Abstract

Context

Genetic variants in melanocortin 3 receptor (MC3R) and melanocortin 4 receptor (MC4R) genes are strongly associated with childhood obesity.

Objective

This study aims to identify and functionally characterize MC3R and MC4R variants in an Asian cohort of children with severe early-onset obesity.

Methods

Whole-exome sequencing was performed to screen for MC3R and MC4R coding variants in 488 Asian children with severe early-onset obesity (body mass index for age ≥97th percentile). Functionality of the identified variants were determined via measurement of intracellular cyclic adenosine monophosphate (cAMP) concentrations and luciferase activity.

Results

Four MC3R and 2 MC4R heterozygous nonsynonymous rare variants were detected. There were 3 novel variants: MC3R c.151G > C (p.Val51Leu), MC4R c.127C > A (p.Gln43Lys), and MC4R c.272T > G (p.Met91Arg), and 3 previously reported variants: MC3R c.127G > A (p.Glu43Lys), MC3R c.97G > A (p.Ala33Thr), and MC3R c.437T > A (p.Ile146Asn). Both MC3R c.127G > A (p.Glu43Lys) and MC4R c.272T > G (p.Met91Arg) variants demonstrated defective downstream cAMP signaling activity. The MC4R c.127C > A (p.Gln43Lys) variant showed reduced cAMP signaling activity at low substrate concentration but the signaling activity was restored at high substrate concentration. The MC3R c.151G > C (p.Val51Leu) variant did not show a significant reduction in cAMP signaling activity compared to wild-type (WT) MC3R. Coexpression studies of the WT and variant MC3R/MC4R showed that the heterozygous variants did not exhibit dominant negative effect.

Conclusion

Our functional assays demonstrated that MC3R c.127G > A (p.Glu43Lys) and MC4R c.272T > G (p.Met91Arg) variants might predispose individuals to early-onset obesity, and further studies are needed to establish the causative effect of these variants in the pathogenesis of obesity.

The prevalence of childhood obesity has been on the rise in Singapore and has been imposing a huge burden on our healthcare infrastructure (1). Obesity is a complex, multifactorial disease involving excess body fat accumulation that increases the risk of debilitating cardiometabolic conditions that can potentially reduce quality of life and life expectancy (2). Obesity evolves from the interaction between modern “obesogenic” environment and genetic susceptibility to excessive weight gain (3). Genetic factors are especially important in the development of early-onset obesity as children have minimal exposure to extrinsic environmental factors (4).

Melanocortin 3 receptor (MC3R) and melanocortin 4 receptor (MC4R) are G protein–coupled receptors that are activated by α-melanocyte stimulating hormone (α-MSH) via stimulatory heterotrimeric G proteins leading to cyclic adenosine monophosphate (cAMP) production that regulates feeding behavior and metabolism pathways (5-10). The knockout (KO) of MC3R and MC4R genes in mice has been shown to generate an obese phenotype (11, 12). However, the obesity features differ between the MC3R KO and MC4R KO models. MC3R KO rats displayed a mild obesity phenotype characterized by hypophagia, higher fat mass, and lower lean mass (12, 13), while MC4R KO rats were shown to exhibit a severe obesity phenotype characterized by hyperphagia, hyperglycemia, and hyperinsulinemia (13).

Studies have revealed multiple mutations in the MC3R and MC4R genes that are strongly associated with early-onset obesity (14-25). MC4R mutations account for about 5% of severe obesity, which makes it the most common cause of monogenic obesity (17), whereas a fewer number of MC3R mutations have been reported to be associated with early-onset obesity (26). MC3R and MC4R mutations have been shown to impair receptor signaling through disruption of receptor expression/trafficking, alteration in substrate binding, and reduction in downstream signaling pathway related to appetite control (16, 18, 21, 22, 27-30).

Groundbreaking pharmacological research has led to the development of the small-molecule MC4R agonist setmelanotide, which has recently been approved by the US Food and Drug Administration for the treatment of obesity associated with genetic defects upstream of the MC4R pathway (31, 32). Studies have shown that setmelanotide can lead to weight loss in individuals who have MC4R mutation-driven obesity via a “chaperone effect” whereby the small-molecule MC4R agonist rescues the variant MC4R by increasing cell surface expression (33-36). Setmelanotide also activates the MC3R signaling pathway (33). The identification and functional characterization of MC3R and MC4R variants may guide the use of genetic-driven drugs, such as setmelanotide, for the treatment of obesity.

In this study, we aimed to identify and functionally characterize MC3R and MC4R variants in an Asian cohort of children with severe early-onset obesity.

Materials and Methods

Study Participants

A total of 571 children with obesity, and of Chinese and Malay ethnicity, were recruited from hospital (National University Hospital) and community settings (Singapore Health Promotion Board) under the OBesity in Singaporean Children (OBiSC) study. The recruitment criteria were 1) onset of obesity at younger than 10 years, 2) body mass index (BMI) for age greater than or equal to the 97th percentile, and 3) no syndromic causes of obesity. Family members of children with obesity were also recruited if available. Family history of obesity was obtained during the study visit interview. Standard anthropometric parameters such as weight, height, and body fat percentage were measured, and pubertal assessment was conducted. Blood samples were collected in a fasted state. The study was performed in accordance with the Declaration of Helsinki, and ethics approval was obtained from Domain Specific Review Board of National Healthcare Group, Singapore (reference No. 2015/00314). Written informed consent was obtained from all study participants. The study is registered under clinicaltrials.gov (NCT02418377).

Whole-Exome Sequencing

Genomic DNA was extracted from peripheral blood leukocytes. Sequencing libraries were constructed from 1 μg of DNA using the Ultra II DNA Library Prep Kit for Illumina (NEBNext). Exonic regions were captured genomewide by hybridization with SeqCap EZ exome v3.0 probes (Nimblegen). Libraries were assessed using a Bioanalyzer High Sensitivity DNA chip (Agilent) sequenced on a HiSeq 4000 (Illumina) platform. To maximize sequencing accuracy, each sample was sequenced to an average of 40× coverage, using 2 × 150 bp read lengths.

