Abstract

Pathogenic microorganisms can infect a variety of niches in the human body. During infection, these microbes can only persist if they adapt adequately to the dynamic host environment and the stresses imposed by the immune system. While viruses entirely rely on host cells to replicate, bacteria and fungi use their pathogenicity mechanisms for the acquisition of essential nutrients that lie under host restriction. An inappropriate deployment of pathogenicity mechanisms will alert host defence mechanisms that aim to eradicate the pathogen. Thus, these adaptations require tight regulation to guarantee nutritional access without eliciting strong immune activation. To work efficiently, the immune system relies on a complex signalling network, involving a myriad of immune mediators, some of which are quite directly associated with imminent danger for the pathogen. To manipulate the host immune system, viruses have evolved cytokine receptors and viral cytokines. However, among bacteria and fungi, selected pathogens have evolved the capacity to use these inflammatory response-specific signals to regulate their pathogenicity. In this review, we explore how bacterial and fungal pathogens can sense the immune system and use adaptive pathogenicity strategies to evade and escape host defence to ensure their persistence in the host.

Introduction

Despite improvements in sanitation, access to health care, and development of vaccines and antimicrobial therapies, infectious diseases remain a major cause of mortality and morbidity globally (Baker et al. 2022, Shinu et al. 2022). Especially in developing countries, these diseases remain an important cause of death (Baker et al. 2022). Yet, modern healthcare practice involves invasive procedures, indwelling medical devices, prosthetics, and immune-compromising treatments, which clear the path for opportunistic pathogens (Clark and Drummond 2019, Lopes and Lionakis 2022, Kreitmann et al. 2024). Further, demographic and climate changes contribute to the rise and spread of new infectious disease outbreaks (Baker et al. 2022). The increasing incidence of antimicrobial resistance and poorly filled drug-development pipelines make infectious diseases pose again an important threat to global health (Shinu et al. 2022). Understanding the adaptive mechanisms that allow microbial pathogens to thrive and cause disease, and the consequences of these strategies on the host–pathogen interplay during infection, can help to identify new therapeutic targets, as well as biomarkers that could aid in the detection and early diagnosis of infection.

While viruses rely on the infection of host cells and spread from host to host, bacteria and fungi can co-exist in the human body as commensals. Under certain circumstances, some microbes can undergo a commensal-to-pathogen shift and act as opportunistic pathogens. Alternatively, bacteria and fungi can also originate from the environment before infecting susceptible individuals. Several stressors in the environment such as extreme pH, temperature, osmolarity, UV light, and nutrient competition selected traits that help microbes to also overcome challenging conditions within the host (Lange et al. 2023, Huang et al. 2024). This process, known as the environmental virulence school, may explain the evolution of immune evasion mechanisms through competition with other environmental microbes and the development of defence mechanisms against predatory amoebae or nematodes in the environment (Steenbergen et al. 2001, Mylonakis 2002, Casadevall 2008, Bliska and Casadevall 2009). Mechanisms that fulfil important functions in the environmental niche can confer fitness benefits when infecting humans. This functional duality is referred to as ‘ready-made’ virulence (Casadevall 2003). Despite the imminent danger of predation and the need to evade it, by far not all microorganisms that developed effective countermeasures are human pathogens. A decisive criterion for pathogenicity in humans is the ability to grow at human body temperature. Acclimatization to elevated temperatures in some microbes might have arisen through thermophilic adaptation to the heat resulting from the fermentation of decaying biomass (Tekaia 2005). Moreover, the increased selective pressure to tolerate higher temperatures posed by global warming is believed to drive the emergence of certain environmental microbes as pathogens, such as the fungi Candida auris and Rhodosporidiobolus fluvialis (Casadevall 2020, 2021, Huang et al. 2024).

In contrast to the environmental virulence school, immune evasion strategies acquired during commensalism define the commensal virulence school (Hube 2009, Lange et al. 2023). Symbiotic host–microbe dynamics promoted their coevolution and regulatory mechanisms leading to a host immune balance (Nenciarini et al. 2024). However, disruption of this fine equilibrium could induce the transition from the commensal to a pathogenic status. To traverse through the commensal virulence school (Hube 2009) or exist as an obligate pathogen, adaptation to body temperature is a fundamental prerequisite. Additionally, the establishment of pathogenicity in the host niche requires the ability to compete with other (commensal) microbes, stressing the protective role of a healthy microbiome (Kamada et al. 2012). However, in parallel to fiercely competing with each other, host-associated microbes are under constant surveillance by the immune system (Neutra et al. 1996, Vallon-Eberhard et al. 2006). The continuous interplay with the host and the selective pressure imposed by the immune system foster the co-evolution of commensals and obligate pathogens with the host. In contrast to environmental pathogens, host-associated microbial pathogens rely on their host niches for successful replication and survival (Hube 2009).

Nutritional immunity is one of the first host defence lines that controls the microbial community, and is an illustrative example of the arms race between host and pathogen (Hood 2012, Núñez et al. 2018). This comprises an active restriction of access to essential nutrients like metal ions such as iron, zinc, or manganese by the host (Hood 2012, Núñez et al. 2018). A common microbial strategy to overcome nutritional immunity is the use of chelators to scavenge host-bound metal ions, such as siderophores, which can acquire iron bound to host proteins such as lactoferrin or transferrin (Hood 2012). Another strategy to acquire essential nutrients is by releasing them through damaging host cells (Almeida 2008, 2009, Citiulo 2012, Spaan et al. 2015, Martins et al. 2016, Ristow and Welch 2016, Núñez et al. 2018). This process has to be tightly regulated by the pathogen, as attacking the host inevitably causes the release of damage-associated molecular patterns (DAMPs) and alarmins that activate the immune response (Caffrey 2016). Employing this nutrient-acquisition strategy is a risky gamble for the pathogen, as it marks a turning point in the host–microbe relationship. The adaptations required for a microbe to persist in the host will drastically change after immune activation is elicited. The alarmins and DAMPs released by damaged host cells as well as recognition of the invading pathogen by tissue-resident immune cells will induce further leukocyte recruitment to the site of infection, and will activate adaptive immune responses at later stages (Pittman 2013, Bertheloot 2017). At this point, microbes face a rapidly developing hostile environment with an arsenal of dangerous host defence mechanisms, including reactive oxygen/nitrogen species, proteases, acidic intracellular environments, antimicrobial peptides, and neutralizing antibodies (Bevins and Salzman 2011, Frick 2011, Pauwels 2017, Lu 2018).

To persist after immune activation, a pathogen will need to apply different strategies: (i) hide from incoming immune cells, (ii) withstand the immune response by increasing its resistance to the hostile environment, or (iii) interfere with immune activation.

Due to the essential relationship of viruses with their human hosts, co-evolution has made them masters of immunomodulation and immune evasion (Alcami and Koszinowski 2000). During infection, viruses directly modulate inflammatory cell death pathways, cytokine synthesis, activation, and signal transduction (Xiang and Moss 1999, Alcami and Koszinowski 2000, Born et al. 2000, Kotwal 2000, Smith et al. 2000, Reading and Smith 2003, Parnian et al. 2024, Wu et al. 2024). Strikingly, viral evolution has numerous times achieved this by molecular mimicry of host cytokine receptors and cytokines, allowing them to efficiently interfere with cytokine signalling as well as control immune responses (Alcami and Koszinowski 2000, Kotwal 2000, Alvarez-de Miranda et al. 2021).

