
Kumate
J
,
Kuusisto
J
,
Kwak
SH
,
Laakso
M
,
Lagou
V
,
Lakka
TA
,
Langenberg
C
,
Langford
C
,
Lawrence
R
,
Leander
K
,
Lee
JM
,
Lee
NR
,
Li
M
,
Li
X
,
Li
Y
,
Liang
J
,
Liju
S
,
Lim
WY
,
Lind
L
,
Lindgren
CM
,
Lindholm
E
,
Liu
CT
,
Liu
JJ
,
Lobbens
S
,
Long
J
,
Loos
RJ
,
Lu
W
,
Luan
J
,
Lyssenko
V
,
Ma
RC
,
Maeda
S
,
Mägi
R
,
Männisto
S
,
Matthews
DR
,
Meigs
JB
,
Melander
O
,
Metspalu
A
,
Meyer
J
,
Mirza
G
,
Mihailov
E
,
Moebus
S
,
Mohan
V
,
Mohlke
KL
,
Morris
AD
,
Mühleisen
TW
,
Müller-Nurasyid
M
,
Musk
B
,
Nakamura
J
,
Nakashima
E
,
Navarro
P
,
Ng
PK
,
Nica
AC
,
Nilsson
PM
,
Njølstad
I
,
Nöthen
MM
,
Ohnaka
K
,
Ong
TH
,
Owen
KR
,
Palmer
CN
,
Pankow
JS
,
Park
KS
,
Parkin
M
,
Pechlivanis
S
,
Pedersen
NL
,
Peltonen
L
,
Perry
JR
,
Peters
A
,
Pinidiyapathirage
JM
,
Platou
CG
,
Potter
S
,
Price
JF
,
Qi
L
,
Radha
V
,
Rallidis
L
,
Rasheed
A
,
Rathman
W
,
Rauramaa
R
,
Raychaudhuri
S
,
Rayner
NW
,
Rees
SD
,
Rehnberg
E
,
Ripatti
S
,
Robertson
N
,
Roden
M
,
Rossin
EJ
,
Rudan
I
,
Rybin
D
,
Saaristo
TE
,
Salomaa
V
,
Saltevo
J
,
Samuel
M
,
Sanghera
DK
,
Saramies
J
,
Scott
J
,
Scott
LJ
,
Scott
RA
,
Segrè
AV
,
Sehmi
J
,
Sennblad
B
,
Shah
N
,
Shah
S
,
Shera
AS
,
Shu
XO
,
Shuldiner
AR
,
Sigurđsson
G
,
Sijbrands
E
,
Silveira
A
,
Sim
X
,
Sivapalaratnam
S
,
Small
KS
,
So
WY
,
Stančáková
A
,
Stefansson
K
,
Steinbach
G
,
Steinthorsdottir
V
,
Stirrups
K
,
Strawbridge
RJ
,
Stringham
HM
,
Sun
Q
,
Suo
C
,
Syvänen
AC
,
Takayanagi
R
,
Takeuchi
F
,
Tay
WT
,
Teslovich
TM
,
Thorand
B
,
Thorleifsson
G
,
Thorsteinsdottir
U
,
Tikkanen
E
,
Trakalo
J
,
Tremoli
E
,
Trip
MD
,
Tsai
FJ
,
Tuomi
T
,
Tuomilehto
J
,
Uitterlinden
AG
,
Valladares-Salgado
A
,
Vedantam
S
,
Veglia
F
,
Voight
BF
,
Wang
C
,
Wareham
NJ
,
Wennauer
R
,
Wickremasinghe
AR
,
Wilsgaard
T
,
Wilson
JF
,
Wiltshire
S
,
Winckler
W
,
Wong
TY
,
Wood
AR
,
Wu
JY
,
Wu
Y
,
Yamamoto
K
,
Yamauchi
T
,
Yang
M
,
Yengo
L
,
Yokota
M
,
Young
R
,
Zabaneh
D
,
Zhang
F
,
Zhang
R
,
Zheng
W
,
Zimmet
PZ
,
Altshuler
D
,
Bowden
DW
,
Cho
YS
,
Cox
NJ
,
Cruz
M
,
Hanis
CL
,
Kooner
J
,
Lee
JY
,
Seielstad
M
,
Teo
YY
,
Boehnke
M
,
Parra
EJ
,
Chambers
JC
,
Tai
ES
,
McCarthy
MI
,
Morris
AP
;
DIAbetes Genetics Replication and Meta-analysis (DIAGRAM) Consortium; Asian Genetic Epidemiology Network Type 2 Diabetes (AGEN-T2D) Consortium; South Asian Type 2 Diabetes (SAT2D) Consortium; Mexican American Type 2 Diabetes (MAT2D) Consortium; Type 2 Diabetes Genetic Exploration by Next-Gneration sequencing in Muyti-Ethnic Samples (T2D-GENES) Consortium
.
Genome-wide trans-ancestry meta-analysis provides insight into the genetic architecture of type 2 diabetes susceptibility
.
Nat Genet
.
2014
;
46
(
3
):
234
244
.

242.

Hertel
JK
,
Johansson
S
,
Sonestedt
E
,
Jonsson
A
,
Lie
RT
,
Platou
CG
,
Nilsson
PM
,
Rukh
G
,
Midthjell
K
,
Hveem
K
,
Melander
O
,
Groop
L
,
Lyssenko
V
,
Molven
A
,
Orho-Melander
M
,
Njølstad
PR
.
FTO, type 2 diabetes, and weight gain throughout adult life: a meta-analysis of 41,504 subjects from the Scandinavian HUNT, MDC, and MPP studies
.
Diabetes
.
2011
;
60
(
5
):
1637
1644
.

243.

Li
H
,
Kilpeläinen
TO
,
Liu
C
,
Zhu
J
,
Liu
Y
,
Hu
C
,
Yang
Z
,
Zhang
W
,
Bao
W
,
Cha
S
,
Wu
Y
,
Yang
T
,
Sekine
A
,
Choi
BY
,
Yajnik
CS
,
Zhou
D
,
Takeuchi
F
,
Yamamoto
K
,
Chan
JC
,
Mani
KR
,
Been
LF
,
Imamura
M
,
Nakashima
E
,
Lee
N
,
Fujisawa
T
,
Karasawa
S
,
Wen
W
,
Joglekar
CV
,
Lu
W
,
Chang
Y
,
Xiang
Y
,
Gao
Y
,
Liu
S
,
Song
Y
,
Kwak
SH
,
Shin
HD
,
Park
KS
,
Fall
CH
,
Kim
JY
,
Sham
PC
,
Lam
KS
,
Zheng
W
,
Shu
X
,
Deng
H
,
Ikegami
H
,
Krishnaveni
GV
,
Sanghera
DK
,
Chuang
L
,
Liu
L
,
Hu
R
,
Kim
Y
,
Daimon
M
,
Hotta
K
,
Jia
W
,
Kooner
JS
,
Chambers
JC
,
Chandak
GR
,
Ma
RC
,
Maeda
S
,
Dorajoo
R
,
Yokota
M
,
Takayanagi
R
,
Kato
N
,
Lin
X
,
Loos
RJ
.
Association of genetic variation in FTO with risk of obesity and type 2 diabetes with data from 96,551 East and South Asians
.
Diabetologia
.
2012
;
55
(
4
):
981
995
.

244.

Grarup
N
,
Sandholt
CH
,
Hansen
T
,
Pedersen
O
.
Genetic susceptibility to type 2 diabetes and obesity: from genome-wide association studies to rare variants and beyond
.
Diabetologia
.
2014
;
57
(
8
):
1528
1541
.

245.

Tung
YCL
,
Yeo
GSH
,
O’Rahilly
S
,
Coll
AP
.
Obesity and FTO: changing focus at a complex locus
.
Cell Metab
.
2014
;
20
(
5
):
710
718
.

246.

Pociot
F
,
Lernmark
Å
.
Genetic risk factors for type 1 diabetes
.
Lancet
.
2016
;
387
(
10035
):
2331
2339
.

247.

Spiegelman
BM
,
Flier
JS
.
Obesity and the regulation of energy balance
.
Cell
.
2001
;
104
(
4
):
531
543
.

248.

Frayling
TM
,
Timpson
NJ
,
Weedon
MN
,
Zeggini
E
,
Freathy
RM
,
Lindgren
CM
,
Perry
JR
,
Elliott
KS
,
Lango
H
,
Rayner
NW
,
Shields
B
,
Harries
LW
,
Barrett
JC
,
Ellard
S
,
Groves
CJ
,
Knight
B
,
Patch
AM
,
Ness
AR
,
Ebrahim
S
,
Lawlor
DA
,
Ring
SM
,
Ben-Shlomo
Y
,
Jarvelin
MR
,
Sovio
U
,
Bennett
AJ
,
Melzer
D
,
Ferrucci
L
,
Loos
RJ
,
Barroso
I
,
Wareham
NJ
,
Karpe
F
,
Owen
KR
,
Cardon
LR
,
Walker
M
,
Hitman
GA
,
Palmer
CN
,
Doney
AS
,
Morris
AD
,
Smith
GD
,
Hattersley
AT
,
McCarthy
MI
.
A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity
.
Science
.
2007
;
316
(
5826
):
889
894
.

249.

Haupt
A
,
Thamer
C
,
Machann
J
,
Kirchhoff
K
,
Stefan
N
,
Tschritter
O
,
Machicao
F
,
Schick
F
,
Häring
HU
,
Fritsche
A
.
Impact of variation in the FTO gene on whole body fat distribution, ectopic fat, and weight loss
.
Obesity (Silver Spring)
.
2008
;
16
(
8
):
1969
1972
.

250.

Loos
RJ
,
Lindgren
CM
,
Li
S
,
Wheeler
E
,
Zhao
JH
,
Prokopenko
I
,
Inouye
M
,
Freathy
RM
,
Attwood
AP
,
Beckmann
JS
,
Berndt
SI
,
Jacobs
KB
,
Chanock
SJ
,
Hayes
RB
,
Bergmann
S
,
Bennett
AJ
,
Bingham
SA
,
Bochud
M
,
Brown
M
,
Cauchi
S
,
Connell
JM
,
Cooper
C
,
Smith
GD
,
Day
I
,
Dina
C
,
De
S
,
Dermitzakis
ET
,
Doney
AS
,
Elliott
KS
,
Elliott
P
,
Evans
DM
,
Sadaf Farooqi
I
,
Froguel
P
,
Ghori
J
,
Groves
CJ
,
Gwilliam
R
,
Hadley
D
,
Hall
AS
,
Hattersley
AT
,
Hebebrand
J
,
Heid
IM
,
Lamina
C
,
Gieger
C
,
Illig
T
,
Meitinger
T
,
Wichmann
HE
,
Herrera
B
,
Hinney
A
,
Hunt
SE
,
Jarvelin
MR
,
Johnson
T
,
Jolley
JD
,
Karpe
F
,
Keniry
A
,
Khaw
KT
,
Luben
RN
,
Mangino
M
,
Marchini
J
,
McArdle
WL
,
McGinnis
R
,
Meyre
D
,
Munroe
PB
,
Morris
AD
,
Ness
AR
,
Neville
MJ
,
Nica
AC
,
Ong
KK
,
O’Rahilly
S
,
Owen
KR
,
Palmer
CN
,
Papadakis
K
,
Potter
S
,
Pouta
A
,
Qi
L
,
Randall
JC
,
Rayner
NW
,
Ring
SM
,
Sandhu
MS
,
Scherag
A
,
Sims
MA
,
Song
K
,
Soranzo
N
,
Speliotes
EK
,
Syddall
HE
,
Teichmann
SA
,
Timpson
NJ
,
Tobias
JH
,
Uda
M
,
Vogel
CI
,
Wallace
C
,
Waterworth
DM
,
Weedon
MN
,
Willer
CJ
,
Wraight
,
Yuan
X
,
Zeggini
E
,
Hirschhorn
JN
,
Strachan
DP
,
Ouwehand
WH
,
Caulfield
MJ
,
Samani
NJ
,
Frayling
TM
,
Vollenweider
P
,
Waeber
G
,
Mooser
V
,
Deloukas
P
,
McCarthy
MI
,
Wareham
NJ
,
Barroso
I
,
Jacobs
KB
,
Chanock
SJ
,
Hayes
RB
,
Lamina
C
,
Gieger
C
,
Illig
T
,
Meitinger
T
,
Wichmann
HE
,
Kraft
P
,
Hankinson
SE
,
Hunter
DJ
,
Hu
FB
,
Lyon
HN
,
Voight
BF
,
Ridderstrale
M
,
Groop
L
,
Scheet
P
,
Sanna
S
,
Abecasis
GR
,
Albai
G
,
Nagaraja
R
,
Schlessinger
D
,
Jackson
AU
,
Tuomilehto
J
,
Collins
FS
,
Boehnke
M
,
Mohlke
KL
;
Prostate, Lung,Colorectal, and Ovarian (PLCO) Cancer Screening Trial; KORA; Nurses’ Health Study; Diabetes Genetics Initiative; SardiNIA Study; Wellcome Trust Case Control Consortium; FUSION
.
Common variants near MC4R are associated with fat mass, weight and risk of obesity
.
Nat Genet
.
2008
;
40
(
6
):
768
775
.