Paired-ended sequencing reads were aligned with the reference sequence MC3R: NM_019888.3, MC4R: NM_005912.3 using Burrows Wheeler Aligner. Variant quality score recalibration was performed in the raw variant call set and single-nucleotide variations (SNVs) with low confidence were excluded based on the following parameters: quality score normalized by depth less than 2, mapping quality less than 40, strand bias greater than 60, mapping quality bias less than 12.5, and bias in read position of alternative allele versus reference allele less than −8. High-quality common variants with a minor allele frequency greater than 5% and SNV call rate greater than 95% were used to perform sample quality control (QC) procedures in our study data set. Samples with depth of coverage less than 10, call rate less than 95%, extreme heterozygosity levels, or with first-degree relation/outliers from reported ethnicity were excluded.

Selection of Melanocortin 3 Receptor and Melanocortin 4 Receptor Genetic Variants

Each variant that passed the QC was annotated using ANNOVAR (ver20140915) (37) for determination of reference sequence gene symbol and the frequency of reference populations from the Exome Aggregation Consortium database (37). Novel (not found in Genome Aggregation Database, gnomAD) or rare nonsynonymous variants with an allele frequency of less than 0.1% in Asian population, from exonic regions of obesity susceptibility genes MC3R and MC4R, were identified among our QC data set of Chinese and Malay childhood-obesity cases, which were compared against controls consisting of 1762 Chinese and Malay normal-weight (BMI 18.5-23) Singaporean adults selected from other cohorts. Sanger sequencing was then performed to reconfirm the identity of these variants. Novel variants were submitted to ClinVar (https://www.ncbi.nlm.nih.gov/clinvar/).

Conservational Analysis and Prediction of Functional Effect of Melanocortin 3 Receptor and Melanocortin 4 Receptor Variants

Clustal Omega (https://www.ebi.ac.uk/Tools/msa/clustalo/) was used to align and compare the degree of conservation of sequence region surrounding the MC3R or MC4R variants among different species. Bioinformatics tools such as Sorting Intolerant From Tolerant (SIFT) (https://sift.bii.a-star.edu.sg/) and Polyphen-2 (http://genetics.bwh.harvard.edu/pph2/) were used to predict the possible impact of the MC3R or MC4R variants on protein function. The classification of variant function (pathogenic, likely pathogenic, uncertain significance, likely benign, and benign) was based on the American College of Medical Genetics and Genomics criteria (38).

Construction of Wild-type and Variant Melanocortin 3 Receptor and Melanocortin 4 Receptor Plasmids

The pBuDCE4.1 dual-expression vector (Thermo Fisher), which consists of 2 multiple cloning sites (MCS) under 2 constitutive promoters respectively, was used to create the MC3R or MC4R plasmids. The sequence for Renilla luciferase (internal control) was first cloned into one of the MCS of the dual-expression vectors using HindIII and BamHI restriction sites, and the wild-type (WT) MC3R or MC4R sequence was then cloned into the other MCS of the same dual-expression vector using the XhoI and BglII restriction sites to generate the MC3R or MC4R WT plasmids. The MC3R or MC4R variants were introduced into the respective MC3R or MC4R WT plasmids via site-directed mutagenesis (GenScript) to generate variant MC3R or MC4R plasmids. The sequences of all plasmids were verified by Sanger sequencing.

Measurement of Cyclic Adenosine Monophosphate Levels

Human embryonic kidney 293 (HEK293) cells were maintained in Dulbecco's Modified Eagle's Medium (Cytiva) supplemented with 10% fetal bovine serum, 100 U/mL penicillin and 100 μg/mL streptomycin (Cytiva). Cells were incubated at 37 °C with 5% CO2. The cells were grown to 90% confluent on a 6-well plate, and each well was transfected with 2 μg of either MC3R or MC4R plasmid (WT or variant plasmid) using Lipofectamine 2000 (Invitrogen). The transfected cells were incubated at 37 °C with 5% CO2 overnight for 24 hours. Thereafter, the cells were stimulated with α-MSH of differing concentrations ranging from 100 pmol/L to 10 µmol/L for 16 hours. Cells were lysed with 0.1 M HCl and assayed for cAMP levels using a cAMP competitive enzyme-linked immunosorbent assay kit (Abcam catalog No. ab65355, RRID: AB_2909598), in which absorbance was measured at OD450 nm by the Synergy 2 Microplate Reader (Biotek Instruments). The assay was performed in technical duplicates and each experiment was performed in biological triplicates. Levels of cAMP were expressed as fold increase over basal reading of nonstimulated cells.

Measurement of Dual Luciferase Activity

HEK293 cells were grown to 90% confluent on a 24-well plate, and each well was cotransfected with 0.5 μg of MC3R or MC4R plasmid (WT or variant plasmid) and 0.5 μg of pGL4.29 cAMP response element (CRE) firefly luciferase reporter vector (Promega) using Lipofectamine 2000 (Invitrogen). On binding of α-MSH to MC3R or MC4R, cAMP will be produced and the cAMP will bind to CRE to promote the expression of firefly luciferase. Concurrently, HEK293 cells were cotransfected with 0.25 μg of variant plasmids, 0.25 μg of WT plasmids and 0.5 μg of CRE reporter vector to investigate for a possible dominant-negative effect of variant MC3R or MC4R. Transfected cells were incubated overnight for 24 hours and then stimulated with α-MSH of differing concentrations ranging from 100 pmol/L to 1 µmol/L for 16 hours. The activity of variant MC3R or MC4R was determined by a dual-luciferase reporter assay (Promega) according to the manufacturer's instructions, and luciferase activity was measured using the Glomax Luminometer (Promega). The assay was performed in technical duplicates and each experiment was performed in biological triplicates. Luciferase activity was normalized against Renilla activity (internal control for transection efficiency) and expressed as fold over basal reading of nonstimulated cells.

Statistical Analysis

A 3-parameter sigmoidal dose-response curve was fitted to normalized cAMP and luciferase values from all replicates using GraphPad Prism 8 for Windows (version 8.4.3 GraphPad Software). Extra sum-of-squares F-test was used to compare the differences in EC50 and Emax.

Relative Emax was determined based on cAMP levels and luciferase activity measured at 10−5M and 10−6M α-MSH, respectively. For variant MC3R or MC4R that did not mount a response with α-MSH stimulation, no fit curve was generated, and t test (SPSS, version 27.0, IBM) was used to compare differences in Emax.