Via convergent evolutionary processes, bacterial and fungal pathogens have developed various strategies to cope with immune-imposed stresses, and even adapted to a lifestyle within professional immune cells (Hmama et al. 2015, Ray and Rappleye 2019, Montano et al. 2022). Thus, a timely induction of the necessary stress and evasion responses requires precise sensing of the current environment and its potential threats. In line with the widespread capacity of viruses to interfere with immune activation, there is evidence that some bacterial and fungal pathogens have similarly evolved efficient strategies to interact with immune mediators. In this review, we discuss how bacterial and fungal pathogens can sense different states of immune activation within the mammalian host and, in consequence, integrate these signals to adaptively engage pathogenicity under adequate circumstances to ensure their persistence during infection.

Adaptation versus adaptive prediction

When microbes exist in a commensal relationship with their host, the environment is relatively stable or follows patterns of systematic fluctuation (‘microbe-host homeostasis’). However, this changes drastically when microbes apply their pathogenicity mechanisms and the host’s immune defences are triggered. Disruption of the commensal status can also be associated with the exchange of one host niche for another, which is accompanied by drastic changes in the environmental conditions that the microbe encounters. Exemplary, the opportunistic pathogenic yeast Candida albicans lyses host cells using its toxin candidalysin (Moyes et al. 2016) and thereby can translocate through the intestinal epithelium (Allert et al. 2018) from the intestine (its commensal niche) to the bloodstream (Kullberg and Arendrup 2015, Kumamoto et al. 2020). Such stressful scenarios within the host demand rapid sensing of environmental changes by pathogens in order to survive and become better suited to their new niche, also known as adaptation (Peck and Waxman 2018). These environmental changes often follow regular sequential or cyclic patterns. By sensing specific markers of these sequential or cyclic patterns, pathogens can ‘predict’ upcoming environmental changes and challenges (Cao and Goodrich-Blair 2017). Microbes capable of such adaptive prediction have a fitness benefit even before encountering the actual threat (Mitchell et al. 2009, Brunke and Hube 2014, Pradhan et al. 2019). These molecular mechanisms of adaptive prediction are driven by evolutionary coupling and reconnecting of stress-response pathways (Brown et al. 2019). Responses to two unrelated stimuli or stresses can be rearranged and connected on a microbial evolutionary timescale, which can be exemplified for specific microbes. Escherichia coli, a commensal bacterium in the human intestine, induces the expression of genes required for maltose metabolism upon exposure to the unrelated metabolite lactose (Savageau 1998). This response is in line with lactose being the first carbon source that E. coli encounters in the upper digestive tract, followed by maltose in the lower digestive tract (Savageau 1998). The predictable pattern in nutrient availability has evolutionarily favoured E. coli variants that express genes for maltose metabolism upon exposure to lactose. This adaptive prediction could be uncoupled by continuously growing E. coli on lactose, without subsequent exposure to maltose (Mitchell et al. 2009). This suggests that E. coli acquired its predictive adaptive trait due to the ‘predictable’ consecutive environmental conditions and the fitness benefits of faster adaption. Nevertheless, when no fitness benefit is achieved, predictive adaptive traits can be lost, likely due to the high costs of sustaining the response (Mitchell et al. 2009).

Besides bacteria, adaptive prediction has also been reported throughout the fungal kingdom from environmental to commensal and human pathogenic fungi. Exemplary, the fungal genetics model organism Neurospora crassa responds to the monosaccharide l-rhamnose not only with upregulation of a broad pectin degradation machinery, but the corresponding master regulator PDR1 also promotes gene expression for the utilization of the hemicellulose arabinan (Thieme et al. 2017). Thereby, the fungus not only expects other sugars present in the polysaccharide pectin, where l-rhamnose is most likely to be encountered but also readies itself to degrade other components of a plant cell wall, where it would naturally find pectin. A later study corroborated and extended this observation of cross-induction of small saccharides for utilization of polysaccharides beyond their common parent structure to a general concept in fungal plant cell wall degradation (Wu et al. 2020). As mentioned above, the opportunistic pathogenic yeast C. albicans can cause systemic infections when it translocates from its commensal niche (Kumamoto et al. 2020). During this process, C. albicans transits from the gut where glucose levels are low, into the bloodstream where higher glucose levels are encountered (Fig. 1). The exposure of C. albicans to blood glucose concentrations increases oxidative and cationic stress resistance via the upregulation of the genes hog1 and cap1, a response that is not required for glucose metabolism (Rodaki et al. 2009). Nevertheless, this response is beneficial for confrontation with immune cells, which can take place when the fungus enters the bloodstream and disseminates to vital organs (Lionakis et al. 2011, Kammer et al. 2020), as both Hog1 and Cap1 proteins are known to regulate the protection against oxidative stresses (Alonso-Monge et al. 2003). Immune cells such as neutrophils eliminate the fungus via oxidative burst, neutrophil extracellular traps formation, or the influx of cations into phagocytic vacuoles (Lee et al. 2003b, Naglik et al. 2017, Hopke et al. 2020). Moreover, bloodstream glucose concentration also induces filamentous growth of C. albicans, which is a crucial feature of its virulence (Alves et al. 2020) and further complicates clearance by the immune system (Austermeier et al. 2020). Such predictive adaptations and pathogenicity programs allow C. albicans to anticipate potential threats, thereby increasing its survival chances and fitness. Besides glucose, other carbohydrates can drive adaptation, mostly leading to immune evasion through cell wall remodelling (reviewed in Hopke et al. 2018, Childers et al. 2020).

Comparison of adaptation and adaptive prediction on the example of C. albicans and glucose (Rodaki et al. 2009). To cause systemic infection, C. albicans needs to translocate from the intestine, a commensal niche, into the bloodstream. There C. albicans encounters a new environment with stressors imposed by the host. The survival of the fungus depends on its acquired adaptation strategies. Adaptation (left panel): (i) Upon entering the bloodstream, C. albicans senses a higher glucose concentration compared to its previous niche. This leads to the expression of genes involved in metabolic remodelling (yellow arrow) to utilize the carbon source efficiently. (ii) Candida albicans will further encounter immune cells, which are perceived as a direct threat, resulting in the expression of genes involved in its defence mechanisms (blue/diagonal-patterned arrow). (iii) The pathogenic yeast was unsuccessful in rapidly adapting to the threat, giving the immune cells the advantage to overcome and eliminate the fungus. In this scenario, adaptation to environmental signals and adaptation to imposed stress are not linked to each other, potentially leading to delayed reactions. Adaptive prediction (right panel): (i) Upon entering the bloodstream, C. albicans senses the elevated glucose concentration and recognizes it not only as a nutrient source but as a potential precursor. Consequently, genes related to glucose metabolism are upregulated along with those involved in oxidative and cationic stress resistance (yellow - blue/diagonal-patterned arrow). (ii) This co-induced response prepares for the upcoming threat imposed by the immune system, thereby providing a proactive advantage. (iii) The mechanism of adaptive prediction provides a fitness benefit and enhances fungal survival. In this scenario, the response to glucose is linked to the ‘microbial memory’ as a prediction for the danger posed by the immune system. Additionally to glucose, other host-derived signals such as lactate (Childers et al. 2020), interleukin (IL)-8 (Ali et al. 2006), IL-17 (Zelante et al. 2012), tumour necrosis factor (TNF; Rocha et al. 2017), and prostaglandin E2 (PGE2) (Kundu and Noverr 2011) are also associated with adaptive prediction in C. albicans.
Figure 1.