251.

Chambers
JC
,
Elliott
P
,
Zabaneh
D
,
Zhang
W
,
Li
Y
,
Froguel
P
,
Balding
D
,
Scott
J
,
Kooner
JS
.
Common genetic variation near MC4R is associated with waist circumference and insulin resistance
.
Nat Genet
.
2008
;
40
(
6
):
716
718
.

252.

Challis
BG
,
Pritchard
LE
,
Creemers
JW
,
Delplanque
J
,
Keogh
JM
,
Luan
J
,
Wareham
NJ
,
Yeo
GS
,
Bhattacharyya
S
,
Froguel
P
,
White
A
,
Farooqi
IS
,
O’Rahilly
S
.
A missense mutation disrupting a dibasic prohormone processing site in pro-opiomelanocortin (POMC) increases susceptibility to early-onset obesity through a novel molecular mechanism
.
Hum Mol Genet
.
2002
;
11
(
17
):
1997
2004
.

253.

Łuczyński
W
,
Fendler
W
,
Ramatowska
A
,
Szypowska
A
,
Szadkowska
A
,
Młynarski
W
,
Chumiecki
M
,
Jarosz-Chobot
P
,
Chrzanowska
J
,
Noczyńska
A
,
Brandt
A
,
Myśliwiec
M
,
Głowińska-Olszewska
B
,
Bernatowicz
P
,
Kowalczuk
O
,
Bossowski
A
.
Polymorphism of the FTO gene influences body weight in children with type 1 diabetes without severe obesity
.
Int J Endocrinol
.
2014
;
2014
:
630712
.

254.

Qu
H-Q
,
Polychronakos
C
.
The TCF7L2 locus and type 1 diabetes
.
BMC Med Genet
.
2007
;
8
(
1
):
51
.

255.

Redondo
MJ
,
Geyer
S
,
Steck
AK
,
Sosenko
J
,
Anderson
M
,
Antinozzi
P
,
Michels
A
,
Wentworth
J
,
Xu
P
,
Pugliese
A
;
Type 1 Diabetes TrialNet Study Group
.
TCF7L2 genetic variants contribute to phenotypic heterogeneity of type 1 diabetes
.
Diabetes Care
.
2018
;
41
(
2
):
311
317
.

256.

Censin
JC
,
Nowak
C
,
Cooper
N
,
Bergsten
P
,
Todd
JA
,
Fall
T
.
Childhood adiposity and risk of type 1 diabetes: a Mendelian randomization study
.
PLoS Med
.
2017
;
14
(
8
):
e1002362
.

257.

Todd
JN
,
Dahlström
EH
,
Salem
RM
,
Sandholm
N
,
Forsblom
C
,
McKnight
AJ
,
Maxwell
AP
,
Brennan
E
,
Sadlier
D
,
Godson
C
,
Groop
PH
,
Hirschhorn
JN
,
Florez
JC
;
FinnDiane Study Group
.
Genetic evidence for a causal role of obesity in diabetic kidney disease
.
Diabetes
.
2015
;
64
(
12
):
4238
4246
.

258.

Donath
MY
,
Dalmas
É
,
Sauter
NS
,
Böni-Schnetzler
M
.
Inflammation in obesity and diabetes: islet dysfunction and therapeutic opportunity
.
Cell Metab
.
2013
;
17
(
6
):
860
872
.

259.

Hu
X
,
Deutsch
AJ
,
Lenz
TL
,
Onengut-Gumuscu
S
,
Han
B
,
Chen
WM
,
Howson
JM
,
Todd
JA
,
de Bakker
PI
,
Rich
SS
,
Raychaudhuri
S
.
Additive and interaction effects at three amino acid positions in HLA-DQ and HLA-DR molecules drive type 1 diabetes risk
.
Nat Genet
.
2015
;
47
(
8
):
898
905
.

260.

Fukaya
M
,
Brorsson
CA
,
Meyerovich
K
,
Catrysse
L
,
Delaroche
D
,
Vanzela
EC
,
Ortis
F
,
Beyaert
R
,
Nielsen
LB
,
Andersen
ML
,
Mortensen
HB
,
Pociot
F
,
van Loo
G
,
Størling
J
,
Cardozo
AK
.
A20 inhibits β-cell apoptosis by multiple mechanisms and predicts residual β-cell function in type 1 diabetes
.
Mol Endocrinol
.
2016
;
30
(
1
):
48
61
.

261.

Marroqui
L
,
Dos Santos
RS
,
Fløyel
T
,
Grieco
FA
,
Santin
I
,
Op de Beeck
A
,
Marselli
L
,
Marchetti
P
,
Pociot
F
,
Eizirik
DL
.
TYK2, a candidate gene for type 1 diabetes, modulates apoptosis and the innate immune response in human pancreatic β-cells
.
Diabetes
.
2015
;
64
(
11
):
3808
3817
.

262.

Wahl
S
,
Drong
A
,
Lehne
B
,
Loh
M
,
Scott
WR
,
Kunze
S
,
Tsai
PC
,
Ried
JS
,
Zhang
W
,
Yang
Y
,
Tan
S
,
Fiorito
G
,
Franke
L
,
Guarrera
S
,
Kasela
S
,
Kriebel
J
,
Richmond
RC
,
Adamo
M
,
Afzal
U
,
Ala-Korpela
M
,
Albetti
B
,
Ammerpohl
O
,
Apperley
JF
,
Beekman
M
,
Bertazzi
PA
,
Black
SL
,
Blancher
C
,
Bonder
MJ
,
Brosch
M
,
Carstensen-Kirberg
M
,
de Craen
AJ
,
de Lusignan
S
,
Dehghan
A
,
Elkalaawy
M
,
Fischer
K
,
Franco
OH
,
Gaunt
TR
,
Hampe
J
,
Hashemi
M
,
Isaacs
A
,
Jenkinson
A
,
Jha
S
,
Kato
N
,
Krogh
V
,
Laffan
M
,
Meisinger
C
,
Meitinger
T
,
Mok
ZY
,
Motta
V
,
Ng
HK
,
Nikolakopoulou
Z
,
Nteliopoulos
G
,
Panico
S
,
Pervjakova
N
,
Prokisch
H
,
Rathmann
W
,
Roden
M
,
Rota
F
,
Rozario
MA
,
Sandling
JK
,
Schafmayer
C
,
Schramm
K
,
Siebert
R
,
Slagboom
PE
,
Soininen
P
,
Stolk
L
,
Strauch
K
,
Tai
ES
,
Tarantini
L
,
Thorand
B
,
Tigchelaar
EF
,
Tumino
R
,
Uitterlinden
AG
,
van Duijn
C
,
van Meurs
JB
,
Vineis
P
,
Wickremasinghe
AR
,
Wijmenga
C
,
Yang
TP
,
Yuan
W
,
Zhernakova
A
,
Batterham
RL
,
Smith
GD
,
Deloukas
P
,
Heijmans
BT
,
Herder
C
,
Hofman
A
,
Lindgren
CM
,
Milani
L
,
van der Harst
P
,
Peters
A
,
Illig
T
,
Relton
CL
,
Waldenberger
M
,
Järvelin
MR
,
Bollati
V
,
Soong
R
,
Spector
TD
,
Scott
J
,
McCarthy
MI
,
Elliott
P
,
Bell
JT
,
Matullo
G
,
Gieger
C
,
Kooner
JS
,
Grallert
H
,
Chambers
JC
.
Epigenome-wide association study of body mass index, and the adverse outcomes of adiposity
.
Nature
.
2017
;
541
(
7635
):
81
86
.

263.

Jerram
ST
,
Dang
MN
,
Leslie
RD
.
The role of epigenetics in type 1 diabetes
.
Curr Diab Rep
.
2017
;
17
(
10
):
89
.

264.

Dellschaft
NS
,
Alexandre-Gouabau
MC
,
Gardner
DS
,
Antignac
JP
,
Keisler
DH
,
Budge
H
,
Symonds
ME
,
Sebert
SP
.
Effect of pre- and postnatal growth and post-weaning activity on glucose metabolism in the offspring
.
J Endocrinol
.
2015
;
224
(
2
):
171
182
.

265.

Férézou-Viala
J
,
Roy
AF
,
Sérougne
C
,
Gripois
D
,
Parquet
M
,
Bailleux
V
,
Gertler
A
,
Delplanque
B
,
Djiane
J
,
Riottot
M
,
Taouis
M
.
Long-term consequences of maternal high-fat feeding on hypothalamic leptin sensitivity and diet-induced obesity in the offspring
.
Am J Physiol Regul Integr Comp Physiol
.
2007
;
293
(
3
):
R1056
R1062
.

266.

Gautier
J-F
,
Wilson
C
,
Weyer
C
,
Mott
D
,
Knowler
WC
,
Cavaghan
M
,
Polonsky
KS
,
Bogardus
C
,
Pratley
RE
.
Low acute insulin secretory responses in adult offspring of people with early onset type 2 diabetes
.
Diabetes
.
2001
;
50
(
8
):
1828
1833
.

267.

Delahaye
F
,
Breton
C
,
Risold
PY
,
Enache
M
,
Dutriez-Casteloot
I
,
Laborie
C
,
Lesage
J
,
Vieau
D
.
Maternal perinatal undernutrition drastically reduces postnatal leptin surge and affects the development of arcuate nucleus proopiomelanocortin neurons in neonatal male rat pups
.
Endocrinology
.
2008
;
149
(
2
):
470
475
.

268.

Chang
GQ
,
Gaysinskaya
V
,
Karatayev
O
,
Leibowitz
SF
.
Maternal high-fat diet and fetal programming: increased proliferation of hypothalamic peptide-producing neurons that increase risk for overeating and obesity
.
J Neurosci
.
2008
;
28
(
46
):
12107
12119
.

269.

Parsons
TJ
,
Power
C
,
Manor
O
.
Fetal and early life growth and body mass index from birth to early adulthood in 1958 British cohort: longitudinal study
.
BMJ
.
2001
;
323
(
7325
):
1331
1335
.

270.

Waterland
RA
,
Travisano
M
,
Tahiliani
KG
,
Rached
MT
,
Mirza
S
.
Methyl donor supplementation prevents transgenerational amplification of obesity
.
Int J Obes
.
2008
;
32
(
9
):
1373
1379
.

271.

Nelson
SM
,
Sattar
N
,
Freeman
DJ
,
Walker
JD
,
Lindsay
RS
.
Inflammation and endothelial activation is evident at birth in offspring of mothers with type 1 diabetes
.
Diabetes
.
2007
;
56
(
11
):
2697
2704
.

272.

Hansen
NS
,
Strasko
KS
,
Hjort
L
,
Kelstrup
L
,
Houshmand-Øregaard
A
,
Schrölkamp
M
,
Schultz
HS
,
Scheele
C
,
Pedersen
BK
,
Ling
C
,
Clausen
TD
,
Damm
P
,
Vaag
A
,
Broholm
C
.
Fetal hyperglycemia changes human preadipocyte function in adult life
.
J Clin Endocrinol Metab
.
2017
;
102
(
4
):
1141
1150
.

273.

Lindsay
RS
,
Nelson
SM
,
Walker
JD
,
Greene
SA
,
Milne
G
,
Sattar
N
,
Pearson
DW
.
Programming of adiposity in offspring of mothers with type 1 diabetes at age 7 years
.
Diabetes Care
.
2010
;
33
(
5
):
1080
1085
.

274.

Ng
PC
,
Lee
CH
,
Lam
CW
,
Wong
E
,
Chan
IH
,
Fok
TF
.
Plasma ghrelin and resistin concentrations are suppressed in infants of insulin-dependent diabetic mothers
.
J Clin Endocrinol Metab
.
2004
;
89
(
11
):
5563
5568
.

275.

Sobngwi
E
,
Boudou
P
,
Mauvais-Jarvis
F
,
Leblanc
H
,
Velho
G
,
Vexiau
P
,
Porcher
R
,
Hadjadj
S
,
Pratley
R
,
Tataranni
PA
,
Calvo
F
,
Gautier
J-F
.
Effect of a diabetic environment in utero on predisposition to type 2 diabetes
.
Lancet
.
2003
;
361
(
9372
):
1861
1865
.

276.