Results

Identification of Novel Melanocortin 3 Receptor and Melanocortin 4 Receptor Variants in Asian Children With Early-Onset Obesity

After sample QC, 211 patients of Malay ethnicity (43.2%) and 277 cases of Chinese ethnicity (56.8%) were available for downstream analysis. These patients were 67.6% male (32.4% female), and had a mean age of 12.7 ± 3.54 years, BMI of 133 ± 20% of 95th percentile, body fat percentage of 47.9 ± 9.93% and waist-to-hip ratio of 0.95 ± 0.07. Six heterozygous rare nonsynonymous variants (4 in MC3R and 2 in MC4R) were identified in 6 unrelated children with severe obesity. These 6 cases did not possess pathogenic variants in other candidate obesity-susceptibility genes. All identified variants were visually inspected using the integrated genomics viewer. The Variant Quality Score Log-odds (VQSLOD) for these variants ranged from 6.37 to 11.36, indicating high sequence quality, and there were good sequencing depths ranging from 12 to 156 in the samples identified to contain these variants. The 6 heterozygous rare nonsynonymous variants were reconfirmed by Sanger sequencing (Supplementary Fig. S1) (39).

We identified 1 novel MC3R variant, NM_019888.3 (MC3R): c.151G > C (p.Val51Leu), and 2 novel MC4R variants, NM_005912.3 (MC4R): c.127C > A (p.Gln43Lys) and NM_005912.3 (MC4R): c.272T > G (p.Met91Arg) in our cohort of Asian children with early-onset obesity. We also identified 3 other MC3R variants that have previously been reported: NM_019888.3 (MC3R): c.127G > A (p.Glu43Lys) is a variant of uncertain significance; NM_019888.3 (MC3R): c.97G > A (p.Ala33Thr) is a likely benign variant; and NM_019888.3 (MC3R): c.437T > A (p.Ile146Asn) is a likely pathogenic variant (21, 25, 40, 41). Characteristics and functional prediction/classification of each variant is represented in Table 1. Human sequences surrounding the MC3R and MC4R variants are highly conserved in other species (Supplementary Fig. S2) (39). The location of each variant on MC3R or MC4R proteins are shown in Supplementary Fig. S3 (39).

Table 1.

Characteristics and functional prediction/classification of MC3R and MC4R variants

SNVLocationGenotypeFrequency in obese cohortFrequency in control cohortSIFT predictionPolyphen-2 predictionAllele frequency in AsiansVariant classificationAccession No.
MC3R (NM_019888.3)
c.97G > A (p.Ala33Thr)Extracellular N-terminusHeterozygous10ToleratedBenign0.00008Likely benignrs770261378
c.127G > A (p.Glu43Lys)First transmembrane helixHeterozygous20ToleratedPossibly damaging<0.00001Uncertain significancers375750359
c.151G > C (p.Val51Leu)aFirst transmembrane helixHeterozygous10ToleratedBenignUncertain significanceSCV002540643
c.437T > A (p.Ile146Asn)Second intracellular loopHeterozygous10DamagingProbably damaging0.00018Likely pathogenicrs74315393
MC4R (NM_005912.3)
c.127C > A (p.Gln43Lys)aExtracellular N-terminusHeterozygous10ToleratedPossibly damagingUncertain significanceSCV002540644
c.272T > G (p.Met91Arg)aSecond transmembrane helixHeterozygous10DamagingProbably damagingLikely pathogenicSCV002540645
SNVLocationGenotypeFrequency in obese cohortFrequency in control cohortSIFT predictionPolyphen-2 predictionAllele frequency in AsiansVariant classificationAccession No.
MC3R (NM_019888.3)
c.97G > A (p.Ala33Thr)Extracellular N-terminusHeterozygous10ToleratedBenign0.00008Likely benignrs770261378
c.127G > A (p.Glu43Lys)First transmembrane helixHeterozygous20ToleratedPossibly damaging<0.00001Uncertain significancers375750359
c.151G > C (p.Val51Leu)aFirst transmembrane helixHeterozygous10ToleratedBenignUncertain significanceSCV002540643
c.437T > A (p.Ile146Asn)Second intracellular loopHeterozygous10DamagingProbably damaging0.00018Likely pathogenicrs74315393
MC4R (NM_005912.3)
c.127C > A (p.Gln43Lys)aExtracellular N-terminusHeterozygous10ToleratedPossibly damagingUncertain significanceSCV002540644
c.272T > G (p.Met91Arg)aSecond transmembrane helixHeterozygous10DamagingProbably damagingLikely pathogenicSCV002540645

Characteristics of each variant including location, genotype, frequency, novelty, and predicted functionality are described. Frequency refers to the number of individuals within each cohort that are found to possess the respective variant. None of the variants were detected in 1762 normal-weight adult controls. Bioinformatics tools such as Sorting Intolerant From Tolerant (SIFT) and Polyphen-2 were used to predict functional significance of these variants. The allele frequency of each variant in Asians were extracted from the Genome Aggregation Database (gnomAD). The pathogenicity of each variant was classified based on the American College of Medical Genetics and Genomics criteria.

Abbreviation: SNV, single-nucleotide variation.

aNovel variants identified in this study.

Table 1.

Characteristics and functional prediction/classification of MC3R and MC4R variants