Comparison of adaptation and adaptive prediction on the example of C. albicans and glucose (Rodaki et al. 2009). To cause systemic infection, C. albicans needs to translocate from the intestine, a commensal niche, into the bloodstream. There C. albicans encounters a new environment with stressors imposed by the host. The survival of the fungus depends on its acquired adaptation strategies. Adaptation (left panel): (i) Upon entering the bloodstream, C. albicans senses a higher glucose concentration compared to its previous niche. This leads to the expression of genes involved in metabolic remodelling (yellow arrow) to utilize the carbon source efficiently. (ii) Candida albicans will further encounter immune cells, which are perceived as a direct threat, resulting in the expression of genes involved in its defence mechanisms (blue/diagonal-patterned arrow). (iii) The pathogenic yeast was unsuccessful in rapidly adapting to the threat, giving the immune cells the advantage to overcome and eliminate the fungus. In this scenario, adaptation to environmental signals and adaptation to imposed stress are not linked to each other, potentially leading to delayed reactions. Adaptive prediction (right panel): (i) Upon entering the bloodstream, C. albicans senses the elevated glucose concentration and recognizes it not only as a nutrient source but as a potential precursor. Consequently, genes related to glucose metabolism are upregulated along with those involved in oxidative and cationic stress resistance (yellow - blue/diagonal-patterned arrow). (ii) This co-induced response prepares for the upcoming threat imposed by the immune system, thereby providing a proactive advantage. (iii) The mechanism of adaptive prediction provides a fitness benefit and enhances fungal survival. In this scenario, the response to glucose is linked to the ‘microbial memory’ as a prediction for the danger posed by the immune system. Additionally to glucose, other host-derived signals such as lactate (Childers et al. 2020), interleukin (IL)-8 (Ali et al. 2006), IL-17 (Zelante et al. 2012), tumour necrosis factor (TNF; Rocha et al. 2017), and prostaglandin E2 (PGE2) (Kundu and Noverr 2011) are also associated with adaptive prediction in C. albicans.

Microbial sensing of immunological communication

Factors that induce adaptive prediction in bacteria and fungi do not necessarily need to be of metabolic nature. Cytokines are signalling molecules that orchestrate intercellular communication between both immune and non-immune cells (Fig. 2A) (Ramesh et al. 2013, Akdis et al. 2016, Taherkhani et al. 2020). Early during infection, cytokines as well as DAMPs are released by tissue-resident immune cells and non-professional immune cells, such as epithelial cells (Stadnyk 2002, Naglik and Moyes 2011), inducing the recruitment of professional phagocytes to the site of infection. Some of the immunological signalling cytokines dictate very specific immune responses, e.g. IL-17 and IL-8 regulate neutrophil recruitment and activation (Azevedo et al. 2021, Matsushima et al. 2022), whereas chemokines such as MCP-1/CCL2 will specifically recruit monocytes to the site of infection (Zhang et al. 1994, Gunn et al. 1997), and IL-1 is a general inducer of potent innate immune activation (Mantovani et al. 2019). Bacterial and fungal pathogens that can sense cytokine production and 'predict' the host immune response would have a major fitness advantage to regulate pathogenicity, immune evasion, and escape mechanisms. On multiple occasions, bacterial and fungal pathogens have been described to interact with specific cytokines and chemokines (Table 1) that execute crucial roles in the host immune system (Table 2). The extent of these interactions varies dramatically between studies. Most commonly shown is the ability of bacteria and fungi to bind host cytokines and chemokines (Fig. 2B).

Microbial sensing of immunological communication signals. (A) Cytokines facilitate communication between immune cells. Upon recognition of pathogenic microbes, immune cells undergo activation and subsequently release an array of cytokines. Other immune cells respond to these signal molecules by killing the pathogen, recruiting more immune cells, or priming downstream signalling pathways to activate several cellular processes. (B) Cytokines can bind to microbial structures on the surface of microbes. Negatively charged lipopolysaccharides (LPS), type III secretion systems (T3SS), receptors such as outer membrane proteins (OMPs), glycosylphosphatidylinositol (GPI) anchor proteins, or cell wall polysaccharides have the capacity of binding cytokines. (C) The interaction between cytokines and pathogens can lead to two distinct outcomes: either the pathogen evades the immune response, or it activates defensive responses to counteract immune cells. (D) Cytokine sensing can induce various adaptive behaviours in pathogens, altering virulence factors and inducing biofilm formation. Cell membrane remodelling and morphological changes may also occur in response to cytokine binding, along with transcriptional and metabolic rewiring.
Figure 2.

Microbial sensing of immunological communication signals. (A) Cytokines facilitate communication between immune cells. Upon recognition of pathogenic microbes, immune cells undergo activation and subsequently release an array of cytokines. Other immune cells respond to these signal molecules by killing the pathogen, recruiting more immune cells, or priming downstream signalling pathways to activate several cellular processes. (B) Cytokines can bind to microbial structures on the surface of microbes. Negatively charged lipopolysaccharides (LPS), type III secretion systems (T3SS), receptors such as outer membrane proteins (OMPs), glycosylphosphatidylinositol (GPI) anchor proteins, or cell wall polysaccharides have the capacity of binding cytokines. (C) The interaction between cytokines and pathogens can lead to two distinct outcomes: either the pathogen evades the immune response, or it activates defensive responses to counteract immune cells. (D) Cytokine sensing can induce various adaptive behaviours in pathogens, altering virulence factors and inducing biofilm formation. Cell membrane remodelling and morphological changes may also occur in response to cytokine binding, along with transcriptional and metabolic rewiring.

Table 1.

Cytokines that are bound by bacteria and fungi.