Bonifacio
E
,
Pflüger
M
,
Marienfeld
S
,
Winkler
C
,
Hummel
M
,
Ziegler
A-G
.
Maternal type 1 diabetes reduces the risk of islet autoantibodies: relationships with birthweight and maternal HbA1c
.
Diabetologia
.
2008
;
51
(
7
):
1245
1252
.

277.

Lindell
N
,
Carlsson
A
,
Josefsson
A
,
Samuelsson
U
.
Maternal obesity as a risk factor for early childhood type 1 diabetes: a nationwide, prospective, population-based case-control study
.
Diabetologia
.
2018
;
61
(
1
):
130
137
.

278.

Hussen
HI
,
Persson
M
,
Moradi
T
.
Maternal overweight and obesity are associated with increased risk of type 1 diabetes in offspring of parents without diabetes regardless of ethnicity
.
Diabetologia
.
2015
;
58
(
7
):
1464
1473
.

279.

Plagemann
A
,
Heidrich
I
,
Götz
F
,
Rohde
W
,
Dörner
G
.
Obesity and enhanced diabetes and cardiovascular risk in adult rats due to early postnatal overfeeding
.
Exp Clin Endocrinol
.
1992
;
99
(
3
):
154
158
.

280.

Waterland
RA
,
Garza
C
.
Early postnatal nutrition determines adult pancreatic glucose-responsive insulin secretion and islet gene expression in rats
.
J Nutr
.
2002
;
132
(
3
):
357
364
.

281.

Li
G
,
Kohorst
JJ
,
Zhang
W
,
Laritsky
E
,
Kunde-Ramamoorthy
G
,
Baker
MS
,
Fiorotto
ML
,
Waterland
RA
.
Early postnatal nutrition determines adult physical activity and energy expenditure in female mice
.
Diabetes
.
2013
;
62
(
8
):
2773
2783
.

282.

Tomar
AS
,
Tallapragada
DS
,
Nongmaithem
SS
,
Shrestha
S
,
Yajnik
CS
,
Chandak
GR
.
Intrauterine programming of diabetes and adiposity
.
Curr Obes Rep
.
2015
;
4
(
4
):
418
428
.

283.

Buckley
J
,
Cohen
JD
,
Kramer
AF
,
McAuley
E
,
Mullen
SP
.
Cognitive control in the self-regulation of physical activity and sedentary behavior
.
Front Hum Neurosci
.
2014
;
8
:
747
.

284.

Bouchard
L
,
Thibault
S
,
Guay
SP
,
Santure
M
,
Monpetit
A
,
St-Pierre
J
,
Perron
P
,
Brisson
D
.
Leptin gene epigenetic adaptation to impaired glucose metabolism during pregnancy
.
Diabetes Care
.
2010
;
33
(
11
):
2436
2441
.

285.

Bouchard
L
,
Hivert
MF
,
Guay
SP
,
St-Pierre
J
,
Perron
P
,
Brisson
D
.
Placental adiponectin gene DNA methylation levels are associated with mothers’ blood glucose concentration
.
Diabetes
.
2012
;
61
(
5
):
1272
1280
.

286.

Bell
CG
,
Finer
S
,
Lindgren
CM
,
Wilson
GA
,
Rakyan
VK
,
Teschendorff
AE
,
Akan
P
,
Stupka
E
,
Down
TA
,
Prokopenko
I
,
Morison
IM
,
Mill
J
,
Pidsley
R
,
Deloukas
P
,
Frayling
TM
,
Hattersley
AT
,
McCarthy
MI
,
Beck
S
,
Hitman
GA
;
International Type 2 Diabetes 1q Consortium
.
Integrated genetic and epigenetic analysis identifies haplotype-specific methylation in the FTO type 2 diabetes and obesity susceptibility locus
.
PLoS One
.
2010
;
5
(
11
):
e14040
.

287.

Grunnet
LG
,
Nilsson
E
,
Ling
C
,
Hansen
T
,
Pedersen
O
,
Groop
L
,
Vaag
A
,
Poulsen
P
.
Regulation and function of FTO mRNA expression in human skeletal muscle and subcutaneous adipose tissue
.
Diabetes
.
2009
;
58
(
10
):
2402
2408
.

288.

Rui
J
,
Deng
S
,
Lebastchi
J
,
Clark
PL
,
Usmani-Brown
S
,
Herold
KC
.
Methylation of insulin DNA in response to proinflammatory cytokines during the progression of autoimmune diabetes in NOD mice
.
Diabetologia
.
2016
;
59
(
5
):
1021
1029
.

289.

Liang
F
,
Kume
S
,
Koya
D
.
SIRT1 and insulin resistance
.
Nat Rev Endocrinol
.
2009
;
5
(
7
):
367
373
.

290.

Banks
AS
,
Kon
N
,
Knight
C
,
Matsumoto
M
,
Gutiérrez-Juárez
R
,
Rossetti
L
,
Gu
W
,
Accili
D
.
SirT1 gain of function increases energy efficiency and prevents diabetes in mice
.
Cell Metab
.
2008
;
8
(
4
):
333
341
.

291.

Feige
JN
,
Lagouge
M
,
Canto
C
,
Strehle
A
,
Houten
SM
,
Milne
JC
,
Lambert
PD
,
Mataki
C
,
Elliott
PJ
,
Auwerx
J
.
Specific SIRT1 activation mimics low energy levels and protects against diet-induced metabolic disorders by enhancing fat oxidation
.
Cell Metab
.
2008
;
8
(
5
):
347
358
.

292.

Milne
JC
,
Lambert
PD
,
Schenk
S
,
Carney
DP
,
Smith
JJ
,
Gagne
DJ
,
Jin
L
,
Boss
O
,
Perni
RB
,
Vu
CB
,
Bemis
JE
,
Xie
R
,
Disch
JS
,
Ng
PY
,
Nunes
JJ
,
Lynch
AV
,
Yang
H
,
Galonek
H
,
Israelian
K
,
Choy
W
,
Iffland
A
,
Lavu
S
,
Medvedik
O
,
Sinclair
DA
,
Olefsky
JM
,
Jirousek
MR
,
Elliott
PJ
,
Westphal
CH
.
Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes
.
Nature
.
2007
;
450
(
7170
):
712
716
.

293.

Lee
SM
,
Yang
H
,
Tartar
DM
,
Gao
B
,
Luo
X
,
Ye
SQ
,
Zaghouani
H
,
Fang
D
.
Prevention and treatment of diabetes with resveratrol in a non-obese mouse model of type 1 diabetes
.
Diabetologia
.
2011
;
54
(
5
):
1136
1146
.

294.

Christensen
DP
,
Gysemans
C
,
Lundh
M
,
Dahllöf
MS
,
Noesgaard
D
,
Schmidt
SF
,
Mandrup
S
,
Birkbak
N
,
Workman
CT
,
Piemonti
L
,
Blaabjerg
L
,
Monzani
V
,
Fossati
G
,
Mascagni
P
,
Paraskevas
S
,
Aikin
RA
,
Billestrup
N
,
Grunnet
LG
,
Dinarello
CA
,
Mathieu
C
,
Mandrup-Poulsen
T
.
Lysine deacetylase inhibition prevents diabetes by chromatin-independent immunoregulation and β-cell protection
.
Proc Natl Acad Sci USA
.
2014
;
111
(
3
):
1055
1059
.

295.

Shah
R
,
Murthy
V
,
Pacold
M
,
Danielson
K
,
Tanriverdi
K
,
Larson
MG
,
Hanspers
K
,
Pico
A
,
Mick
E
,
Reis
J
,
de Ferranti
S
,
Freinkman
E
,
Levy
D
,
Hoffmann
U
,
Osganian
S
,
Das
S
,
Freedman
JE
.
Extracellular RNAs are associated with insulin resistance and metabolic phenotypes
.
Diabetes Care
.
2017
;
40
(
4
):
546
553
.

296.

Arner
P
,
Kulyté
A
.
MicroRNA regulatory networks in human adipose tissue and obesity
.
Nat Rev Endocrinol
.
2015
;
11
(
5
):
276
288
.

297.

Assmann
TS
,
Recamonde-Mendoza
M
,
De Souza
BM
,
Crispim
D
.
MicroRNA expression profiles and type 1 diabetes mellitus: systematic review and bioinformatic analysis
.
Endocr Connect
.
2017
;
6
(
8
):
773
790
.

298.

Guay
C
,
Regazzi
R
.
Circulating microRNAs as novel biomarkers for diabetes mellitus
.
Nat Rev Endocrinol
.
2013
;
9
(
9
):
513
521
.

299.

Frost
RJ
,
Olson
EN
.
Control of glucose homeostasis and insulin sensitivity by the Let-7 family of microRNAs
.
Proc Natl Acad Sci USA
.
2011
;
108
(
52
):
21075
21080
.

300.

Plaisance
V
,
Abderrahmani
A
,
Perret-Menoud
V
,
Jacquemin
P
,
Lemaigre
F
,
Regazzi
R
.
MicroRNA-9 controls the expression of Granuphilin/Slp4 and the secretory response of insulin-producing cells
.
J Biol Chem
.
2006
;
281
(
37
):
26932
26942
.

301.

Wijesekara
N
,
Zhang
LH
,
Kang
MH
,
Abraham
T
,
Bhattacharjee
A
,
Warnock
GL
,
Verchere
CB
,
Hayden
MR
.
miR-33a modulates ABCA1 expression, cholesterol accumulation, and insulin secretion in pancreatic islets
.
Diabetes
.
2012
;
61
(
3
):
653
658
.

302.

Lovis
P
,
Roggli
E
,
Laybutt
DR
,
Gattesco
S
,
Yang
JY
,
Widmann
C
,
Abderrahmani
A
,
Regazzi
R
.
Alterations in microRNA expression contribute to fatty acid-induced pancreatic β-cell dysfunction
.
Diabetes
.
2008
;
57
(
10
):
2728
2736
.

303.

Sun
LL
,
Jiang
BG
,
Li
WT
,
Zou
JJ
,
Shi
YQ
,
Liu
ZM
.
MicroRNA-15a positively regulates insulin synthesis by inhibiting uncoupling protein-2 expression
.
Diabetes Res Clin Pract
.
2011
;
91
(
1
):
94
100
.

304.

Fred
RG
,
Bang-Berthelsen
CH
,
Mandrup-Poulsen
T
,
Grunnet
LG
,
Welsh
N
.
High glucose suppresses human islet insulin biosynthesis by inducing miR-133a leading to decreased polypyrimidine tract binding protein-expression
.
PLoS One
.
2010
;
5
(
5
):
e10843
.

305.

Nieto
M
,
Hevia
P
,
Garcia
E
,
Klein
D
,
Alvarez-Cubela
S
,
Bravo-Egana
V
,
Rosero
S
,
Damaris Molano
R
,
Vargas
N
,
Ricordi
C
,
Pileggi
A
,
Diez
J
,
Domínguez-Bendala
J
,
Pastori
RL
.
Antisense miR-7 impairs insulin expression in developing pancreas and in cultured pancreatic buds
.
Cell Transplant
.
2012
;
21
(
8
):
1761
1774
.

306.

Baroukh
N
,
Ravier
MA
,
Loder
MK
,
Hill
EV
,
Bounacer
A
,
Scharfmann
R
,
Rutter
GA
,
Van Obberghen
E
.
MicroRNA-124a regulates Foxa2 expression and intracellular signaling in pancreatic β-cell lines
.
J Biol Chem
.
2007
;
282
(
27
):
19575
19588
.

307.

Kloosterman
WP
,
Lagendijk
AK
,
Ketting
RF
,
Moulton
JD
,
Plasterk
RH
.
Targeted inhibition of miRNA maturation with morpholinos reveals a role for miR-375 in pancreatic islet development
.
PLoS Biol
.
2007
;
5
(
8
):
e203
.

308.

Poy
MN
,
Hausser
J
,
Trajkovski
M
,
Braun
M
,
Collins
S
,
Rorsman
P
,
Zavolan
M
,
Stoffel
M
.
miR-375 maintains normal pancreatic α- and β-cell mass
.
Proc Natl Acad Sci USA
.
2009
;
106
(
14
):
5813
5818
.

309.

Seyhan
AA
,
Nunez Lopez
YO
,
Xie
H
,
Yi
F
,
Mathews
C
,
Pasarica
M
,
Pratley
RE
.
Pancreas-enriched miRNAs are altered in the circulation of subjects with diabetes: a pilot cross-sectional study
.
Sci Rep
.
2016
;
6
(
1
):
31479
.

310.

Cui
X
,
You
L
,
Zhu
L
,
Wang
X
,
Zhou
Y
,
Li
Y
,
Wen
J
,
Xia
Y
,
Wang
X
,
Ji
C
,
Guo
X
.
Change in circulating microRNA profile of obese children indicates future risk of adult diabetes
.
Metabolism
.
2018
;
78
:
95
105
.