SNVLocationGenotypeFrequency in obese cohortFrequency in control cohortSIFT predictionPolyphen-2 predictionAllele frequency in AsiansVariant classificationAccession No.
MC3R (NM_019888.3)
c.97G > A (p.Ala33Thr)Extracellular N-terminusHeterozygous10ToleratedBenign0.00008Likely benignrs770261378
c.127G > A (p.Glu43Lys)First transmembrane helixHeterozygous20ToleratedPossibly damaging<0.00001Uncertain significancers375750359
c.151G > C (p.Val51Leu)aFirst transmembrane helixHeterozygous10ToleratedBenignUncertain significanceSCV002540643
c.437T > A (p.Ile146Asn)Second intracellular loopHeterozygous10DamagingProbably damaging0.00018Likely pathogenicrs74315393
MC4R (NM_005912.3)
c.127C > A (p.Gln43Lys)aExtracellular N-terminusHeterozygous10ToleratedPossibly damagingUncertain significanceSCV002540644
c.272T > G (p.Met91Arg)aSecond transmembrane helixHeterozygous10DamagingProbably damagingLikely pathogenicSCV002540645
SNVLocationGenotypeFrequency in obese cohortFrequency in control cohortSIFT predictionPolyphen-2 predictionAllele frequency in AsiansVariant classificationAccession No.
MC3R (NM_019888.3)
c.97G > A (p.Ala33Thr)Extracellular N-terminusHeterozygous10ToleratedBenign0.00008Likely benignrs770261378
c.127G > A (p.Glu43Lys)First transmembrane helixHeterozygous20ToleratedPossibly damaging<0.00001Uncertain significancers375750359
c.151G > C (p.Val51Leu)aFirst transmembrane helixHeterozygous10ToleratedBenignUncertain significanceSCV002540643
c.437T > A (p.Ile146Asn)Second intracellular loopHeterozygous10DamagingProbably damaging0.00018Likely pathogenicrs74315393
MC4R (NM_005912.3)
c.127C > A (p.Gln43Lys)aExtracellular N-terminusHeterozygous10ToleratedPossibly damagingUncertain significanceSCV002540644
c.272T > G (p.Met91Arg)aSecond transmembrane helixHeterozygous10DamagingProbably damagingLikely pathogenicSCV002540645

Characteristics of each variant including location, genotype, frequency, novelty, and predicted functionality are described. Frequency refers to the number of individuals within each cohort that are found to possess the respective variant. None of the variants were detected in 1762 normal-weight adult controls. Bioinformatics tools such as Sorting Intolerant From Tolerant (SIFT) and Polyphen-2 were used to predict functional significance of these variants. The allele frequency of each variant in Asians were extracted from the Genome Aggregation Database (gnomAD). The pathogenicity of each variant was classified based on the American College of Medical Genetics and Genomics criteria.

Abbreviation: SNV, single-nucleotide variation.

aNovel variants identified in this study.

Clinical Characteristics and Family Pedigrees of Probands

MC3R c.97G > A (p.Ala33Thr)

This heterozygous variant was detected in a 17-year-old Chinese girl who developed obesity at age 3 years. She had a BMI of 105.6% of 95th percentile and body fat percentage of 41.2%. She had type 2 diabetes mellitus and was on metformin treatment. Her 52-year-old father was overweight, while her 45-year-old mother and 14-year-old brother were not overweight. However, DNA of family members were not available for genotyping (Fig. 1).

Family pedigrees of probands with MC3R or MC4R variants. Age (in years), race, onset of obesity (in years), genotype, body mass index (BMI), percentage (%) of 95th percentile BMI, and body fat percentage (%) are listed. Heterozygotes are half shaded. Arrows represent probands with early-onset obesity. A, Family of proband with MC3R c.97G > A (p.Ala33Thr) variant. B, Family of proband with MC3R c.127G > A (p.Glu43Lys) variant. C, Family of proband with both MC3R c.127G > A (p.Glu43Lys) and c.151G > C (p.Val51Leu) variants. D, Family of proband with MC3R c.437T > A (p.Ile146Asn) variant. E, Family of proband with MC4R c.127C > A (p.Gln43Lys) variant. F, Family of proband with MC4R c.272T > G (p.Met91Arg) variant.
Figure 1.

Family pedigrees of probands with MC3R or MC4R variants. Age (in years), race, onset of obesity (in years), genotype, body mass index (BMI), percentage (%) of 95th percentile BMI, and body fat percentage (%) are listed. Heterozygotes are half shaded. Arrows represent probands with early-onset obesity. A, Family of proband with MC3R c.97G > A (p.Ala33Thr) variant. B, Family of proband with MC3R c.127G > A (p.Glu43Lys) variant. C, Family of proband with both MC3R c.127G > A (p.Glu43Lys) and c.151G > C (p.Val51Leu) variants. D, Family of proband with MC3R c.437T > A (p.Ile146Asn) variant. E, Family of proband with MC4R c.127C > A (p.Gln43Lys) variant. F, Family of proband with MC4R c.272T > G (p.Met91Arg) variant.

MC3R c.127G > A (p.Glu43Lys)

This heterozygous variant was identified in a 7-year-old Malay boy who developed obesity at age 3 years. He had a BMI of 188.4% of 95th percentile and body fat percentage of 66.9% with no existing medical conditions. His 43-year-old mother was overweight, but the DNA of the mother was not available for genotyping (see Fig. 1).

MC3R c.151G > C (p.Val51Leu) and c.127G > A (p.Glu43Lys)

This novel heterozygous variant, MC3R c.151 G > C, was discovered in a 9-year-old Malay boy who had been obese since infancy. This boy was also found to have the MC3R c.127G > A (p.Glu43Lys) variant. He had a BMI of 160.6% of 95th percentile and body fat percentage of 66.2% with no existing medical conditions. His 41-year-old mother was found to have the heterozygous MC3R c.151G > C (p.Val51Leu) variant but not the MC3R c.127G > A (p.Glu43Lys) variant. His mother had been obese since age 7. She had a BMI of 36.3 kg/m2 and body fat percentage of 50.6%. His 14-year-old brother was overweight, while his 49-year-old father and 11-year-old sister were not overweight. However, DNA samples of the father and siblings were not available for genotyping (see Fig. 1).

MC3R c.437T > A (p.Ile146Asn)

This heterozygous variant was detected in a 14-year-old Malay girl who had been obese since age 3. She had a BMI of 170.0% of 95th percentile and body fat percentage of 68.3% with no existing medical conditions. Her 53-year-old mother who was obese with a BMI of 29.30 kg/m2 and body fat percentage of 40.4% did not possess the variant. Her father and 20-year-old sister were overweight, while her 19-year-old sister was not overweight. However, DNA samples of the father and siblings were not available for genotyping (see Fig. 1).

MC4R c.127C > A (p.Gln43Lys)

This novel heterozygous variant was discovered in a 14-year-old Malay girl who had been obese since age 1. She had a BMI of 149.8% of 95th percentile and body fat percentage of 57.3% with no existing medical conditions. Both her 41-year-old father and 39-year-old mother were obese, while her older sister and younger brother were not overweight. However, DNA of family members were not available for genotyping (see Fig. 1).