Cytokine
*Cytokine has a lectin-like domain
ReceptorMicrobeMicrobial response
# No microbial response has been described
Reference
INF-γMmp10M. tuberculosisIncreased oxygen consumption rate and upregulation of virulence factorsAhmed et al. (2022)
 T3SSY. pestis#Zav'yalov et al. (1995)
 OprFP. aeruginosaUpregulation of PA-I expressionWu et al. (2005)
 BilRIA. actinomycetemcomitans#Ahlstrand et al. (2017)
IL-8*UnknownM. tuberculosis#Krupa et al. (2015)
 PilQN. meningitidisCell wall remodelling, modified transcriptional profiling, and enhanced complement resistanceMahdavi et al. (2013)
 BilRIA. actinomycetemcomitansReduced biofilm eDNA contentAhlstrand et al. (2017)
 LPS #Ahlstrand et al. (2019)
 UnknownC. albicansModified hyphal tip growth patternAli et al. (2006)
TNF*FN3 proteinB. longum#Dyakov et al. (2020)
 Glycosilated PilEN. meningitidisCell wall remodelling, modified transcriptional profiling, and enhanced complement resistanceMahdavi et al. 2013)
 UnknownS. flexneriIncreased invasion of non-professional phagocytic cellsLuo et al. (1993)
  S. typhimurium# 
  E. coliIncreased planktonic growth of virulent, but not avirulent, strainsLuo et al. (1993)
 BilRIA. actinomycetemcomitans#Lee et al. (2003a)
 N,N′-diacetylchitobioseC. albicansReduced biofilm metabolism and yeast-to-hyphae transitionAhlstrand et al. (2017)
IL-1β*Caf1AY. pestis#Rocha et al. (2017)
 UnknownE. coliIncreased planktonic growth of virulent, but not avirulent, strainsZav'yalov et al. (1995)
 BilRIA. actinomycetemcomitansReduced biofilm eDNA contentPorat et al. (1991)
IL-2*IrmA proteinE. coliIncreased planktonic growth of virulent, but not avirulent, strainsPaino et al. (2013)
IL-4*IrmA proteinE. coli#Ahlstrand et al. (2017)
IL-10IrmA proteinE. coli#Denis et al. (1991)
 BilRIA. actinomycetemcomitans#Moriel et al. (2016)
IL-17ACrh11pC. albicansAugmented filamentous growth, adhesion, and autophagyMoriel et al. (2016)
 Crf1A. fumigatusIncreased biofilm formation and autophagy 
CXCL9, CXCL10UnknownS. aureusProtein A (SPA) releaseAhlstrand et al. (2017)
Cytokine
*Cytokine has a lectin-like domain
ReceptorMicrobeMicrobial response
# No microbial response has been described
Reference
INF-γMmp10M. tuberculosisIncreased oxygen consumption rate and upregulation of virulence factorsAhmed et al. (2022)
 T3SSY. pestis#Zav'yalov et al. (1995)
 OprFP. aeruginosaUpregulation of PA-I expressionWu et al. (2005)
 BilRIA. actinomycetemcomitans#Ahlstrand et al. (2017)
IL-8*UnknownM. tuberculosis#Krupa et al. (2015)
 PilQN. meningitidisCell wall remodelling, modified transcriptional profiling, and enhanced complement resistanceMahdavi et al. (2013)
 BilRIA. actinomycetemcomitansReduced biofilm eDNA contentAhlstrand et al. (2017)
 LPS #Ahlstrand et al. (2019)
 UnknownC. albicansModified hyphal tip growth patternAli et al. (2006)
TNF*FN3 proteinB. longum#Dyakov et al. (2020)
 Glycosilated PilEN. meningitidisCell wall remodelling, modified transcriptional profiling, and enhanced complement resistanceMahdavi et al. 2013)
 UnknownS. flexneriIncreased invasion of non-professional phagocytic cellsLuo et al. (1993)
  S. typhimurium# 
  E. coliIncreased planktonic growth of virulent, but not avirulent, strainsLuo et al. (1993)
 BilRIA. actinomycetemcomitans#Lee et al. (2003a)
 N,N′-diacetylchitobioseC. albicansReduced biofilm metabolism and yeast-to-hyphae transitionAhlstrand et al. (2017)
IL-1β*Caf1AY. pestis#Rocha et al. (2017)
 UnknownE. coliIncreased planktonic growth of virulent, but not avirulent, strainsZav'yalov et al. (1995)
 BilRIA. actinomycetemcomitansReduced biofilm eDNA contentPorat et al. (1991)
IL-2*IrmA proteinE. coliIncreased planktonic growth of virulent, but not avirulent, strainsPaino et al. (2013)
IL-4*IrmA proteinE. coli#Ahlstrand et al. (2017)
IL-10IrmA proteinE. coli#Denis et al. (1991)
 BilRIA. actinomycetemcomitans#Moriel et al. (2016)
IL-17ACrh11pC. albicansAugmented filamentous growth, adhesion, and autophagyMoriel et al. (2016)
 Crf1A. fumigatusIncreased biofilm formation and autophagy 
CXCL9, CXCL10UnknownS. aureusProtein A (SPA) releaseAhlstrand et al. (2017)
Table 1.

Cytokines that are bound by bacteria and fungi.

Cytokine
*Cytokine has a lectin-like domain
ReceptorMicrobeMicrobial response
# No microbial response has been described
Reference
INF-γMmp10M. tuberculosisIncreased oxygen consumption rate and upregulation of virulence factorsAhmed et al. (2022)
 T3SSY. pestis#Zav'yalov et al. (1995)
 OprFP. aeruginosaUpregulation of PA-I expressionWu et al. (2005)
 BilRIA. actinomycetemcomitans#Ahlstrand et al. (2017)
IL-8*UnknownM. tuberculosis#Krupa et al. (2015)
 PilQN. meningitidisCell wall remodelling, modified transcriptional profiling, and enhanced complement resistanceMahdavi et al. (2013)
 BilRIA. actinomycetemcomitansReduced biofilm eDNA contentAhlstrand et al. (2017)
 LPS #Ahlstrand et al. (2019)
 UnknownC. albicansModified hyphal tip growth patternAli et al. (2006)
TNF*FN3 proteinB. longum#Dyakov et al. (2020)
 Glycosilated PilEN. meningitidisCell wall remodelling, modified transcriptional profiling, and enhanced complement resistanceMahdavi et al. 2013)
 UnknownS. flexneriIncreased invasion of non-professional phagocytic cellsLuo et al. (1993)
  S. typhimurium# 
  E. coliIncreased planktonic growth of virulent, but not avirulent, strainsLuo et al. (1993)
 BilRIA. actinomycetemcomitans#Lee et al. (2003a)
 N,N′-diacetylchitobioseC. albicansReduced biofilm metabolism and yeast-to-hyphae transitionAhlstrand et al. (2017)
IL-1β*Caf1AY. pestis#Rocha et al. (2017)
 UnknownE. coliIncreased planktonic growth of virulent, but not avirulent, strainsZav'yalov et al. (1995)
 BilRIA. actinomycetemcomitansReduced biofilm eDNA contentPorat et al. (1991)
IL-2*IrmA proteinE. coliIncreased planktonic growth of virulent, but not avirulent, strainsPaino et al. (2013)
IL-4*IrmA proteinE. coli#Ahlstrand et al. (2017)
IL-10IrmA proteinE. coli#Denis et al. (1991)
 BilRIA. actinomycetemcomitans#Moriel et al. (2016)
IL-17ACrh11pC. albicansAugmented filamentous growth, adhesion, and autophagyMoriel et al. (2016)
 Crf1A. fumigatusIncreased biofilm formation and autophagy 
CXCL9, CXCL10UnknownS. aureusProtein A (SPA) releaseAhlstrand et al. (2017)
Cytokine
*Cytokine has a lectin-like domain
ReceptorMicrobeMicrobial response
# No microbial response has been described
Reference
INF-γMmp10M. tuberculosisIncreased oxygen consumption rate and upregulation of virulence factorsAhmed et al. (2022)
 T3SSY. pestis#Zav'yalov et al. (1995)
 OprFP. aeruginosaUpregulation of PA-I expressionWu et al. (2005)
 BilRIA. actinomycetemcomitans#Ahlstrand et al. (2017)
IL-8*UnknownM. tuberculosis#Krupa et al. (2015)
 PilQN. meningitidisCell wall remodelling, modified transcriptional profiling, and enhanced complement resistanceMahdavi et al. (2013)
 BilRIA. actinomycetemcomitansReduced biofilm eDNA contentAhlstrand et al. (2017)
 LPS #Ahlstrand et al. (2019)
 UnknownC. albicansModified hyphal tip growth patternAli et al. (2006)
TNF*FN3 proteinB. longum#Dyakov et al. (2020)
 Glycosilated PilEN. meningitidisCell wall remodelling, modified transcriptional profiling, and enhanced complement resistanceMahdavi et al. 2013)
 UnknownS. flexneriIncreased invasion of non-professional phagocytic cellsLuo et al. (1993)
  S. typhimurium# 
  E. coliIncreased planktonic growth of virulent, but not avirulent, strainsLuo et al. (1993)
 BilRIA. actinomycetemcomitans#Lee et al. (2003a)
 N,N′-diacetylchitobioseC. albicansReduced biofilm metabolism and yeast-to-hyphae transitionAhlstrand et al. (2017)
IL-1β*Caf1AY. pestis#Rocha et al. (2017)
 UnknownE. coliIncreased planktonic growth of virulent, but not avirulent, strainsZav'yalov et al. (1995)
 BilRIA. actinomycetemcomitansReduced biofilm eDNA contentPorat et al. (1991)
IL-2*IrmA proteinE. coliIncreased planktonic growth of virulent, but not avirulent, strainsPaino et al. (2013)
IL-4*IrmA proteinE. coli#Ahlstrand et al. (2017)
IL-10IrmA proteinE. coli#Denis et al. (1991)
 BilRIA. actinomycetemcomitans#Moriel et al. (2016)
IL-17ACrh11pC. albicansAugmented filamentous growth, adhesion, and autophagyMoriel et al. (2016)
 Crf1A. fumigatusIncreased biofilm formation and autophagy 
CXCL9, CXCL10UnknownS. aureusProtein A (SPA) releaseAhlstrand et al. (2017)
Table 2.