311.

Miao
F
,
Chen
Z
,
Genuth
S
,
Paterson
A
,
Zhang
L
,
Wu
X
,
Li
SM
,
Cleary
P
,
Riggs
A
,
Harlan
DM
,
Lorenzi
G
,
Kolterman
O
,
Sun
W
,
Lachin
JM
,
Natarajan
R
;
DCCT/EDIC Research Group
.
Evaluating the role of epigenetic histone modifications in the metabolic memory of type 1 diabetes
.
Diabetes
.
2014
;
63
(
5
):
1748
1762
.

312.

Chen
Z
,
Miao
F
,
Paterson
AD
,
Lachin
JM
,
Zhang
L
,
Schones
DE
,
Wu
X
,
Wang
J
,
Tompkins
JD
,
Genuth
S
,
Braffett
BH
,
Riggs
AD
,
Natarajan
R
;
DCCT/EDIC Research Group
.
Epigenomic profiling reveals an association between persistence of DNA methylation and metabolic memory in the DCCT/EDIC type 1 diabetes cohort
.
Proc Natl Acad Sci USA
.
2016
;
113
(
21
):
E3002
E3011
.

313.

Agardh
E
,
Lundstig
A
,
Perfilyev
A
,
Volkov
P
,
Freiburghaus
T
,
Lindholm
E
,
Rönn
T
,
Agardh
CD
,
Ling
C
.
Genome-wide analysis of DNA methylation in subjects with type 1 diabetes identifies epigenetic modifications associated with proliferative diabetic retinopathy
.
BMC Med
.
2015
;
13
(
1
):
182
.

314.

Gu
T
,
Falhammar
H
,
Gu
HF
,
Brismar
K
.
Epigenetic analyses of the insulin-like growth factor binding protein 1 gene in type 1 diabetes and diabetic nephropathy
.
Clin Epigenetics
.
2014
;
6
(
1
):
10
.

315.

Rakyan
VK
,
Beyan
H
,
Down
TA
,
Hawa
MI
,
Maslau
S
,
Aden
D
,
Daunay
A
,
Busato
F
,
Mein
CA
,
Manfras
B
,
Dias
KR
,
Bell
CG
,
Tost
J
,
Boehm
BO
,
Beck
S
,
Leslie
RD
.
Identification of type 1 diabetes-associated DNA methylation variable positions that precede disease diagnosis
.
PLoS Genet
.
2011
;
7
(
9
):
e1002300
.

316.

Reddy
MA
,
Zhang
E
,
Natarajan
R
.
Epigenetic mechanisms in diabetic complications and metabolic memory
.
Diabetologia
.
2015
;
58
(
3
):
443
455
.

317.

Väremo
L
,
Henriksen
TI
,
Scheele
C
,
Broholm
C
,
Pedersen
M
,
Uhlén
M
,
Pedersen
BK
,
Nielsen
J
.
Type 2 diabetes and obesity induce similar transcriptional reprogramming in human myocytes
.
Genome Med
.
2017
;
9
(
1
):
47
.

318.

Pinnick
KE
,
Nicholson
G
,
Manolopoulos
KN
,
McQuaid
SE
,
Valet
P
,
Frayn
KN
,
Denton
N
,
Min
JL
,
Zondervan
KT
,
Fleckner
J
,
McCarthy
MI
,
Holmes
CC
,
Karpe
F
;
MolPAGE Consortium
.
Distinct developmental profile of lower-body adipose tissue defines resistance against obesity-associated metabolic complications
.
Diabetes
.
2014
;
63
(
11
):
3785
3797
.

319.

Turnbaugh
PJ
,
Ley
RE
,
Mahowald
MA
,
Magrini
V
,
Mardis
ER
,
Gordon
JI
.
An obesity-associated gut microbiome with increased capacity for energy harvest
.
Nature
.
2006
;
444
(
7122
):
1027
1031
.

320.

Leung
C
,
Rivera
L
,
Furness
JB
,
Angus
PW
.
The role of the gut microbiota in NAFLD
.
Nat Rev Gastroenterol Hepatol
.
2016
;
13
(
7
):
412
425
.

321.

Musso
G
,
Gambino
R
,
Cassader
M
.
Interactions between gut microbiota and host metabolism predisposing to obesity and diabetes
.
Annu Rev Med
.
2011
;
62
(
1
):
361
380
.

322.

Bäckhed
F
,
Ding
H
,
Wang
T
,
Hooper
LV
,
Koh
GY
,
Nagy
A
,
Semenkovich
CF
,
Gordon
JI
.
The gut microbiota as an environmental factor that regulates fat storage
.
Proc Natl Acad Sci USA
.
2004
;
101
(
44
):
15718
15723
.

323.

Ley
RE
,
Turnbaugh
PJ
,
Klein
S
,
Gordon
JI
.
Microbial ecology: human gut microbes associated with obesity
.
Nature
.
2006
;
444
(
7122
):
1022
1023
.

324.

Turnbaugh
PJ
,
Hamady
M
,
Yatsunenko
T
,
Cantarel
BL
,
Duncan
A
,
Ley
RE
,
Sogin
ML
,
Jones
WJ
,
Roe
BA
,
Affourtit
JP
,
Egholm
M
,
Henrissat
B
,
Heath
AC
,
Knight
R
,
Gordon
JI
.
A core gut microbiome in obese and lean twins
.
Nature
.
2009
;
457
(
7228
):
480
484
.

325.

Dumas
ME
,
Barton
RH
,
Toye
A
,
Cloarec
O
,
Blancher
C
,
Rothwell
A
,
Fearnside
J
,
Tatoud
R
,
Blanc
V
,
Lindon
JC
,
Mitchell
SC
,
Holmes
E
,
McCarthy
MI
,
Scott
J
,
Gauguier
D
,
Nicholson
JK
.
Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice
.
Proc Natl Acad Sci USA
.
2006
;
103
(
33
):
12511
12516
.

326.

Spencer
MD
,
Hamp
TJ
,
Reid
RW
,
Fischer
LM
,
Zeisel
SH
,
Fodor
AA
.
Association between composition of the human gastrointestinal microbiome and development of fatty liver with choline deficiency
.
Gastroenterology
.
2011
;
140
(
3
):
976
986
.

327.

Qin
J
,
Li
Y
,
Cai
Z
,
Li
S
,
Zhu
J
,
Zhang
F
,
Liang
S
,
Zhang
W
,
Guan
Y
,
Shen
D
,
Peng
Y
,
Zhang
D
,
Jie
Z
,
Wu
W
,
Qin
Y
,
Xue
W
,
Li
J
,
Han
L
,
Lu
D
,
Wu
P
,
Dai
Y
,
Sun
X
,
Li
Z
,
Tang
A
,
Zhong
S
,
Li
X
,
Chen
W
,
Xu
R
,
Wang
M
,
Feng
Q
,
Gong
M
,
Yu
J
,
Zhang
Y
,
Zhang
M
,
Hansen
T
,
Sanchez
G
,
Raes
J
,
Falony
G
,
Okuda
S
,
Almeida
M
,
LeChatelier
E
,
Renault
P
,
Pons
N
,
Batto
JM
,
Zhang
Z
,
Chen
H
,
Yang
R
,
Zheng
W
,
Li
S
,
Yang
H
,
Wang
J
,
Ehrlich
SD
,
Nielsen
R
,
Pedersen
O
,
Kristiansen
K
,
Wang
J
.
A metagenome-wide association study of gut microbiota in type 2 diabetes
.
Nature
.
2012
;
490
(
7418
):
55
60
.

328.

Forslund
K
,
Hildebrand
F
,
Nielsen
T
,
Falony
G
,
Le Chatelier
E
,
Sunagawa
S
,
Prifti
E
,
Vieira-Silva
S
,
Gudmundsdottir
V
,
Pedersen
HK
,
Arumugam
M
,
Kristiansen
K
,
Voigt
AY
,
Vestergaard
H
,
Hercog
R
,
Costea
PI
,
Kultima
JR
,
Li
J
,
Jørgensen
T
,
Levenez
F
,
Dore
J
,
Nielsen
HB
,
Brunak
S
,
Raes
J
,
Hansen
T
,
Wang
J
,
Ehrlich
SD
,
Bork
P
,
Pedersen
O
;
MetaHIT consortium
.
Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota
.
Nature
.
2015
;
528
(
7581
):
262
266
.

329.

Malin
SK
,
Kashyap
SR
.
Effects of various gastrointestinal procedures on β-cell function in obesity and type 2 diabetes
.
Surg Obes Relat Dis
.
2016
;
12
(
6
):
1213
1219
.

330.

Knip
M
,
Honkanen
J
.
Modulation of type 1 diabetes risk by the intestinal microbiome
.
Curr Diab Rep
.
2017
;
17
(
11
):
105
.

331.

Needell
JC
,
Zipris
D
.
The role of the intestinal microbiome in type 1 diabetes pathogenesis
.
Curr Diab Rep
.
2016
;
16
(
10
):
89
.

332.

Wen
L
,
Ley
RE
,
Volchkov
PY
,
Stranges
PB
,
Avanesyan
L
,
Stonebraker
AC
,
Hu
C
,
Wong
FS
,
Szot
GL
,
Bluestone
JA
,
Gordon
JI
,
Chervonsky
AV
.
Innate immunity and intestinal microbiota in the development of type 1 diabetes
.
Nature
.
2008
;
455
(
7216
):
1109
1113
.

333.

Brown
CT
,
Davis-Richardson
AG
,
Giongo
A
,
Gano
KA
,
Crabb
DB
,
Mukherjee
N
,
Casella
G
,
Drew
JC
,
Ilonen
J
,
Knip
M
,
Hyöty
H
,
Veijola
R
,
Simell
T
,
Simell
O
,
Neu
J
,
Wasserfall
CH
,
Schatz
D
,
Atkinson
MA
,
Triplett
EW
.
Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes
.
PLoS One
.
2011
;
6
(
10
):
e25792
.

334.

Soyucen
E
,
Gulcan
A
,
Aktuglu-Zeybek
AC
,
Onal
H
,
Kiykim
E
,
Aydin
A
.
Differences in the gut microbiota of healthy children and those with type 1 diabetes
.
Pediatr Int
.
2014
;
56
(
3
):
336
343
.

335.

Murri
M
,
Leiva
I
,
Gomez-Zumaquero
JM
,
Tinahones
FJ
,
Cardona
F
,
Soriguer
F
,
Queipo-Ortuño
MI
.
Gut microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study
.
BMC Med
.
2013
;
11
(
1
):
46
.

336.

Pellegrini
S
,
Sordi
V
,
Bolla
AM
,
Saita
D
,
Ferrarese
R
,
Canducci
F
,
Clementi
M
,
Invernizzi
F
,
Mariani
A
,
Bonfanti
R
,
Barera
G
,
Testoni
PA
,
Doglioni
C
,
Bosi
E
,
Piemonti
L
.
Duodenal mucosa of patients with type 1 diabetes shows distinctive inflammatory profile and microbiota
.
J Clin Endocrinol Metab
.
2017
;
102
(
5
):
1468
1477
.

337.

de Goffau
MC
,
Luopajärvi
K
,
Knip
M
,
Ilonen
J
,
Ruohtula
T
,
Härkönen
T
,
Orivuori
L
,
Hakala
S
,
Welling
GW
,
Harmsen
HJ
,
Vaarala
O
.
Fecal microbiota composition differs between children with β-cell autoimmunity and those without
.
Diabetes
.
2013
;
62
(
4
):
1238
1244
.

338.

Alkanani
AK
,
Hara
N
,
Gottlieb
PA
,
Ir
D
,
Robertson
CE
,
Wagner
BD
,
Frank
DN
,
Zipris
D
.
Alterations in intestinal microbiota correlate with susceptibility to type 1 diabetes
.
Diabetes
.
2015
;
64
(
10
):
3510
3520
.

339.

Kostic
AD
,
Gevers
D
,
Siljander
H
,
Vatanen
T
,
Hyötyläinen
T
,
Hämäläinen
AM
,
Peet
A
,
Tillmann
V
,
Pöhö
P
,
Mattila
I
,
Lähdesmäki
H
,
Franzosa
EA
,
Vaarala
O
,
de Goffau
M
,
Harmsen
H
,
Ilonen
J
,
Virtanen
SM
,
Clish
CB
,
Orešič
M
,
Huttenhower
C
,
Knip
M
,
Xavier
RJ
;
DIABIMMUNE Study Group
.
The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes
.
Cell Host Microbe
.
2015
;
17
(
2
):
260
273
.

340.