MC4R c.272T > G (p.Met91Arg)

This novel heterozygous variant was discovered in an 11-year-old Chinese boy who had been obese since age 5. He had a BMI of 107.4% of 95th percentile and body fat percentage of 50.6% with no existing medical conditions. His 49-year-old father was overweight, while his younger brother and sister were not overweight. However, DNA samples of family members were not available for genotyping (see Fig. 1).

Onset of Puberty and Stature Among Probands With Melanocortin 3 Receptor or Melanocortin 4 Receptor Variants

MC3R rare variants were found to be associated with delayed onset of puberty and reduced linear growth (42), while MC4R is reported to be involved in regulation of growth (43). Hence, we examined the onset of puberty and stature in our probands with MC3R and MC4R variants. There was no delayed onset of puberty among the female probands while all male probands are at prepubertal stage likely due to young age. We did not observe short stature among the probands (Supplementary Table S1) (39).

Signaling Activities of Variant Melanocortin 3 Receptor or Melanocortin 4 Receptor

The activities of variant MC3R or MC4R were assessed by the intracellular level of cAMP, which is a downstream messenger of the MC3R and MC4R signaling pathways. MC3R c.97G > A (p.Ala33Thr) and MC3R c.151G > C (p.Val51Leu) variants demonstrated similar signaling activities compared to MC3R WT with no significant differences in EC50 and Emax (Fig. 2A and 2C). In contrast, MC3R c.127G > A (p.Glu43Lys) and MC3R c.437T > A (p.Ile146Asn) variants resulted in a complete loss of receptor function with negligible response to increasing α-MSH concentrations (Figs. 2B and 2D). The presence of both MC3R c.127G > A (p.Glu43Lys) and MC3R c.151G > C (p.Val51Leu) variants also resulted in a complete loss of receptor function (Fig. 2E).

Figure 2.

Signaling activities of variant MC3R or MC4R. Cyclic adenosine monophosphate (cAMP) results were normalized against basal nonstimulated readings. Each data point represents the mean and SE of at least 3 independent experiments performed in technical duplicates. A, MC3R c.97G > A (p.Ala33Thr) variant; B, MC3R c.127G > A (p.Glu43Lys) variant; C, MC3R c.151G > C (p.Val51Leu) variant; D, MC3R c.437T > A (p.Ile146Asn) variant; E, MC3R c.[127G > A;151G > C] (p.[Glu43Lys;Val51Leu]) variant; F, MC4R c.127C > A (p.Gln43Lys) variant; G, MC4R c.272T > G (p.Met91Arg) variant. ^MC4R c.127C > A (p.Gln43Lys) included stimulation with 50 µM α-melanocyte stimulating hormone (α-MSH). Three-parameter sigmoidal dose-response curve was fitted to normalized cAMP values. EC50 values are presented in µM. Emax values are a measure of normalized ratios against basal readings. Data are represented as mean ± SE of at least 3 independent experiments performed in technical duplicates. The asterisk indicates a statistically significant (P < .05) difference in EC50 or Emax values when the variant MC3R or MC4R was compared against wild-type MC3R or MC4R.

The MC4R c.127C > A (p.Gln43Lys) variant demonstrated a modest reduction in receptor signaling activity at lower concentrations of α-MSH compared to WT but the function of the variant MC4R was restored to that of WT at higher concentrations of α-MSH (Fig. 2F). In contrast, MC4R c.272T > G (p.Met91Arg) resulted in a complete loss of receptor function with negligible response to increasing α-MSH concentrations (Fig. 2G).

Figure 3.

Dominant negative effect of variant MC3R or MC4R. Luciferase levels were normalized against the internal control, Renilla levels, to account for variation in transfection efficiency. Each data point represents the mean and SE of at least 3 independent experiments performed in technical duplicates. A, MC3R c.97G > A (p.Ala33Thr) variant; B, MC3R c.127G > A (p.Glu43Lys) variant; C, MC3R c.151G > C (p.Val51Leu) variant; D, MC3R c.437T > A (p.Ile146Asn) variant; E, MC3R c.[127G > A;151G >C] (p.[Glu43Lys;Val51Leu]) variant; F, MC4R c.127C > A (p.Gln43Lys) variant; G, MC4R c.272T > G (p.Met91Arg) variant. Three-parameter sigmoidal dose-response curve was fitted to normalized luciferase values. EC50 values are presented in nM. Emax values are a measure of normalized ratios against basal readings. Data are represented as mean ± SE of at least 3 independent experiments performed in duplicates. The asterisk indicates a statistically significant (P < .05) difference in EC50 or Emax values when comparing among the wild-type, heterozygous variant, and homozygous variant MC3R or MC4R.

Dominant Negative Effect of Variant Melanocortin 3 Receptor or Melanocortin 4 Receptor

The downstream signaling activities of the variant MC3R or MC4R were also assessed by cAMP-driven luciferase activity. The luciferase activities of the variant MC3R or MC4R were similar to that demonstrated in the cAMP assay studies. There was no significant difference in luciferase activities of MC3R c.97G > A (p.Ala33Thr) and MC3R c.151G > C (p.Val51Leu) compared to MC3R WT (Fig. 3A and 3C). MC3R c.127G > A (p.Glu43Lys), MC3R c.437T > A (p.Ile146Asn), and MC3R c.[127G > A;151G > C] (p.[Glu43Lys;Val51Leu]) showed significantly reduced luciferase activity compared to MC3R WT (Fig. 3B, 3D, and 3E). MC4R c.127C > A (p.Gln43Lys) demonstrated an impaired luciferase activity at lower α-MSH concentrations but had a similar Emax compared to MC4R WT at higher α-MSH concentrations (Fig. 3F). MC4R c.272T > G (p.Met91Arg) demonstrated minimal luciferase activity despite increasing concentrations of α-MSH (Fig. 3G).

In addition, the cAMP-driven luciferase activity was used to investigate the possible dominant-negative effect of the heterozygous MC3R or MC4R variants on the receptor function. The presence of both WT and variant alleles resulted in luciferase activity that is higher than the variant allele but lower than the WT allele. These variants did not exhibit a dominant-negative effect (see Fig. 3).