Immune mediators and their function that microbes could benefit from interfering with.

Immune mediatorFunction
IL-1αGeneral alarmin released upon tissue damage that potently activates immune responses
IL-1βPotent broad-spectrum mediator of inflammation released upon innate immune activation
IL-1RaEndogenous antagonist of the interleukin-1 receptor
IL-2Activator of T-cell proliferation
IL-3Hematopoietic growth factor of myeloid cells and their progenitors
IL-4Potent driver of T-helper 2 and B-cell responses
IL-6Mediator of systemic inflammatory responses
IL-7Stimulation of T and B-cell proliferation
IL-8 (CXCL8)Neutrophil chemoattractant and activator
IL-10Potent anti-inflammatory cytokine
IL-17APotent driver of neutrophil-mediated inflammation and activator of mucosal host defence
IL-18Activation of T-helper 1 immunity and driving IFN-γ responses
α1-antitrypsinAcute-phase protein with potent protease inhibitory function
CXCL9 (MIG)IFNγ-induced broad-spectrum chemokine
CXCL10 (IP10)IFNγ-induced broad-spectrum chemokine
GM-CSFActivation, survival, and proliferation factor for myeloid cells
MCP3 (CCL7)Broad-spectrum chemokine
MIP1α (CCL3)Recruitment and activation of granulocytes
PGE2Immunoregulatory lipid mediator
TGF-βBroad spectrum immunoregulatory cytokine with antiproliferative effects
IFN-γActivator of myeloid phagocytes, promotes killing of intracellular and phagocytosed pathogens
TNFPotent broad-spectrum mediator of inflammation released upon innate immune activation
Immune mediatorFunction
IL-1αGeneral alarmin released upon tissue damage that potently activates immune responses
IL-1βPotent broad-spectrum mediator of inflammation released upon innate immune activation
IL-1RaEndogenous antagonist of the interleukin-1 receptor
IL-2Activator of T-cell proliferation
IL-3Hematopoietic growth factor of myeloid cells and their progenitors
IL-4Potent driver of T-helper 2 and B-cell responses
IL-6Mediator of systemic inflammatory responses
IL-7Stimulation of T and B-cell proliferation
IL-8 (CXCL8)Neutrophil chemoattractant and activator
IL-10Potent anti-inflammatory cytokine
IL-17APotent driver of neutrophil-mediated inflammation and activator of mucosal host defence
IL-18Activation of T-helper 1 immunity and driving IFN-γ responses
α1-antitrypsinAcute-phase protein with potent protease inhibitory function
CXCL9 (MIG)IFNγ-induced broad-spectrum chemokine
CXCL10 (IP10)IFNγ-induced broad-spectrum chemokine
GM-CSFActivation, survival, and proliferation factor for myeloid cells
MCP3 (CCL7)Broad-spectrum chemokine
MIP1α (CCL3)Recruitment and activation of granulocytes
PGE2Immunoregulatory lipid mediator
TGF-βBroad spectrum immunoregulatory cytokine with antiproliferative effects
IFN-γActivator of myeloid phagocytes, promotes killing of intracellular and phagocytosed pathogens
TNFPotent broad-spectrum mediator of inflammation released upon innate immune activation
Table 2.

Immune mediators and their function that microbes could benefit from interfering with.

Immune mediatorFunction
IL-1αGeneral alarmin released upon tissue damage that potently activates immune responses
IL-1βPotent broad-spectrum mediator of inflammation released upon innate immune activation
IL-1RaEndogenous antagonist of the interleukin-1 receptor
IL-2Activator of T-cell proliferation
IL-3Hematopoietic growth factor of myeloid cells and their progenitors
IL-4Potent driver of T-helper 2 and B-cell responses
IL-6Mediator of systemic inflammatory responses
IL-7Stimulation of T and B-cell proliferation
IL-8 (CXCL8)Neutrophil chemoattractant and activator
IL-10Potent anti-inflammatory cytokine
IL-17APotent driver of neutrophil-mediated inflammation and activator of mucosal host defence
IL-18Activation of T-helper 1 immunity and driving IFN-γ responses
α1-antitrypsinAcute-phase protein with potent protease inhibitory function
CXCL9 (MIG)IFNγ-induced broad-spectrum chemokine
CXCL10 (IP10)IFNγ-induced broad-spectrum chemokine
GM-CSFActivation, survival, and proliferation factor for myeloid cells
MCP3 (CCL7)Broad-spectrum chemokine
MIP1α (CCL3)Recruitment and activation of granulocytes
PGE2Immunoregulatory lipid mediator
TGF-βBroad spectrum immunoregulatory cytokine with antiproliferative effects
IFN-γActivator of myeloid phagocytes, promotes killing of intracellular and phagocytosed pathogens
TNFPotent broad-spectrum mediator of inflammation released upon innate immune activation
Immune mediatorFunction
IL-1αGeneral alarmin released upon tissue damage that potently activates immune responses
IL-1βPotent broad-spectrum mediator of inflammation released upon innate immune activation
IL-1RaEndogenous antagonist of the interleukin-1 receptor
IL-2Activator of T-cell proliferation
IL-3Hematopoietic growth factor of myeloid cells and their progenitors
IL-4Potent driver of T-helper 2 and B-cell responses
IL-6Mediator of systemic inflammatory responses
IL-7Stimulation of T and B-cell proliferation
IL-8 (CXCL8)Neutrophil chemoattractant and activator
IL-10Potent anti-inflammatory cytokine
IL-17APotent driver of neutrophil-mediated inflammation and activator of mucosal host defence
IL-18Activation of T-helper 1 immunity and driving IFN-γ responses
α1-antitrypsinAcute-phase protein with potent protease inhibitory function
CXCL9 (MIG)IFNγ-induced broad-spectrum chemokine
CXCL10 (IP10)IFNγ-induced broad-spectrum chemokine
GM-CSFActivation, survival, and proliferation factor for myeloid cells
MCP3 (CCL7)Broad-spectrum chemokine
MIP1α (CCL3)Recruitment and activation of granulocytes
PGE2Immunoregulatory lipid mediator
TGF-βBroad spectrum immunoregulatory cytokine with antiproliferative effects
IFN-γActivator of myeloid phagocytes, promotes killing of intracellular and phagocytosed pathogens
TNFPotent broad-spectrum mediator of inflammation released upon innate immune activation