Neu
J
,
Reverte
CM
,
Mackey
AD
,
Liboni
K
,
Tuhacek-Tenace
LM
,
Hatch
M
,
Li
N
,
Caicedo
RA
,
Schatz
DA
,
Atkinson
M
.
Changes in intestinal morphology and permeability in the biobreeding rat before the onset of type 1 diabetes
.
J Pediatr Gastroenterol Nutr
.
2005
;
40
(
5
):
589
595
.

341.

Maffeis
C
,
Martina
A
,
Corradi
M
,
Quarella
S
,
Nori
N
,
Torriani
S
,
Plebani
M
,
Contreas
G
,
Felis
GE
.
Association between intestinal permeability and faecal microbiota composition in Italian children with beta cell autoimmunity at risk for type 1 diabetes
.
Diabetes Metab Res Rev
.
2016
;
32
(
7
):
700
709
.

342.

de Goffau
MC
,
Fuentes
S
,
van den Bogert
B
,
Honkanen
H
,
de Vos
WM
,
Welling
GW
,
Hyöty
H
,
Harmsen
HJ
.
Aberrant gut microbiota composition at the onset of type 1 diabetes in young children
.
Diabetologia
.
2014
;
57
(
8
):
1569
1577
.

343.

Rouxel
O
,
Da Silva
J
,
Beaudoin
L
,
Nel
I
,
Tard
C
,
Cagninacci
L
,
Kiaf
B
,
Oshima
M
,
Diedisheim
M
,
Salou
M
,
Corbett
A
,
Rossjohn
J
,
McCluskey
J
,
Scharfmann
R
,
Battaglia
M
,
Polak
M
,
Lantz
O
,
Beltrand
J
,
Lehuen
A
.
Cytotoxic and regulatory roles of mucosal-associated invariant T cells in type 1 diabetes
.
Nat Immunol
.
2017
;
18
(
12
):
1321
1331
.

344.

Pound
LD
,
Patrick
C
,
Eberhard
CE
,
Mottawea
W
,
Wang
GS
,
Abujamel
T
,
Vandenbeek
R
,
Stintzi
A
,
Scott
FW
.
Cathelicidin antimicrobial peptide: a novel regulator of islet function, islet regeneration, and selected gut bacteria
.
Diabetes
.
2015
;
64
(
12
):
4135
4147
.

345.

Perry
RJ
,
Peng
L
,
Barry
NA
,
Cline
GW
,
Zhang
D
,
Cardone
RL
,
Petersen
KF
,
Kibbey
RG
,
Goodman
AL
,
Shulman
GI
.
Acetate mediates a microbiome–brain–β-cell axis to promote metabolic syndrome
.
Nature
.
2016
;
534
(
7606
):
213
217
.

346.

Mariño
E
,
Richards
JL
,
McLeod
KH
,
Stanley
D
,
Yap
YA
,
Knight
J
,
McKenzie
C
,
Kranich
J
,
Oliveira
AC
,
Rossello
FJ
,
Krishnamurthy
B
,
Nefzger
CM
,
Macia
L
,
Thorburn
A
,
Baxter
AG
,
Morahan
G
,
Wong
LH
,
Polo
JM
,
Moore
RJ
,
Lockett
TJ
,
Clarke
JM
,
Topping
DL
,
Harrison
LC
,
Mackay
CR
.
Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes
.
Nat Immunol
.
2017
;
18
(
5
):
552
562
.

347.

Needell
JC
,
Ir
D
,
Robertson
CE
,
Kroehl
ME
,
Frank
DN
,
Zipris
D
.
Maternal treatment with short-chain fatty acids modulates the intestinal microbiota and immunity and ameliorates type 1 diabetes in the offspring
.
PLoS One
.
2017
;
12
(
9
):
e0183786
.

348.

Liu
J
,
Brutkiewicz
RR
.
Reduced circulating MAIT cells in obesity
.
J Immunol
.
2017
;
198
(
1
,
Suppl
):
197.17
.

349.

Magalhaes
I
,
Pingris
K
,
Poitou
C
,
Bessoles
S
,
Venteclef
N
,
Kiaf
B
,
Beaudoin
L
,
Da Silva
J
,
Allatif
O
,
Rossjohn
J
,
Kjer-Nielsen
L
,
McCluskey
J
,
Ledoux
S
,
Genser
L
,
Torcivia
A
,
Soudais
C
,
Lantz
O
,
Boitard
C
,
Aron-Wisnewsky
J
,
Larger
E
,
Clément
K
,
Lehuen
A
.
Mucosal-associated invariant T cell alterations in obese and type 2 diabetic patients
.
J Clin Invest
.
2015
;
125
(
4
):
1752
1762
.

350.

Bosi
E
,
Molteni
L
,
Radaelli
MG
,
Folini
L
,
Fermo
I
,
Bazzigaluppi
E
,
Piemonti
L
,
Pastore
MR
,
Paroni
R
.
Increased intestinal permeability precedes clinical onset of type 1 diabetes
.
Diabetologia
.
2006
;
49
(
12
):
2824
2827
.

351.

Odenwald
MA
,
Turner
JR
.
The intestinal epithelial barrier: a therapeutic target
?
Nat Rev Gastroenterol Hepatol
.
2017
;
14
(
1
):
9
21
.

352.

Tremaroli
V
,
Bäckhed
F
.
Functional interactions between the gut microbiota and host metabolism
.
Nature
.
2012
;
489
(
7415
):
242
249
.

353.

Flint
HJ
.
The impact of nutrition on the human microbiome
.
Nutr Rev
.
2012
;
70
(
Suppl 1
):
S10
S13
.

354.

Shortt
C
,
Hasselwander
O
,
Meynier
A
,
Nauta
A
,
Fernández
EN
,
Putz
P
,
Rowland
I
,
Swann
J
,
Türk
J
,
Vermeiren
J
,
Antoine
JM
.
Systematic review of the effects of the intestinal microbiota on selected nutrients and non-nutrients
.
Eur J Nutr
.
2018
;
57
(
1
):
25
49
.

355.

Zeevi
D
,
Korem
T
,
Zmora
N
,
Israeli
D
,
Rothschild
D
,
Weinberger
A
,
Ben-Yacov
O
,
Lador
D
,
Avnit-Sagi
T
,
Lotan-Pompan
M
,
Suez
J
,
Mahdi
JA
,
Matot
E
,
Malka
G
,
Kosower
N
,
Rein
M
,
Zilberman-Schapira
G
,
Dohnalová
L
,
Pevsner-Fischer
M
,
Bikovsky
R
,
Halpern
Z
,
Elinav
E
,
Segal
E
.
Personalized nutrition by prediction of glycemic responses
.
Cell
.
2015
;
163
(
5
):
1079
1094
.

356.

Suez
J
,
Korem
T
,
Zeevi
D
,
Zilberman-Schapira
G
,
Thaiss
CA
,
Maza
O
,
Israeli
D
,
Zmora
N
,
Gilad
S
,
Weinberger
A
,
Kuperman
Y
,
Harmelin
A
,
Kolodkin-Gal
I
,
Shapiro
H
,
Halpern
Z
,
Segal
E
,
Elinav
E
.
Artificial sweeteners induce glucose intolerance by altering the gut microbiota
.
Nature
.
2014
;
514
(
7521
):
181
186
.

357.

Neis
EP
,
Dejong
CH
,
Rensen
SS
.
The role of microbial amino acid metabolism in host metabolism
.
Nutrients
.
2015
;
7
(
4
):
2930
2946
.

358.

Pedersen
HK
,
Gudmundsdottir
V
,
Nielsen
HB
,
Hyotylainen
T
,
Nielsen
T
,
Jensen
BA
,
Forslund
K
,
Hildebrand
F
,
Prifti
E
,
Falony
G
,
Le Chatelier
E
,
Levenez
F
,
Doré
J
,
Mattila
I
,
Plichta
DR
,
Pöhö
P
,
Hellgren
LI
,
Arumugam
M
,
Sunagawa
S
,
Vieira-Silva
S
,
Jørgensen
T
,
Holm
JB
,
Trošt
K
,
Kristiansen
K
,
Brix
S
,
Raes
J
,
Wang
J
,
Hansen
T
,
Bork
P
,
Brunak
S
,
Oresic
M
,
Ehrlich
SD
,
Pedersen
O
;
MetaHIT Consortium
.
Human gut microbes impact host serum metabolome and insulin sensitivity
.
Nature
.
2016
;
535
(
7612
):
376
381
.

359.

Mayer-Davis
EJ
,
Dabelea
D
,
Crandell
JL
,
Crume
T
,
D’Agostino
RB
Jr
,
Dolan
L
,
King
IB
,
Lawrence
JM
,
Norris
JM
,
Pihoker
C
,
The
N
.
Nutritional factors and preservation of C-peptide in youth with recently diagnosed type 1 diabetes: SEARCH Nutrition Ancillary Study
.
Diabetes Care
.
2013
;
36
(
7
):
1842
1850
.

360.

Montandon
SA
,
Jornayvaz
FR
.
Effects of antidiabetic drugs on gut microbiota composition
.
Genes (Basel)
.
2017
;
8
(
10
):
250
.

361.

Karlsson
FH
,
Tremaroli
V
,
Nookaew
I
,
Bergström
G
,
Behre
CJ
,
Fagerberg
B
,
Nielsen
J
,
Bäckhed
F
.
Gut metagenome in European women with normal, impaired and diabetic glucose control
.
Nature
.
2013
;
498
(
7452
):
99
103
.

362.

Shin
NR
,
Lee
JC
,
Lee
HY
,
Kim
MS
,
Whon
TW
,
Lee
MS
,
Bae
JW
.
An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice
.
Gut
.
2014
;
63
(
5
):
727
735
.

363.

de la Cuesta-Zuluaga
J
,
Mueller
NT
,
Corrales-Agudelo
V
,
Velásquez-Mejía
EP
,
Carmona
JA
,
Abad
JM
,
Escobar
JS
.
Metformin is associated with higher relative abundance of mucin-degrading Akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the gut
.
Diabetes Care
.
2017
;
40
(
1
):
54
62
.

364.

Plovier
H
,
Everard
A
,
Druart
C
,
Depommier
C
,
Van Hul
M
,
Geurts
L
,
Chilloux
J
,
Ottman
N
,
Duparc
T
,
Lichtenstein
L
,
Myridakis
A
,
Delzenne
NM
,
Klievink
J
,
Bhattacharjee
A
,
van der Ark
KC
,
Aalvink
S
,
Martinez
LO
,
Dumas
ME
,
Maiter
D
,
Loumaye
A
,
Hermans
MP
,
Thissen
JP
,
Belzer
C
,
de Vos
WM
,
Cani
PD
.
A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice
.
Nat Med
.
2017
;
23
(
1
):
107
113
.

365.

Wilson
ID
,
Nicholson
JK
.
Gut microbiome interactions with drug metabolism, efficacy, and toxicity
.
Transl Res
.
2017
;
179
:
204
222
.

366.

Schmidt
TSB
,
Raes
J
,
Bork
P
.
The human gut microbiome: from association to modulation
.
Cell
.
2018
;
172
(
6
):
1198
1215
.

367.

Ouchi
N
,
Parker
JL
,
Lugus
JJ
,
Walsh
K
.
Adipokines in inflammation and metabolic disease
.
Nat Rev Immunol
.
2011
;
11
(
2
):
85
97
.

368.

Crewe
C
,
An
YA
,
Scherer
PE
.
The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis
.
J Clin Invest
.
2017
;
127
(
1
):
74
82
.

369.

Pou
KM
,
Massaro
JM
,
Hoffmann
U
,
Lieb
K
,
Vasan
RS
,
O’Donnell
CJ
,
Fox
CS
.
Patterns of abdominal fat distribution: the Framingham Heart Study
.
Diabetes Care
.
2009
;
32
(
3
):
481
485
.

370.

Bonora
E
,
Del Prato
S
,
Bonadonna
RC
,
Gulli
G
,
Solini
A
,
Shank
ML
,
Ghiatas
AA
,
Lancaster
JL
,
Kilcoyne
RF
,
Alyassin
AM
,
DeFronzo
RA
.
Total body fat content and fat topography are associated differently with in vivo glucose metabolism in nonobese and obese nondiabetic women
.
Diabetes
.
1992
;
41
(
9
):
1151
1159
.

371.

Després
JP
,
Nadeau
A
,
Tremblay
A
,
Ferland
M
,
Moorjani
S
,
Lupien
PJ
,
Thériault
G
,
Pinault
S
,
Bouchard
C
.
Role of deep abdominal fat in the association between regional adipose tissue distribution and glucose tolerance in obese women
.
Diabetes
.
1989
;
38
(
3
):
304
309
.

372.

Dubé
MC
,
Joanisse
DR
,
Prud’homme
D
,
Lemieux
S
,
Bouchard
C
,
Pérusse
L
,
Lavoie
C
,
Weisnagel
SJ
.
Muscle adiposity and body fat distribution in type 1 and type 2 diabetes: varying relationships according to diabetes type
.
Int J Obes
.
2006
;
30
(
12
):
1721
1728
.