Discussion

We identified 4 MC3R and 2 MC4R heterozygous variants in a cohort of Asian children with severe obesity. The 6 variants were found in 6 unrelated children with severe obesity, and the variants were absent in 1762 normal-weight adult controls. Of the 6 variants, there were 3 novel variants: MC3R c.151G > C (p.Val51Leu), MC4R c.127C > G (p.Gln43Lys), and MC4R c.272T > G (p.Met91Arg), and 3 variants that have previously been reported: MC3R c.127G > A (p.Glu43Lys) of uncertain significance, c.97G > A (p.Ala33Thr) of likely benign function, and MC3R c.437T > A (p.Ile146Asn) of likely pathogenic function (21, 25).

A 9-year-old Malay boy was found to have both MC3R c.151G > C (p.Val51Leu) and MC3R c.127G > A (p.Glu43Lys) variants. Although the novel MC3R c.151G > C (p.Val51Leu) variant did not affect downstream receptor signaling activity, the previously reported MC3R c.127G > A (p.Glu43Lys) variant of uncertain significance was found to reduce downstream receptor signaling activity. Coexpression of both variants resulted in decreased receptor signaling activity similar to that of the MC3R c.127G > A (p.Glu43Lys) variant alone. This suggests that the boy's obese phenotype was likely attributed to the MC3R c.127G > A (p.Glu43Lys) variant. The boy likely inherited the MC3R c.151G > C (p.Val51Leu) variant from his mother who was obese and had the MC3R c.151G > C (p.Val51Leu) variant but not the MC3R c.127G > A (p.Glu43Lys) variant. His mother's obese phenotype would not have been contributed by the MC3R c.151G > C (p.Val51Leu) variant.

The MC3R c.127G > A (p.Glu43Lys) variant is located on a highly conserved residue in the first transmembrane helix that is critical for ligand binding, which supports its possible pathogenic function (44). Our analysis also showed that the site of the MC3R variant is conserved among all the 8 tested species (human, chimpanzee, monkey, pig, dog, mouse, chicken, and zebrafish). In silico functional prediction programs such as SIFT predicted that the MC3R c.127G > A (p.Glu43Lys) variant would be tolerated but our in vitro experiments demonstrated pathogenic function with significant reduction in MC3R signaling activity. This highlights the importance of in vitro experiments in functionally characterizing novel variants (45).

MC3R c.97G > A (p.Ala33Thr) resides in the N-terminus, which has been shown to have a role in cell surface expression of the receptor (21). Lee et al (25) first discovered the MC3R c.97G > A (p.Ala33Thr) variant and reported that there was a significant, modest reduction in receptor signaling activity with normal cell surface expression and no dominant-negative effect of the variant. Subsequently, Yang and Tao (21) showed that this variant resulted in a nonsignificant slight reduction in signaling activity with normal ligand binding and total cell surface expression. In the present study, we revalidated the function of the MC3R c.97G > A (p.Ala33Thr) variant and found that it caused a slight reduction in receptor signaling activity compared to the WT MC3R receptor, albeit the difference was not statistically significant. We also showed that the MC3R c.97G > A (p.Ala33Thr) variant did not exert a dominant-negative effect. The lack of a significant difference as reported by Yang and Tao (21) may be attributed to the variation between biological triplicates.

MC3R c.437T > A (p.Ile146Asn), which is in the second intracellular loop that is vital for G protein–coupled receptor activation, is a pathogenic variant that leads to complete loss of protein function (25, 40). While Lee et al and Tao et al report an absence of dominant-negative effect (21, 25, 40), Rached et al (41) demonstrated a possible dominant-negative effect. We validated previous results showing that the MC3R c.437T > A (p.Ile146Asn) variant led to a loss of receptor function and did not exert a dominant-negative effect (25, 40).

A recent study by Lam et al (42) reported that rare and common MC3R variants were associated with short stature and delayed puberty. However, our children with rare MC3R variants did not have short stature or delayed puberty. The lack of significant associations between rare MC3R variants and short stature or delayed puberty in our study may be attributed to the boys who possessed the rare MC3R variants being young and at the prepubertal stage, and our small sample size. A longitudinal study of children with and without obesity will be required to examine the association between MC3R variants and childhood growth/puberty.

The novel MC4R c.272T > G (p.Met91Arg) variant resulted in a complete loss of receptor function with no downstream signaling activity. This variant resides in the second transmembrane helix, which has previously been shown to be critical for ligand binding, and conformational change in the structure of the second transmembrane helix may significantly reduce ligand binding affinity and decimate cAMP production (46).

The MC4R c.127C > A (p.Gln43Lys) variant is located on the N-terminus of the MC4R protein, which has been shown to be vital for constitutive receptor signaling activity (47). We showed that the MC4R c.127C > A (p.Gln43Lys) variant had significantly higher EC50 compared to WT, but similar Emax compared to WT. The MC4R c.127C > A (p.Gln43Lys) variant may cause a decrease in binding affinity between ligand and receptor, and the binding affinity is restored with higher ligand concentration.

Our study is limited by the lack of complete family genotype data, hence we were unable to examine the variant segregation with the obese phenotype.

In conclusion, our functional assays demonstrated that the MC3R c.127G > A (p.Glu43Lys) and MC4R c.272T > G (p.Met91Arg) variants might predispose individuals to early-onset obesity, and further studies are needed to establish the causative effect of these variants in the pathogenesis of obesity.

Funding

This work was supported by National Medical Research Council (NMRC), Singapore (NMRC/CIRG/1407/2014).

Disclosures

The authors declare no conflict of interest.

Data Availability

Original data generated and analyzed during this study are included in this published article or in the data repositories listed in “References.”

References

1

Dehghan
M
,
Akhtar-Danesh
N
,
Merchant
AT
.
Childhood obesity, prevalence and prevention
.
Nutr J
.
2005
;
4
(
1
):
24
.

2

Haslam
DW
,
James
WP
.
Obesity
.
Lancet
.
2005
;
366
(
9492
):
1197
1209
.

3

Bouchard
C
.
Current understanding of the etiology of obesity: genetic and nongenetic factors
.
Am J Clin Nutr
.
1991
;
53
(
6
):
1561S
1565S
.

4

Stunkard
AJ
,
Sorensen
TI
,
Hanis
C
, et al.
An adoption study of human obesity
.
N Engl J Med
.
1986
;
314
(
4
):
193
198
.