Binding of mammalian cytokines by microbial pathogens

Many microbe–cytokine binding interactions have been associated with the lectin-like properties of cytokines. Lectin-like domains are present in cytokines such as IL-1α (Cebo et al. 2002), IL-1β (Fukushima et al. 1997, Cebo et al. 2002), IL-2 (Sherblom et al. 1989, Cebo et al. 2002), IL-3, IL-4, IL-6, IL-7 (Cebo et al. 2002), and TNF (Cebo et al. 2002, Rocha et al. 2017). These lectin-like domains in cytokines were suggested to play a role in receptor and cell specificity of cytokines depending on carbohydrate recognition (Cebo et al. 2002) and can bind to glycans and glycosylated proteins constituting microbial cell walls.

The yeast C. albicans was found to bind a variety of cytokines with lectin-like domains. Abundantly exposed mannose residues on the fungal cell wall enable IL-2 binding, which can be inhibited by the addition of exogenous mannan (Treseler et al. 1992, Zanetta et al. 1998). Similarly, TNF binds C. albicans via its lectin-like properties (Rocha et al. 2017). Lectin-mediated effects were also proposed as the mechanism of IL-8 and TNF binding by the type four pili (Tpf) subunit PilE of Neisseria meningitidis. Cytokine binding and internalization were associated with pili glycosylation, as the binding of these cytokines could be competed away using lectins (Mahdavi et al. 2013).

The interaction of oppositely charged molecules may also facilitate cytokine binding by microbes. Aggregatibacter actinomycetemcomitans negatively charged lipopolysaccharide (LPS) on the outer membrane was found to bind host positively charged IL-8. This cytokine also binds to A. actinomycetemcomitans outer membrane vesicles (OMVs), of which LPS is one of the major components, supporting the existence of opposite-charge-based interactions that could also be involved in cytokine binding by other Gram-negative bacteria (Ahlstrand et al. 2019).

Collectively, this suggests that the binding of human cytokines to many microbes relies on natural lectin-like properties and molecular charges of cytokines. Although the function of these interactions remains elusive, this may have evolved by the immune system to serve as an opsonizing mechanism. This was observed for Mycobacterium tuberculosis binding of IL-8, which promotes phagocytic uptake and killing by neutrophils (Krupa et al. 2015). Similarly, C. albicans with bound IL-2 was shown to elicit increased proliferation rates of PBMCs compared to C. albicans without bound cytokine (Treseler et al. 1992).

Even though cytokine binding by microbes can be mediated by non-specific interactions, several studies elucidated specific bacterial and fungal receptors involved in host cytokine binding (Zav'yalov et al. 1995, Wu et al. 2005, Gendrin et al. 2010, Yung et al. 2011, Zelante et al. 2012, Mahdavi et al. 2013, Paino et al. 2013, Moriel et al. 2016, Ahlstrand et al. 2017, Hogbom and Ihalin 2017, Dyakov et al. 2020, Ahmed et al. 2022), a feature that is also widespread among viruses (Hernaez and Alcamí 2020). Besides minor structural similarities, no remarkable commonly shared structures can be observed, neither between the different bacterial cytokine-binding receptors nor between bacterial and human cytokine receptors. This could indicate that bacterial cytokine-binding proteins did not arise from a single origin, suggesting that cytokine-binding proteins evolved convergently (Hogbom and Ihalin 2017).

Specific receptors for host cytokines have been found in the outer membrane of Gram-negative bacteria and classified within the family of outer membrane proteins (OMPs), which serve essential roles in cell functions and as virulence factors in immune evasion (Rollauer et al. 2015). Yersinia pestis capsule antigen F1 assembly protein (Caf1A) was the first pore-forming OMP identified to bind host cytokines. Caf1A binds host IL-1β through its PapC domain, overlapping with Caf1 capsular protein binding site (Zav'yalov et al. 1995). Other pore-forming OMPs of Pseudomonas aeruginosa (OprF) and N. meningitidis (PilQ) were found to bind host interferon-γ (IFN-γ) (Wu et al. 2005) or bind and internalize IL-8 and TNF, respectively (Mahdavi et al. 2013). PilQ is involved in type IV pili (Tfp) assembly, interacting with Tfp PilE subunit, which has also shown the ability to bind these cytokines in a glycosylation-dependent manner (Mahdavi et al. 2013). In A. actinomycetemcomitans, an intrinsically disordered outer membrane lipoprotein, bacterial interleukin receptor I (BilRI), located both on the bacterial outer membrane and OMVs, was demonstrated to interact with IL-1β, IL-8, IL-10, TNF, TGF-β, and IFN-γ, and also mediates internalization of IL-1β (Paino et al. 2013, Ahlstrand et al. 2017). Not attached to the membrane but in the extracellular space, a secreted soluble protein termed IrmA (interleukin receptor mimic protein A) of E. coli was found to bind IL-2, IL-4, and, to a lesser extent, IL-10, due to its structural similarity with those cytokines’ corresponding receptors (Moriel et al. 2016).

Apart from IL-1β binding through Caf1A protein, Y. pestis interacts with IFN-γ and TNF via its type 3 secretion system (T3SS) protein LcrV (V-antigen) (Nakajima and Brubaker 1993, Nakajima et al. 1995, Gendrin et al. 2010), which constitutes a major virulence factor. This interaction occurs in a concentration-dependent fashion, and was suggested not to disturb the association of IFN-γ with its receptor (Gendrin et al. 2010). However, it was suggested to suppress IFN-γ and TNF levels in mice (Nakajima and Brubaker 1993, Nakajima et al. 1995). Mycobacterium tuberculosis can also associate with IFN-γ through the membrane protein large 10 (Mmp10), modulating host–pathogen interplay (Ahmed et al. 2022). Mycobacterium tuberculosis interaction with IL-8 was also reported, even though the bacterial receptor implicated has not been clarified (Krupa et al. 2015).