373.

Yusuf
S
,
Hawken
S
,
Ounpuu
S
,
Bautista
L
,
Franzosi
MG
,
Commerford
P
,
Lang
CC
,
Rumboldt
Z
,
Onen
CL
,
Lisheng
L
,
Tanomsup
S
,
Wangai
P
Jr
,
Razak
F
,
Sharma
AM
,
Anand
SS
;
INTERHEART Study Investigators
.
Obesity and the risk of myocardial infarction in 27,000 participants from 52 countries: a case-control study
.
Lancet
.
2005
;
366
(
9497
):
1640
1649
.

374.

Goodpaster
BH
,
Thaete
FL
,
Kelley
DE
.
Thigh adipose tissue distribution is associated with insulin resistance in obesity and in type 2 diabetes mellitus
.
Am J Clin Nutr
.
2000
;
71
(
4
):
885
892
.

375.

Perseghin
G
,
Lattuada
G
,
Danna
M
,
Sereni
LP
,
Maffi
P
,
De Cobelli
F
,
Battezzati
A
,
Secchi
A
,
Del Maschio
A
,
Luzi
L
.
Insulin resistance, intramyocellular lipid content, and plasma adiponectin in patients with type 1 diabetes
.
Am J Physiol Endocrinol Metab
.
2003
;
285
(
6
):
E1174
E1181
.

376.

Ding
J
,
Hsu
FC
,
Harris
TB
,
Liu
Y
,
Kritchevsky
SB
,
Szklo
M
,
Ouyang
P
,
Espeland
MA
,
Lohman
KK
,
Criqui
MH
,
Allison
M
,
Bluemke
DA
,
Carr
JJ
.
The association of pericardial fat with incident coronary heart disease: the Multi-Ethnic Study of Atherosclerosis (MESA)
.
Am J Clin Nutr
.
2009
;
90
(
3
):
499
504
.

377.

Yang
FS
,
Yun
CH
,
Wu
TH
,
Hsieh
YC
,
Bezerra
HG
,
Liu
CC
,
Wu
YJ
,
Kuo
JY
,
Hung
CL
,
Hou
CJ
,
Yeh
HI
,
Lee
JJ
,
Bulwer
BE
,
Cury
RC
.
High pericardial and peri-aortic adipose tissue burden in pre-diabetic and diabetic subjects
.
BMC Cardiovasc Disord
.
2013
;
13
(
1
):
98
.

378.

Alman
AC
,
Smith
SR
,
Eckel
RH
,
Hokanson
JE
,
Burkhardt
BR
,
Sudini
PR
,
Wu
Y
,
Schauer
IE
,
Pereira
RI
,
Snell-Bergeon
JK
.
The ratio of pericardial to subcutaneous adipose tissues is associated with insulin resistance
.
Obesity (Silver Spring)
.
2017
;
25
(
7
):
1284
1291
.

379.

Divoux
A
,
Karastergiou
K
,
Xie
H
,
Guo
W
,
Perera
RJ
,
Fried
SK
,
Smith
SR
.
Identification of a novel lncRNA in gluteal adipose tissue and evidence for its positive effect on preadipocyte differentiation
.
Obesity (Silver Spring)
.
2014
;
22
(
8
):
1781
1785
.

380.

Divoux
A
,
Xie
H
,
Li
JL
,
Karastergiou
K
,
Perera
RJ
,
Chang
RJ
,
Fried
SK
,
Smith
SR
.
MicroRNA-196 regulates HOX gene expression in human gluteal adipose tissue
.
Obesity (Silver Spring)
.
2017
;
25
(
8
):
1375
1383
.

381.

Cummings
BP
,
Bettaieb
A
,
Graham
JL
,
Stanhope
KL
,
Dill
R
,
Morton
GJ
,
Haj
FG
,
Havel
PJ
.
Subcutaneous administration of leptin normalizes fasting plasma glucose in obese type 2 diabetic UCD-T2DM rats
.
Proc Natl Acad Sci USA
.
2011
;
108
(
35
):
14670
14675
.

382.

Miller
RA
,
Chu
Q
,
Le Lay
J
,
Scherer
PE
,
Ahima
RS
,
Kaestner
KH
,
Foretz
M
,
Viollet
B
,
Birnbaum
MJ
.
Adiponectin suppresses gluconeogenic gene expression in mouse hepatocytes independent of LKB1-AMPK signaling
.
J Clin Invest
.
2011
;
121
(
6
):
2518
2528
.

383.

Sugii
S
,
Olson
P
,
Sears
DD
,
Saberi
M
,
Atkins
AR
,
Barish
GD
,
Hong
SH
,
Castro
GL
,
Yin
YQ
,
Nelson
MC
,
Hsiao
G
,
Greaves
DR
,
Downes
M
,
Yu
RT
,
Olefsky
JM
,
Evans
RM
.
PPARγ activation in adipocytes is sufficient for systemic insulin sensitization
.
Proc Natl Acad Sci USA
.
2009
;
106
(
52
):
22504
22509
.

384.

Pereira
RI
,
Snell-Bergeon
JK
,
Erickson
C
,
Schauer
IE
,
Bergman
BC
,
Rewers
M
,
Maahs
DM
.
Adiponectin dysregulation and insulin resistance in type 1 diabetes
.
J Clin Endocrinol Metab
.
2012
;
97
(
4
):
E642
E647
.

385.

Martos-Moreno
GA
,
Barrios
V
,
Soriano-Guillén
L
,
Argente
J
.
Relationship between adiponectin levels, acylated ghrelin levels, and short-term body mass index changes in children with diabetes mellitus type 1 at diagnosis and after insulin therapy
.
Eur J Endocrinol
.
2006
;
155
(
5
):
757
761
.

386.

Azar
ST
,
Zalloua
PA
,
Zantout
MS
,
Shahine
CH
,
Salti
I
.
Leptin levels in patients with type 1 diabetes receiving intensive insulin therapy compared with those in patients receiving conventional insulin therapy
.
J Endocrinol Invest
.
2002
;
25
(
8
):
724
726
.

387.

Yoshioka
M
,
Kayo
T
,
Ikeda
T
,
Koizumi
A
.
A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice
.
Diabetes
.
1997
;
46
(
5
):
887
894
.

388.

Ueno
N
,
Inui
A
,
Kalra
PS
,
Kalra
SP
.
Leptin transgene expression in the hypothalamus enforces euglycemia in diabetic, insulin-deficient nonobese Akita mice and leptin-deficient obese ob/ob mice
.
Peptides
.
2006
;
27
(
9
):
2332
2342
.

389.

Naito
M
,
Fujikura
J
,
Ebihara
K
,
Miyanaga
F
,
Yokoi
H
,
Kusakabe
T
,
Yamamoto
Y
,
Son
C
,
Mukoyama
M
,
Hosoda
K
,
Nakao
K
.
Therapeutic impact of leptin on diabetes, diabetic complications, and longevity in insulin-deficient diabetic mice
.
Diabetes
.
2011
;
60
(
9
):
2265
2273
.

390.

Yu
X
,
Park
BH
,
Wang
MY
,
Wang
ZV
,
Unger
RH
.
Making insulin-deficient type 1 diabetic rodents thrive without insulin
.
Proc Natl Acad Sci USA
.
2008
;
105
(
37
):
14070
14075
.

391.

Wang
MY
,
Chen
L
,
Clark
GO
,
Lee
Y
,
Stevens
RD
,
Ilkayeva
OR
,
Wenner
BR
,
Bain
JR
,
Charron
MJ
,
Newgard
CB
,
Unger
RH
.
Leptin therapy in insulin-deficient type I diabetes
.
Proc Natl Acad Sci USA
.
2010
;
107
(
11
):
4813
4819
.

392.

Fukushima
M
,
Hattori
Y
,
Tsukada
H
,
Koga
K
,
Kajiwara
E
,
Kawano
K
,
Kobayashi
T
,
Kamata
K
,
Maitani
Y
.
Adiponectin gene therapy of streptozotocin-induced diabetic mice using hydrodynamic injection
.
J Gene Med
.
2007
;
9
(
11
):
976
985
.

393.

Kruger
AJ
,
Yang
C
,
Lipson
KL
,
Pino
SC
,
Leif
JH
,
Hogan
CM
,
Whalen
BJ
,
Guberski
DL
,
Lee
Y
,
Unger
RH
,
Greiner
DL
,
Rossini
AA
,
Bortell
R
.
Leptin treatment confers clinical benefit at multiple stages of virally induced type 1 diabetes in BB rats
.
Autoimmunity
.
2011
;
44
(
2
):
137
148
.

394.

Chen
H
,
Zheng
C
,
Zhang
X
,
Li
J
,
Li
J
,
Zheng
L
,
Huang
K
.
Apelin alleviates diabetes-associated endoplasmic reticulum stress in the pancreas of Akita mice
.
Peptides
.
2011
;
32
(
8
):
1634
1639
.

395.

Vasandani
C
,
Clark
GO
,
Adams-Huet
B
,
Quittner
C
,
Garg
A
.
Efficacy and safety of metreleptin therapy in patients with type 1 diabetes: a pilot study
.
Diabetes Care
.
2017
;
40
(
5
):
694
697
.

396.

Sun
K
,
Park
J
,
Gupta
OT
,
Holland
WL
,
Auerbach
P
,
Zhang
N
,
Goncalves Marangoni
R
,
Nicoloro
SM
,
Czech
MP
,
Varga
J
,
Ploug
T
,
An
Z
,
Scherer
PE
.
Endotrophin triggers adipose tissue fibrosis and metabolic dysfunction
.
Nat Commun
.
2014
;
5
(
1
):
3485
.

397.

Khan
T
,
Muise
ES
,
Iyengar
P
,
Wang
ZV
,
Chandalia
M
,
Abate
N
,
Zhang
BB
,
Bonaldo
P
,
Chua
S
,
Scherer
PE
.
Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI
.
Mol Cell Biol
.
2009
;
29
(
6
):
1575
1591
.

398.

Konrad
D
,
Rudich
A
,
Schoenle
EJ
.
Improved glucose tolerance in mice receiving intraperitoneal transplantation of normal fat tissue
.
Diabetologia
.
2007
;
50
(
4
):
833
839
.

399.

Bahr
J
,
Klöting
N
,
Klöting
I
,
Follak
N
.
Transplantation of adipose tissue protects BB/OK rats from type 1 diabetes development
.
Transpl Immunol
.
2011
;
24
(
4
):
238
240
.

400.

Sparks
LM
,
Pasarica
M
,
Sereda
O
,
deJonge
L
,
Thomas
S
,
Loggins
H
,
Xie
H
,
Miles
JM
,
Smith
SR
.
Effect of adipose tissue on the sexual dimorphism in metabolic flexibility
.
Metabolism
.
2009
;
58
(
11
):
1564
1571
.

401.

Gunawardana
SC
,
Piston
DW
.
Reversal of type 1 diabetes in mice by brown adipose tissue transplant
.
Diabetes
.
2012
;
61
(
3
):
674
682
.

402.

Kim
JH
,
Bae
KH
,
Choi
YK
,
Go
Y
,
Choe
M
,
Jeon
YH
,
Lee
HW
,
Koo
SH
,
Perfield
JW
II
,
Harris
RA
,
Lee
IK
,
Park
KG
.
Fibroblast growth factor 21 analogue LY2405319 lowers blood glucose in streptozotocin-induced insulin-deficient diabetic mice by restoring brown adipose tissue function
.
Diabetes Obes Metab
.
2015
;
17
(
2
):
161
169
.

403.

Gaich
G
,
Chien
JY
,
Fu
H
,
Glass
LC
,
Deeg
MA
,
Holland
WL
,
Kharitonenkov
A
,
Bumol
T
,
Schilske
HK
,
Moller
DE
.
The effects of LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes
.
Cell Metab
.
2013
;
18
(
3
):
333
340
.

404.

Tran
TT
,
Kahn
CR
.
Transplantation of adipose tissue and stem cells: role in metabolism and disease
.
Nat Rev Endocrinol
.
2010
;
6
(
4
):
195
213
.

405.

Butler
AE
,
Janson
J
,
Bonner-Weir
S
,
Ritzel
R
,
Rizza
RA
,
Butler
PC
.
β-Cell deficit and increased β-cell apoptosis in humans with type 2 diabetes
.
Diabetes
.
2003
;
52
(
1
):
102
110
.

406.

Lorenzo
C
,
Wagenknecht
LE
,
D’Agostino
RB
Jr
,
Rewers
MJ
,
Karter
AJ
,
Haffner
SM
.
Insulin resistance, β-cell dysfunction, and conversion to type 2 diabetes in a multiethnic population: the Insulin Resistance Atherosclerosis Study
.
Diabetes Care
.
2010
;
33
(
1
):
67
72
.