5

Girardet
C
,
Begriche
K
,
Ptitsyn
A
,
Koza
RA
,
Butler
AA
.
Unravelling the mysterious roles of melanocortin-3 receptors in metabolic homeostasis and obesity using mouse genetics
.
Int J Obes Suppl
.
2014
;
4
(
S1
):
S37
S44
.

6

Begriche
K
,
Levasseur
PR
,
Zhang
J
, et al.
Genetic dissection of the functions of the melanocortin-3 receptor, a seven-transmembrane G-protein-coupled receptor, suggests roles for central and peripheral receptors in energy homeostasis
.
J Biol Chem
.
2011
;
286
(
47
):
40771
40781
.

7

Gantz
I
,
Miwa
H
,
Konda
Y
, et al.
Molecular cloning, expression, and gene localization of a fourth melanocortin receptor
.
J Biol Chem
.
1993
;
268
(
20
):
15174
15179
.

8

Mountjoy
KG
,
Mortrud
MT
,
Low
MJ
,
Simerly
RB
,
Cone
RD
.
Localization of the melanocortin-4 receptor (MC4-R) in neuroendocrine and autonomic control circuits in the brain
.
Mol Endocrinol
.
1994
;
8
(
10
):
1298
1308
.

9

Tao
YX
.
The melanocortin-4 receptor: physiology, pharmacology, and pathophysiology
.
Endocr Rev
.
2010
;
31
(
4
):
506
543
.

10

Adan
RA
,
Tiesjema
B
,
Hillebrand
JJ
,
la Fleur
SE
,
Kas
MJ
,
de Krom
M
.
The MC4 receptor and control of appetite
.
Br J Pharmacol
.
2006
;
149
(
7
):
815
827
.

11

Huszar
D
,
Lynch
CA
,
Fairchild-Huntress
V
, et al.
Targeted disruption of the melanocortin-4 receptor results in obesity in mice
.
Cell
.
1997
;
88
(
1
):
131
141
.

12

Chen
AS
,
Marsh
DJ
,
Trumbauer
ME
, et al.
Inactivation of the mouse melanocortin-3 receptor results in increased fat mass and reduced lean body mass
.
Nat Genet
.
2000
;
26
(
1
):
97
102
.

13

You
P
,
Hu
H
,
Chen
Y
, et al.
Effects of melanocortin 3 and 4 receptor deficiency on energy homeostasis in rats
.
Sci Rep
.
2016
;
6
(
1
):
34938
.

14

Vaisse
C
,
Clement
K
,
Guy-Grand
B
,
Froguel
P
.
A frameshift mutation in human MC4R is associated with a dominant form of obesity
.
Nat Genet
.
1998
;
20
(
2
):
113
114
.

15

Yeo
GS
,
Farooqi
IS
,
Aminian
S
,
Halsall
DJ
,
Stanhope
RG
,
O'Rahilly
S
.
A frameshift mutation in MC4R associated with dominantly inherited human obesity
.
Nat Genet
.
1998
;
20
(
2
):
111
112
.

16

Tan
K
,
Pogozheva
ID
,
Yeo
GS
, et al.
Functional characterization and structural modeling of obesity associated mutations in the melanocortin 4 receptor
.
Endocrinology
.
2009
;
150
(
1
):
114
125
.

17

Farooqi
IS
,
Keogh
JM
,
Yeo
GS
,
Lank
EJ
,
Cheetham
T
,
O'Rahilly
S
.
Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene
.
N Engl J Med
.
2003
;
348
(
12
):
1085
1095
.

18

Yeo
GS
,
Lank
EJ
,
Farooqi
IS
,
Keogh
J
,
Challis
BG
,
O'Rahilly
S
.
Mutations in the human melanocortin-4 receptor gene associated with severe familial obesity disrupts receptor function through multiple molecular mechanisms
.
Hum Mol Genet
.
2003
;
12
(
5
):
561
574
.

19

Vaisse
C
,
Clement
K
,
Durand
E
,
Hercberg
S
,
Guy-Grand
B
,
Froguel
P
.
Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity
.
J Clin Invest
.
2000
;
106
(
2
):
253
262
.

20

Hinney
A
,
Bettecken
T
,
Tarnow
P
, et al.
Prevalence, spectrum, and functional characterization of melanocortin-4 receptor gene mutations in a representative population-based sample and obese adults from Germany
.
J Clin Endocrinol Metab
.
2006
;
91
(
5
):
1761
1769
.

21

Yang
F
,
Tao
YX
.
Functional characterization of nine novel naturally occurring human melanocortin-3 receptor mutations
.
Biochim Biophys Acta
.
2012
;
1822
(
11
):
1752
1761
.

22

Tao
YX
.
Functional characterization of novel melanocortin-3 receptor mutations identified from obese subjects
.
Biochim Biophys Acta
.
2007
;
1772
(
10
):
1167
1174
.

23

Wang
W
,
Lin
YJ
,
Chen
ZX
,
Guo
DY
.
Identification and characterization of two novel melanocortin-3 receptor mutations in Chinese obese individuals
.
Biochim Biophys Acta Mol Basis Dis
.
2021
;
1867
(
6
):
166107
.

24

Demidowich
AP
,
Jun
JY
,
Yanovski
JA
.
Polymorphisms and mutations in the melanocortin-3 receptor and their relation to human obesity
.
Biochim Biophys Acta Mol Basis Dis
.
2017
;
1863
(
10
):
2468
2476
.

25

Lee
YS
,
Poh
LK
,
Kek
BL
,
Loke
KY
.
The role of melanocortin 3 receptor gene in childhood obesity
.
Diabetes
.
2007
;
56
(
10
):
2622
2630
.

26

Ehtesham
S
,
Qasim
A
,
Meyre
D
.
Loss-of-function mutations in the melanocortin-3 receptor gene confer risk for human obesity: A systematic review and meta-analysis
.
Obes Rev
.
2019
;
20
(
8
):
1085
1092
.

27

Lubrano-Berthelier
C
,
Dubern
B
,
Lacorte
JM
, et al.
Melanocortin 4 receptor mutations in a large cohort of severely obese adults: prevalence, functional classification, genotype-phenotype relationship, and lack of association with binge eating
.
J Clin Endocrinol Metab
.
2006
;
91
(
5
):
1811
1818
.