The Gram-positive bacterium, Staphylococcus aureus, was observed to interact with a striking myriad of 31 human chemokines out of a screening of 42 chemokines. Further characterization of the chemokines CXCL9 and CXCL10 revealed direct binding of the chemokines to both, the cell wall and the membrane (Yung et al. 2011). Regarding bifidobacteria, most species carry the PFNA gene cluster, which is thought to be involved in microbe–host communication. A purified fragment of FN3 protein (encoded in the PFNA gene cluster) of Bifidobacterium longum was shown to bind human TNF, presumably due to the presence of two fibronectin domains similar to cytokine-binding sites of human receptors (Dyakov et al. 2020). Further interactions of E. coli with IL-1β (Porat et al. 1991) and TNF (Luo et al. 1993), as well as Shigella flexneri and Salmonella typhimurium with TNF (Luo et al. 1993), were reported, although the bacterial receptors involved in these interactions have not been identified yet.

In fungi, the C. albicans Crh11p protein, a GPI-anchored fungal protein involved in cell wall assembly and regeneration (Pardini et al. 2006), was identified to bind human IL-17A, leading to changes in fungal virulence. Similarly, IL-17A binding was observed in the environmental fungus Aspergillus fumigatus depending on Crf1, an orthologue of C. albicans CRH GPI proteins (Zelante et al. 2012). Candida albicans also binds the neutrophil chemoattractant IL-8 during germination (Ali et al. 2006). Interestingly, antibody staining for IL-8 receptor A (IL-8RA) also stained the tips of the hyphae (Ali et al. 2006). This could suggest the presence of proteins in the fungal cell wall similar to IL-8RA that exhibit cross-reactivity with anti-IL-8RA antibodies.

Microorganisms can also indirectly bind various host glycoproteins, including cytokines such as the monocyte chemotactic protein 3 (MCP3), the macrophage inflammatory protein 1α (MIP-1α/CCL3) or IFN-γ, via heparin, leading to reduced chemokine-induced chemotaxis (Duensing et al. 1999). Staphylococcus aureus protein A (SpA), known for its ability to bind the Fc region of immunoglobulins (Foster 2005), was also found to bind heparin (Shi et al. 2021), and thereby potentially several cytokines with it.

Some microbial cytokine-binding molecules seem to have broad binding capacity, such as BilRI. This interestingly can be explained based on its properties as an intrinsically disordered protein, which lacks a properly defined 3D structure, allowing the interaction with several different molecules (Ahlstrand et al. 2017), while for cytokines with lectin-like domains, it appears plausible that specific carbohydrate structures are preferred binding partners.

For many of the interactions of microorganisms with immune mediators through binding, downstream microbial responses, physiological adaptations, or consequences have not been described or studied in depth (Table 1). However, even without inducing microbial responses, cytokine binding can already impact host–pathogen interactions drastically. Based on literature on other consequences of protein binding, we can extrapolate some potential effects resulting from binding of cytokines to microbial pathogens. Although it has not been described for cytokines, binding of host proteins (e.g. extracellular matrix components) can mask pathogen-associated molecular patterns (PAMPs) that are recognized by innate immune cells (Bhattacharya and Horswill 2024) or avoid complement activation, promoting immune evasion (Tomlin and Piccinini 2018). This is similar to how immunologically inert proteins mask PAMPs of spores of fungi such as A. fumigatus or how a capsule masks Cryptococcus neoformans PAMPs (Erwig and Gow 2016, van de Veerdonk et al. 2017). The binding of complement regulatory proteins by pathogens is a strategy to decrease complement activation, which increases survival (Kotwal 2000, Reuter et al. 2010, Meri et al. 2013, Hovingh et al. 2016, Moore et al. 2021).

In line with this, cytokine binding by pathogenic fungi and bacteria could, under specific circumstances, serve as a mechanism to sequester cytokines from reaching the cognate receptor on immune cells, thus hindering their subsequent initiation or chemoattraction. This strategy was shown for several poxviruses, which express a viral IL-18 bp to sequester host IL-18 and impair the subsequent IL-18-induced IFN-γ production and natural killer cell responses (Xiang and Moss 1999, Born et al. 2000, Smith et al. 2000). Yet, bacterial cytokine sequestration also has been shown to impact cells expressing the appropriate receptors (Duensing et al. 1999). Moreover, internalization of cytokines by bacteria (Mahdavi et al. 2013, Paino et al. 2013, Ahlstrand et al. 2017) will limit their availability for the corresponding recipient immune cells.

In conclusion, cytokine binding has been identified in numerous bacterial and fungal pathogens mediated by several diverse microbial binding components. These range from oppositely charged proteins to cell wall structures, secretory proteins, and microbial cytokine receptors (Table 1). Cytokine–microbe interaction can occur through biophysical mechanisms, often involving the lectin-like properties of cytokines. Cytokine binding may serve as a strategy for immune evasion or escape (Fig. 2C), suggesting an adaption by microbes to exploit host immune signals. While the precise functions of these interactions remain to be fully elucidated, in a variety of cases microbes have been observed to activate anticipatory responses to the cytokines they interact with.

Microbial responses to mammalian cytokines and immune mediators

While viruses can efficiently modulate immune responses, several bacterial and fungal pathogens have been shown to actively respond when interacting with host immune mediators. A microbial response to host cytokines requires a reciprocal interaction between both, the host and the pathogen. Exemplary, incubation of peripheral blood mononuclear cells (PBMCs) with P. aeruginosa biofilms activates an inflammatory response reflected in the corresponding cytokine secretion. Interestingly, the PBMC:Biofilm co-culture is beneficial for the pathogen, as it increases the number of biofilm-associated bacteria (Kaya et al. 2020). Pseudomonas aeruginosa biofilms cultured with challenged PBMC supernatants, which contain pro-inflammatory and anti-inflammatory cytokines, also increased the number of viable colony-forming units, suggesting bacterial feedback to the host inflammatory environment (Kaya et al. 2020). In addition to physical interaction, specific mammalian immune mediators have been observed to trigger microbial responses in terms of metabolism, growth, morphology, and/or biofilm formation. These responses could suggest that adaptations favouring pathogenicity are induced by prior recognition of host cytokines before immune cells are encountered.

Similar to the example above, a variety of bacterial pathogens induce adaptations in response to cytokines. Growth of virulent, but not avirulent, E. coli strains increases when cultured in the presence of IL-1β (Porat et al. 1991), IL-2, GM-CSF (Denis et al. 1991), and TNF (Lee et al. 2003a), suggesting that cytokine sensing is a virulence-associated trait. The effects of IL-1β on E. coli growth could be neutralized by the host endogenous IL-1 receptor antagonist (IL-1Ra) (Porat et al. 1991). This could suggest that IL-1β sensing by the bacteria is mediated by an IL-1-receptor-like structure (Porat et al. 1991). Despite enhancing planktonic bacterial growth, IL-1β seems to reduce biofilm formation and haemolytic activity of uropathogenic E. coli (UPEC; Engelsoy et al. 2019). Effects of TNF on E. coli growth were also validated in a mouse model. While TNF-deficient mice show a drastically increased susceptibility to E. coli infections due to a compromised host response, in the background of neutropenia, the bacterial burden was reduced in TNF-deficient mice (Lee et al. 2003a).