407.

Kashyap
S
,
Belfort
R
,
Gastaldelli
A
,
Pratipanawatr
T
,
Berria
R
,
Pratipanawatr
W
,
Bajaj
M
,
Mandarino
L
,
DeFronzo
R
,
Cusi
K
.
A sustained increase in plasma free fatty acids impairs insulin secretion in nondiabetic subjects genetically predisposed to develop type 2 diabetes
.
Diabetes
.
2003
;
52
(
10
):
2461
2474
.

408.

Maedler
K
,
Spinas
GA
,
Lehmann
R
,
Sergeev
P
,
Weber
M
,
Fontana
A
,
Kaiser
N
,
Donath
MY
.
Glucose induces β-cell apoptosis via upregulation of the Fas receptor in human islets
.
Diabetes
.
2001
;
50
(
8
):
1683
1690
.

409.

Villareal
DT
,
Banks
MR
,
Patterson
BW
,
Polonsky
KS
,
Klein
S
.
Weight loss therapy improves pancreatic endocrine function in obese older adults
.
Obesity (Silver Spring)
.
2008
;
16
(
6
):
1349
1354
.

410.

Li
G
,
Zhang
P
,
Wang
J
,
Gregg
EW
,
Yang
W
,
Gong
Q
,
Li
H
,
Li
H
,
Jiang
Y
,
An
Y
,
Shuai
Y
,
Zhang
B
,
Zhang
J
,
Thompson
TJ
,
Gerzoff
RB
,
Roglic
G
,
Hu
Y
,
Bennett
PH
.
The long-term effect of lifestyle interventions to prevent diabetes in the China Da Qing Diabetes Prevention Study: a 20-year follow-up study
.
Lancet
.
2008
;
371
(
9626
):
1783
1789
.

411.

Saaristo
T
,
Moilanen
L
,
Korpi-Hyövälti
E
,
Vanhala
M
,
Saltevo
J
,
Niskanen
L
,
Jokelainen
J
,
Peltonen
M
,
Oksa
H
,
Tuomilehto
J
,
Uusitupa
M
,
Keinänen-Kiukaanniemi
S
.
Lifestyle intervention for prevention of type 2 diabetes in primary health care: one-year follow-up of the Finnish National Diabetes Prevention Program (FIN-D2D)
.
Diabetes Care
.
2010
;
33
(
10
):
2146
2151
.

412.

Hamman
RF
,
Wing
RR
,
Edelstein
SL
,
Lachin
JM
,
Bray
GA
,
Delahanty
L
,
Hoskin
M
,
Kriska
AM
,
Mayer-Davis
EJ
,
Pi-Sunyer
X
,
Regensteiner
J
,
Venditti
B
,
Wylie-Rosett
J
.
Effect of weight loss with lifestyle intervention on risk of diabetes
.
Diabetes Care
.
2006
;
29
(
9
):
2102
2107
.

413.

Yu
HW
,
Lee
YJ
,
Cho
WI
,
Lee
YA
,
Shin
CH
,
Yang
SW
.
Preserved C-peptide levels in overweight or obese compared with underweight children upon diagnosis of type 1 diabetes mellitus
.
Ann Pediatr Endocrinol Metab
.
2015
;
20
(
2
):
92
97
.

414.

Gunawardana
SC
.
Benefits of healthy adipose tissue in the treatment of diabetes
.
World J Diabetes
.
2014
;
5
(
4
):
420
430
.

415.

McGee
P
,
Steffes
M
,
Nowicki
M
,
Bayless
M
,
Gubitosi-Klug
R
,
Cleary
P
,
Lachin
J
,
Palmer
J
;
DCCT/EDIC Research Group
.
Insulin secretion measured by stimulated C-peptide in long-established type 1 diabetes in the Diabetes Control and Complications Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications (EDIC) cohort: a pilot study
.
Diabet Med
.
2014
;
31
(
10
):
1264
1268
.

416.

Davis
AK
,
DuBose
SN
,
Haller
MJ
,
Miller
KM
,
DiMeglio
LA
,
Bethin
KE
,
Goland
RS
,
Greenberg
EM
,
Liljenquist
DR
,
Ahmann
AJ
,
Marcovina
SM
,
Peters
AL
,
Beck
RW
,
Greenbaum
CJ
;
T1D Exchange Clinic Network
.
Prevalence of detectable C-peptide according to age at diagnosis and duration of type 1 diabetes
.
Diabetes Care
.
2015
;
38
(
3
):
476
481
.

417.

Krogvold
L
,
Skog
O
,
Sundström
G
,
Edwin
B
,
Buanes
T
,
Hanssen
KF
,
Ludvigsson
J
,
Grabherr
M
,
Korsgren
O
,
Dahl-Jørgensen
K
.
Function of isolated pancreatic islets from patients at onset of type 1 diabetes: insulin secretion can be restored after some days in a nondiabetogenic environment in vitro: results from the DiViD study
.
Diabetes
.
2015
;
64
(
7
):
2506
2512
.

418.

Wang
Z
,
York
NW
,
Nichols
CG
,
Remedi
MS
.
Pancreatic β cell dedifferentiation in diabetes and redifferentiation following insulin therapy
.
Cell Metab
.
2014
;
19
(
5
):
872
882
.

419.

Carlsson
PO
,
Schwarcz
E
,
Korsgren
O
,
Le Blanc
K
.
Preserved β-cell function in type 1 diabetes by mesenchymal stromal cells
.
Diabetes
.
2015
;
64
(
2
):
587
592
.

420.

Kelley
DE
,
Goodpaster
B
,
Wing
RR
,
Simoneau
JA
.
Skeletal muscle fatty acid metabolism in association with insulin resistance, obesity, and weight loss
.
Am J Physiol
.
1999
;
277
(
6 Pt 1
):
E1130
E1141
.

421.

Simoneau
JA
,
Veerkamp
JH
,
Turcotte
LP
,
Kelley
DE
.
Markers of capacity to utilize fatty acids in human skeletal muscle: relation to insulin resistance and obesity and effects of weight loss
.
FASEB J
.
1999
;
13
(
14
):
2051
2060
.

422.

Heinonen
S
,
Buzkova
J
,
Muniandy
M
,
Kaksonen
R
,
Ollikainen
M
,
Ismail
K
,
Hakkarainen
A
,
Lundbom
J
,
Lundbom
N
,
Vuolteenaho
K
,
Moilanen
E
,
Kaprio
J
,
Rissanen
A
,
Suomalainen
A
,
Pietiläinen
KH
.
Impaired mitochondrial biogenesis in adipose tissue in acquired obesity
.
Diabetes
.
2015
;
64
(
9
):
3135
3145
.

423.

Kelley
DE
,
He
J
,
Menshikova
EV
,
Ritov
VB
.
Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes
.
Diabetes
.
2002
;
51
(
10
):
2944
2950
.

424.

Ritov
VB
,
Menshikova
EV
,
He
J
,
Ferrell
RE
,
Goodpaster
BH
,
Kelley
DE
.
Deficiency of subsarcolemmal mitochondria in obesity and type 2 diabetes
.
Diabetes
.
2005
;
54
(
1
):
8
14
.

425.

Mootha
VK
,
Lindgren
CM
,
Eriksson
KF
,
Subramanian
A
,
Sihag
S
,
Lehar
J
,
Puigserver
P
,
Carlsson
E
,
Ridderstråle
M
,
Laurila
E
,
Houstis
N
,
Daly
MJ
,
Patterson
N
,
Mesirov
JP
,
Golub
TR
,
Tamayo
P
,
Spiegelman
B
,
Lander
ES
,
Hirschhorn
JN
,
Altshuler
D
,
Groop
LC
.
PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes
.
Nat Genet
.
2003
;
34
(
3
):
267
273
.

426.

Patti
ME
,
Butte
AJ
,
Crunkhorn
S
,
Cusi
K
,
Berria
R
,
Kashyap
S
,
Miyazaki
Y
,
Kohane
I
,
Costello
M
,
Saccone
R
,
Landaker
EJ
,
Goldfine
AB
,
Mun
E
,
DeFronzo
R
,
Finlayson
J
,
Kahn
CR
,
Mandarino
LJ
.
Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: potential role of PGC1 and NRF1
.
Proc Natl Acad Sci USA
.
2003
;
100
(
14
):
8466
8471
.

427.

Mitchell
T
,
Johnson
MS
,
Ouyang
X
,
Chacko
BK
,
Mitra
K
,
Lei
X
,
Gai
Y
,
Moore
DR
,
Barnes
S
,
Zhang
J
,
Koizumi
A
,
Ramanadham
S
,
Darley-Usmar
VM
.
Dysfunctional mitochondrial bioenergetics and oxidative stress in Akita+/Ins2-derived β-cells
.
Am J Physiol Endocrinol Metab
.
2013
;
305
(
5
):
E585
E599
.

428.

Goodpaster
BH
,
Theriault
R
,
Watkins
SC
,
Kelley
DE
.
Intramuscular lipid content is increased in obesity and decreased by weight loss
.
Metabolism
.
2000
;
49
(
4
):
467
472
.

429.

Coen
PM
,
Dubé
JJ
,
Amati
F
,
Stefanovic-Racic
M
,
Ferrell
RE
,
Toledo
FG
,
Goodpaster
BH
.
Insulin resistance is associated with higher intramyocellular triglycerides in type I but not type II myocytes concomitant with higher ceramide content
.
Diabetes
.
2010
;
59
(
1
):
80
88
.

430.

Shulman
GI
.
Cellular mechanisms of insulin resistance
.
J Clin Invest
.
2000
;
106
(
2
):
171
176
.

431.

Petersen
KF
,
Dufour
S
,
Befroy
D
,
Garcia
R
,
Shulman
GI
.
Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes
.
N Engl J Med
.
2004
;
350
(
7
):
664
671
.

432.

Chavez
AO
,
Kamath
S
,
Jani
R
,
Sharma
LK
,
Monroy
A
,
Abdul-Ghani
MA
,
Centonze
VE
,
Sathyanarayana
P
,
Coletta
DK
,
Jenkinson
CP
,
Bai
Y
,
Folli
F
,
Defronzo
RA
,
Tripathy
D
.
Effect of short-term free fatty acids elevation on mitochondrial function in skeletal muscle of healthy individuals
.
J Clin Endocrinol Metab
.
2010
;
95
(
1
):
422
429
.

433.

Richardson
DK
,
Kashyap
S
,
Bajaj
M
,
Cusi
K
,
Mandarino
SJ
,
Finlayson
J
,
DeFronzo
RA
,
Jenkinson
CP
,
Mandarino
LJ
.
Lipid infusion decreases the expression of nuclear encoded mitochondrial genes and increases the expression of extracellular matrix genes in human skeletal muscle
.
J Biol Chem
.
2005
;
280
(
11
):
10290
10297
.

434.

Bugger
H
,
Chen
D
,
Riehle
C
,
Soto
J
,
Theobald
HA
,
Hu
XX
,
Ganesan
B
,
Weimer
BC
,
Abel
ED
.
Tissue-specific remodeling of the mitochondrial proteome in type 1 diabetic Akita mice
.
Diabetes
.
2009
;
58
(
9
):
1986
1997
.

435.

Brissova
M
,
Shiota
M
,
Nicholson
WE
,
Gannon
M
,
Knobel
SM
,
Piston
DW
,
Wright
CV
,
Powers
AC
.
Reduction in pancreatic transcription factor PDX-1 impairs glucose-stimulated insulin secretion
.
J Biol Chem
.
2002
;
277
(
13
):
11225
11232
.

436.

Soleimanpour
SA
,
Ferrari
AM
,
Raum
JC
,
Groff
DN
,
Yang
J
,
Kaufman
BA
,
Stoffers
DA
.
Diabetes susceptibility genes Pdx1 and Clec16a function in a pathway regulating mitophagy in β-cells
.
Diabetes
.
2015
;
64
(
10
):
3475
3484
.

437.

Stump
CS
,
Short
KR
,
Bigelow
ML
,
Schimke
JM
,
Nair
KS
.
Effect of insulin on human skeletal muscle mitochondrial ATP production, protein synthesis, and mRNA transcripts
.
Proc Natl Acad Sci USA
.
2003
;
100
(
13
):
7996
8001
.

438.

Cheng
Z
,
Guo
S
,
Copps
K
,
Dong
X
,
Kollipara
R
,
Rodgers
JT
,
Depinho
RA
,
Puigserver
P
,
White
MF
.
Foxo1 integrates insulin signaling with mitochondrial function in the liver
.
Nat Med
.
2009
;
15
(
11
):
1307
1311
.

439.