28

Nijenhuis
WA
,
Garner
KM
,
van Rozen
RJ
,
Adan
RA
.
Poor cell surface expression of human melanocortin-4 receptor mutations associated with obesity
.
J Biol Chem
.
2003
;
278
(
25
):
22939
22945
.

29

Tao
YX
.
Molecular mechanisms of the neural melanocortin receptor dysfunction in severe early onset obesity
.
Mol Cell Endocrinol
.
2005
;
239
(
1-2
):
1
14
.

30

Xiang
Z
,
Litherland
SA
,
Sorensen
NB
, et al.
Pharmacological characterization of 40 human melanocortin-4 receptor polymorphisms with the endogenous proopiomelanocortin-derived agonists and the agouti-related protein (AGRP) antagonist
.
Biochemistry
.
2006
;
45
(
23
):
7277
7288
.

31

Clement
K
,
van den Akker
E
,
Argente
J
, et al.
Efficacy and safety of setmelanotide, an MC4R agonist, in individuals with severe obesity due to LEPR or POMC deficiency: single-arm, open-label, multicentre, phase 3 trials
.
Lancet Diabetes Endocrinol
.
2020
;
8
(
12
):
960
970
.

32

Administration UFaD
.
FDA approves first treatment for weight management for people with certain rare genetic conditions. Vol 20222020
.

33

Collet
TH
,
Dubern
B
,
Mokrosinski
J
, et al.
Evaluation of a melanocortin-4 receptor (MC4R) agonist (Setmelanotide) in MC4R deficiency
.
Mol Metab
.
2017
;
6
(
10
):
1321
1329
.

34

René
P
,
Le Gouill
C
,
Pogozheva
ID
, et al.
Pharmacological chaperones restore function to MC4R mutants responsible for severe early-onset obesity
.
J Pharmacol Exp Ther
.
2010
;
335
(
3
):
520
532
.

35

Meimaridou
E
,
Gooljar
SB
,
Ramnarace
N
,
Anthonypillai
L
,
Clark
AJ
,
Chapple
JP
.
The cytosolic chaperone Hsc70 promotes traffic to the cell surface of intracellular retained melanocortin-4 receptor mutants
.
Mol Endocrinol
.
2011
;
25
(
9
):
1650
1660
.

36

Granell
S
,
Mohammad
S
,
Ramanagoudr-Bhojappa
R
,
Baldini
G
.
Obesity-linked variants of melanocortin-4 receptor are misfolded in the endoplasmic reticulum and can be rescued to the cell surface by a chemical chaperone
.
Mol Endocrinol
.
2010
;
24
(
9
):
1805
1821
.

37

Wang
K
,
Li
M
,
Hakonarson
H
.
ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data
.
Nucleic Acids Res
.
2010
;
38
(
16
):
e164
.

38

Richards
S
,
Aziz
N
,
Bale
S
, et al.
Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American college of medical genetics and genomics and the association for molecular pathology
.
Genet Med
.
2015
;
17
(
5
):
405
424
.

39

SG Ong
RD
,
R
Dorajoo
,
J-J
Liu
,
AA
Sng
,
YS
Lee
,
DSQ
Ooi
.
Data from: Novel melanocortin-3 and −4 receptor functional variants in Asian children with severe obesity. Scholarbank@NUS 2023 2023;Deposited 22nd August 2023. https://doi.org/10.25540/0JXP-5FP8
.

40

Tao
YX
,
Segaloff
DL
.
Functional characterization of melanocortin-3 receptor variants identify a loss-of-function mutation involving an amino acid critical for G protein-coupled receptor activation
.
J Clin Endocrinol Metab
.
2004
;
89
(
8
):
3936
3942
.

41

Rached
M
,
Buronfosse
A
,
Begeot
M
,
Penhoat
A
.
Inactivation and intracellular retention of the human I183N mutated melanocortin 3 receptor associated with obesity
.
Biochim Biophys Acta
.
2004
;
1689
(
3
):
229
234
.

42

Lam
BYH
,
Williamson
A
,
Finer
S
, et al.
MC3R Links nutritional state to childhood growth and the timing of puberty
.
Nature
.
2021
;
599
(
7885
):
436
441
.

43

Liu
R
,
Kinoshita
M
,
Adolfi
MC
,
Schartl
M
.
Analysis of the role of the Mc4r system in development, growth, and puberty of medaka
.
Front Endocrinol (Lausanne)
.
2019
;
10
:
213
.

44

Wang
SX
,
Fan
ZC
,
Tao
YX
.
Functions of acidic transmembrane residues in human melanocortin-3 receptor binding and activation
.
Biochem Pharmacol
.
2008
;
76
(
4
):
520
530
.

45

Mencarelli
M
,
Dubern
B
,
Alili
R
, et al.
Rare melanocortin-3 receptor mutations with in vitro functional consequences are associated with human obesity
.
Hum Mol Genet
.
2011
;
20
(
2
):
392
399
.

46

Yang
YK
,
Fong
TM
,
Dickinson
CJ
, et al.
Molecular determinants of ligand binding to the human melanocortin-4 receptor
.
Biochemistry
.
2000
;
39
(
48
):
14900
14911
.

47

Srinivasan
S
,
Lubrano-Berthelier
C
,
Govaerts
C
, et al.
Constitutive activity of the melanocortin-4 receptor is maintained by its N-terminal domain and plays a role in energy homeostasis in humans
.
J Clin Invest
.
2004
;
114
(
8
):
1158
1164
.

Abbreviations

     
  • α-MSH

    α-melanocyte stimulating hormone

  •  
  • BMI

    body mass index

  •  
  • cAMP

    cyclic adenosine monophosphate

  •  
  • CRE

    cAMP response element

  •  
  • KO

    knockout

  •  
  • MC3R

    melanocortin 3 receptor

  •  
  • MC4R

    melanocortin 4 receptor

  •  
  • MCS

    multiple cloning sites

  •  
  • QC

    quality control

  •  
  • SIFT

    Sorting Intolerant From Tolerant

  •  
  • SNV

    single-nucleotide variation

  •  
  • WT

    wild-type

Author notes

Siong Gim Ong and Roghayeh Dehghan are joint first authors of this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/pages/standard-publication-reuse-rights)