Dallon
BW
,
Parker
BA
,
Hodson
AE
,
Tippetts
TS
,
Harrison
ME
,
Appiah
MMA
,
Witt
JE
,
Gibbs
JL
,
Gray
HM
,
Sant
TM
,
Bikman
BT
.
Insulin selectively reduces mitochondrial uncoupling in brown adipose tissue in mice
.
Biochem J
.
2018
;
475
(
3
):
561
569
.

440.

Bregman
JA
,
Herren
DJ
,
Estopinal
CB
,
Chocron
IM
,
Harlow
PA
,
Warden
C
,
Brantley
MA
Jr
,
Samuels
DC
.
Mitochondrial haplogroups affect severity but not prevalence of diabetic retinopathy
.
Invest Ophthalmol Vis Sci
.
2017
;
58
(
2
):
1346
1351
.

441.

Swan
EJ
,
Salem
RM
,
Sandholm
N
,
Tarnow
L
,
Rossing
P
,
Lajer
M
,
Groop
PH
,
Maxwell
AP
,
McKnight
AJ
;
GENIE Consortium
.
Genetic risk factors affecting mitochondrial function are associated with kidney disease in people with type 1 diabetes
.
Diabet Med
.
2015
;
32
(
8
):
1104
1109
.

442.

Qi
H
,
Casalena
G
,
Shi
S
,
Yu
L
,
Ebefors
K
,
Sun
Y
,
Zhang
W
,
D’Agati
V
,
Schlondorff
D
,
Haraldsson
B
,
Böttinger
E
,
Daehn
I
.
Glomerular endothelial mitochondrial dysfunction is essential and characteristic of diabetic kidney disease susceptibility
.
Diabetes
.
2017
;
66
(
3
):
763
778
.

443.

Swan
EJ
,
Maxwell
AP
,
McKnight
AJ
.
Distinct methylation patterns in genes that affect mitochondrial function are associated with kidney disease in blood-derived DNA from individuals with type 1 diabetes
.
Diabet Med
.
2015
;
32
(
8
):
1110
1115
.

444.

Lei
H
,
Nahum-Shani
I
,
Lynch
K
,
Oslin
D
,
Murphy
SAA
.
A “SMART” design for building individualized treatment sequences
.
Annu Rev Clin Psychol
.
2012
;
8
(
1
):
21
48
.

445.

Gusberti
FA
,
Syed
SA
,
Bacon
G
,
Grossman
N
,
Loesche
WJ
.
Puberty gingivitis in insulin-dependent diabetic children. I. Cross-sectional observations
.
J Periodontol
.
1983
;
54
(
12
):
714
720
.

446.

Stewart
CJ
,
Nelson
A
,
Campbell
MD
,
Walker
M
,
Stevenson
EJ
,
Shaw
JA
,
Cummings
SP
,
West
DJ
.
Gut microbiota of type 1 diabetes patients with good glycaemic control and high physical fitness is similar to people without diabetes: an observational study
.
Diabet Med
.
2017
;
34
(
1
):
127
134
.

447.

Burrows
MP
,
Volchkov
P
,
Kobayashi
KS
,
Chervonsky
AV
.
Microbiota regulates type 1 diabetes through Toll-like receptors
.
Proc Natl Acad Sci USA
.
2015
;
112
(
32
):
9973
9977
.

448.

Alam
C
,
Bittoun
E
,
Bhagwat
D
,
Valkonen
S
,
Saari
A
,
Jaakkola
U
,
Eerola
E
,
Huovinen
P
,
Hänninen
A
.
Effects of a germ-free environment on gut immune regulation and diabetes progression in non-obese diabetic (NOD) mice
.
Diabetologia
.
2011
;
54
(
6
):
1398
1406
.

449.

De Riva
A
,
Wållberg
M
,
Ronchi
F
,
Coulson
R
,
Sage
A
,
Thorne
L
,
Goodfellow
I
,
McCoy
KD
,
Azuma
M
,
Cooke
A
,
Busch
R
.
Regulation of type 1 diabetes development and B-cell activation in nonobese diabetic mice by early life exposure to a diabetogenic environment
.
PLoS One
.
2017
;
12
(
8
):
e0181964
.

450.

Pinto
E
,
Anselmo
M
,
Calha
M
,
Bottrill
A
,
Duarte
I
,
Andrew
PW
,
Faleiro
ML
.
The intestinal proteome of diabetic and control children is enriched with different microbial and host proteins
.
Microbiology
.
2017
;
163
(
2
):
161
174
.

451.

Roesch
LF
,
Lorca
GL
,
Casella
G
,
Giongo
A
,
Naranjo
A
,
Pionzio
AM
,
Li
N
,
Mai
V
,
Wasserfall
CH
,
Schatz
D
,
Atkinson
MA
,
Neu
J
,
Triplett
EW
.
Culture-independent identification of gut bacteria correlated with the onset of diabetes in a rat model
.
ISME J
.
2009
;
3
(
5
):
536
548
.

452.

de Groot
PF
,
Belzer
C
,
Aydin
Ö
,
Levin
E
,
Levels
JH
,
Aalvink
S
,
Boot
F
,
Holleman
F
,
van Raalte
DH
,
Scheithauer
TP
,
Simsek
S
,
Schaap
FG
,
Olde Damink
SWM
,
Roep
BO
,
Hoekstra
JB
,
de Vos
WM
,
Nieuwdorp
M
.
Distinct fecal and oral microbiota composition in human type 1 diabetes, an observational study
.
PLoS One
.
2017
;
12
(
12
):
e0188475
.

453.

Davis-Richardson
AG
,
Ardissone
AN
,
Dias
R
,
Simell
V
,
Leonard
MT
,
Kemppainen
KM
,
Drew
JC
,
Schatz
D
,
Atkinson
MA
,
Kolaczkowski
B
,
Ilonen
J
,
Knip
M
,
Toppari
J
,
Nurminen
N
,
Hyöty
H
,
Veijola
R
,
Simell
T
,
Mykkänen
J
,
Simell
O
,
Triplett
EW
.
Bacteroides dorei dominates gut microbiome prior to autoimmunity in Finnish children at high risk for type 1 diabetes
.
Front Microbiol
.
2014
;
5
:
678
.

454.

Patterson
E
,
Marques
TM
,
O’Sullivan
O
,
Fitzgerald
P
,
Fitzgerald
GF
,
Cotter
PD
,
Dinan
TG
,
Cryan
JF
,
Stanton
C
,
Ross
RP
.
Streptozotocin-induced type-1-diabetes disease onset in Sprague-Dawley rats is associated with an altered intestinal microbiota composition and decreased diversity
.
Microbiology
.
2015
;
161
(
Pt 1
):
182
193
.

455.

Henschel
AM
,
Cabrera
SM
,
Kaldunski
ML
,
Jia
S
,
Geoffrey
R
,
Roethle
MF
,
Lam
V
,
Chen
YG
,
Wang
X
,
Salzman
NH
,
Hessner
MJ
.
Modulation of the diet and gastrointestinal microbiota normalizes systemic inflammation and β-cell chemokine expression associated with autoimmune diabetes susceptibility
.
PLoS One
.
2018
;
13
(
1
):
e0190351
.

456.

Davis-Richardson
AG
,
Triplett
EW
.
A model for the role of gut bacteria in the development of autoimmunity for type 1 diabetes
.
Diabetologia
.
2015
;
58
(
7
):
1386
1393
.

457.

Jia
L
,
Shan
K
,
Pan
LL
,
Feng
N
,
Lv
Z
,
Sun
Y
,
Li
J
,
Wu
C
,
Zhang
H
,
Chen
W
,
Diana
J
,
Sun
J
,
Chen
YQ
.
Clostridium butyricum CGMCC0313.1 protects against autoimmune diabetes by modulating intestinal immune homeostasis and inducing pancreatic regulatory T cells
.
Front Immunol
.
2017
;
8
:
1345
.

458.

Needell
JC
,
Dinarello
CA
,
Ir
D
,
Robertson
CE
,
Ryan
SM
,
Kroehl
ME
,
Frank
DN
,
Zipris
D
.
Implication of the intestinal microbiome as a potential surrogate marker of immune responsiveness to experimental therapies in autoimmune diabetes
.
PLoS One
.
2017
;
12
(
3
):
e0173968
.

459.

Hansen
CH
,
Krych
L
,
Nielsen
DS
,
Vogensen
FK
,
Hansen
LH
,
Sørensen
SJ
,
Buschard
K
,
Hansen
AK
.
Early life treatment with vancomycin propagates Akkermansia muciniphila and reduces diabetes incidence in the NOD mouse
.
Diabetologia
.
2012
;
55
(
8
):
2285
2294
.

460.

Livanos
AE
,
Greiner
TU
,
Vangay
P
,
Pathmasiri
W
,
Stewart
D
,
McRitchie
S
,
Li
H
,
Chung
J
,
Sohn
J
,
Kim
S
,
Gao
Z
,
Barber
C
,
Kim
J
,
Ng
S
,
Rogers
AB
,
Sumner
S
,
Zhang
XS
,
Cadwell
K
,
Knights
D
,
Alekseyenko
A
,
Bäckhed
F
,
Blaser
MJ
.
Antibiotic-mediated gut microbiome perturbation accelerates development of type 1 diabetes in mice
.
Nat Microbiol
.
2016
;
1
(
11
):
16140
.

461.

Brown
K
,
Godovannyi
A
,
Ma
C
,
Zhang
Y
,
Ahmadi-Vand
Z
,
Dai
C
,
Gorzelak
MA
,
Chan
Y
,
Chan
JM
,
Lochner
A
,
Dutz
JP
,
Vallance
BA
,
Gibson
DL
.
Prolonged antibiotic treatment induces a diabetogenic intestinal microbiome that accelerates diabetes in NOD mice
.
ISME J
.
2016
;
10
(
2
):
321
332
.

462.

Virtanen
SM
,
Takkinen
HM
,
Nwaru
BI
,
Kaila
M
,
Ahonen
S
,
Nevalainen
J
,
Niinistö
S
,
Siljander
H
,
Simell
O
,
Ilonen
J
,
Hyöty
H
,
Veijola
R
,
Knip
M
.
Microbial exposure in infancy and subsequent appearance of type 1 diabetes mellitus-associated autoantibodies: a cohort study
.
JAMA Pediatr
.
2014
;
168
(
8
):
755
763
.

463.

Kemppainen
KM
,
Ardissone
AN
,
Davis-Richardson
AG
,
Fagen
JR
,
Gano
KA
,
León-Novelo
LG
,
Vehik
K
,
Casella
G
,
Simell
O
,
Ziegler
AG
,
Rewers
MJ
,
Lernmark
Å
,
Hagopian
W
,
She
JX
,
Krischer
JP
,
Akolkar
B
,
Schatz
DA
,
Atkinson
MA
,
Triplett
EW
;
TEDDY Study Group
.
Early childhood gut microbiomes show strong geographic differences among subjects at high risk for type 1 diabetes
.
Diabetes Care
.
2015
;
38
(
2
):
329
332
.

464.

Mejía-León
ME
,
López-Domínguez
L
,
Aguayo-Patrón
SV
,
Caire-Juvera
G
,
Calderón de la Barca
AM
.
Dietary changes and gut dysbiosis in children with type 1 diabetes
.
J Am Coll Nutr
.
2018
;
37
(
6
):
501
507
.

465.

Uusitalo
U
,
Liu
X
,
Yang
J
,
Aronsson
CA
,
Hummel
S
,
Butterworth
M
,
Lernmark
Å
,
Rewers
M
,
Hagopian
W
,
She
JX
,
Simell
O
,
Toppari
J
,
Ziegler
AG
,
Akolkar
B
,
Krischer
J
,
Norris
JM
,
Virtanen
SM
;
TEDDY Study Group
.
Association of early exposure of probiotics and islet autoimmunity in the TEDDY Study
.
JAMA Pediatr
.
2016
;
170
(
1
):
20
28
.

466.

Silverman
M
,
Kua
L
,
Tanca
A
,
Pala
M
,
Palomba
A
,
Tanes
C
,
Bittinger
K
,
Uzzau
S
,
Benoist
C
,
Mathis
D
.
Protective major histocompatibility complex allele prevents type 1 diabetes by shaping the intestinal microbiota early in ontogeny
.
Proc Natl Acad Sci USA
.
2017
;
114
(
36
):
9671
9676
.

467.

Mullaney
JA
,
Stephens
JE
,
Costello
ME
,
Fong
C
,
Geeling
BE
,
Gavin
PG
,
Wright
CM
,
Spector
TD
,
Brown
MA
,
Hamilton-Williams
EE
.
Type 1 diabetes susceptibility alleles are associated with distinct alterations in the gut microbiota [published correction appear in Microbiome. 2018;6(1):51]
.
Microbiome
.
2018
;
6
(
1
):
35
.