Clinical isolates of S. aureus and Acinetobacter sp. also show enhanced growth when incubated in the presence of IL-1β, while P. aeruginosa growth can be increased by the presence of IL-6 (Meduri et al. 1999). Interestingly, continuous passages of the clinical isolates in vitro were associated with a loss of the phenotype (Meduri et al. 1999), which could indicate that the ability to adapt in response to mammalian cytokines is transient and only coupled to adaptation when bacteria have recently encountered the immune system in the host. IL-6 also promoted the growth of M. avium, a bacterium that survives within macrophages, demonstrated by increased intracellular growth rates when IL-6 was present (Denis and Gregg 1991). Strikingly, growth was also augmented by IL-6 in the absence of macrophages, suggesting a direct effect of IL-6 on bacterial growth. Staphylococcus aureus biofilms showed an increased biomass when exposed to proinflammatory IL-1β (McLaughlin and Hoogewerf 2006, Gutierrez Jauregui et al. 2019). Interestingly, nasal carrier strains of S. aureus induce lower IL-1α and IL-1β responses in naïve nasal epithelial cells compared to non-carrier strains. This modulation of the immune response favours the bacterium, since IL-1α can drive inflammatory responses as well as negatively affect bacterial planktonic growth directly (Quinn et al. 2009). Aggregatibacter actinomycetemcomitans biofilm composition changes in response to IL-1β and IL-8, which seems mediated by its BilRI (Ahlstrand et al. 2017). Strikingly, BilRI-deficient mutants (bilRI) and thus unable to bind these cytokines, exhibited a different biofilm composition compared to wild-type (Ahlstrand et al. 2017).

In M. tuberculosis, exposure to IFN-γ increases oxygen consumption rate and improves microbial persistence in vitro in a 3D granuloma model (Ahmed et al. 2022). Of note, IFN-γ did not increase M. tuberculosis growth in the absence of immune cells, suggesting that the augmented bacterial burden is due to increased fitness and resistance to the immune attack, rather than increased bacterial growth itself (Ahmed et al. 2022). Despite the effects of IFN-γ on potentiating M. tuberculosis growth, the enhanced metabolic activity translates to increased sensitivity to isoniazid (Ahmed et al. 2022), for which normally extremely long treatment regimens are required due to slow mycobacterial growth.

Further studies characterized specific bacterial and fungal responses to mammalian cytokines such as altered virulence factor expression and/or increased virulence in infection models. Pseudomonas aeruginosa senses IFN-γ through OprF, and upregulates expression of P. aeruginosa type I lectin (PA-I), one of its key adhesins and virulence factors, which is involved in epithelial barrier disruption (Wu et al. 2005). Pseudomonas response to IFN-γ was proved to be mediated by activation of a quorum sensing (QS) signalling system, as the increase of PA-I after IFN-γ exposure was abrogated in mutant strains lacking the required genes for QS system activation (rhlI and rhlR). Similarly, sensing of IFN-γ by M. tuberculosis results in upregulation of virulence factor genes, including the RNase toxin vapC14 and ESAT-6-like protein esxP (Ahmed et al. 2022). Sensing of TNF by S. typhimurium alters expression of virulence effectors, consequently increasing invasiveness of epithelial cells and altering the host immune responses (Ma et al. 2010). Chronic Salmonella infection was associated with an increased risk of inflammatory bowel disease development, where TNF is increased (Ma et al. 2010). This association is due to the altered immune response triggered by the bacterium (Zha et al. 2019). Neisseria meningitidis induces bacterial cell membrane remodelling, transcriptional responses, and enhanced virulence following uptake of pro-inflammatory cytokines (Mahdavi et al. 2013). Mutant strains lacking the specific receptors result in reduced animal mortality (Mahdavi et al. 2013). UPEC sensing of cytokines increases the expression of iron acquisition systems and those related to fimbriae production, resulting in enhanced virulence in a C. elegans infection model (Engelsoy et al. 2019). Staphylococcus aureus sensing of IL-1β increases its growth but is also related to changes in virulence gene expression, including leukotoxins and surface proteins involved in extracellular matrix binding such as the fibronectin-binding protein or the collagen-binding protein (Kanangat et al. 2007). This was suggested to potentially contribute towards improved invasion and persistence within the host inflammatory environment. Further, the binding of CXCL9 and CXCL10 by S. aureus was observed to induce protein A (SpA) release (Yung et al. 2011), a wall-anchored protein involved in immune evasion, avoiding opsonization (Foster 2005). SpA can also be secreted to the extracellular compartment, where it acts as a superantigen (Bear et al. 2023). Protein A deletion increases bacterial killing by phagocytes and results in decreased virulence in vivo (Foster 2005). The relevance of S. aureus responding to CXCL9 is reflected by the fact that this chemokine is present in high amounts in the skin, the natural niche of S. aureus (Yung et al. 2011). Additionally, both CXCL9 and CXCL10 are upregulated by the type 1 immunity hallmark cytokine IFN-γ (Kanda et al. 2007, Corbera-Bellalta et al. 2016). This could suggest that S. aureus uses the chemokine presence to evaluate the current host immune status. Although CXCL9 and CXCL10 were not addressed specifically, another study showed that IFN-γ-associated CXCL-type chemokine production can support S. aureus infection in the skin (McLoughlin et al. 2008). A detrimental involvement of IFN-γ in S. aureus-caused disease suggests that the bacterium uses CXCL chemokines to recognize a state of host susceptibility, before engaging a virulence program (Wei et al. 1999, McLoughlin et al. 2008, Satorres et al. 2009, Yung et al. 2011). Similarly, this could be the case for E. coli IrmA protein, which mimics IL-4 and IL-2 receptors, thereby sensing a Th2-dominated environment (Moriel et al. 2016). As IL-4/IL-13-driven type 2 immunity generally is considered inadequate for efficient antibacterial defence (Hultgren et al. 1998, Potian 2011, Woytschak 2016), IrmA thus might also act as a host susceptibility sensor and downstream lead to engagement of a virulence program.

Similar to bacteria, fungi also exhibit responses to host cytokines, promoting their persistence during infection. The sensing of the neutrophil chemoattractant IL-8 by C. albicans leads to a modification in the hyphal tip growth pattern, away from areas with high IL-8 concentrations (Ali et al. 2006). Since the hyphal tip is likely more sensitive to neutrophil attack and IL-8 promotes their recruitment, such a response could have evolved as a strategy to avoid a neutrophil attack. Sensing of IL-17A by C. albicans induces not only changes in fungal growth but also in filamentation dynamics and transcriptional responses, characterized by the upregulation of genes involved in autophagy and hyphal morphogenesis, which was associated with improved fungal fitness against neutrophils (Zelante et al. 2012). Similar observations were made with A. fumigatus (Zelante et al. 2012), which is surprising since A. fumigatus evolved its pathogenicity in environmental niches (Casadevall 2003, Tekaia 2005, Hube 2009). Thus, A. fumigatus unlikely would have co-evolved with a host expressing IL-17. However, proteins with structural similarity to IL-17A have been found in C. elegans (Chen et al. 2017). As nematodes and amoebae can represent a major predatory threat to environmental fungi (Mylonakis 2002, Casadevall 2003), this raises the possibility of the development of IL-17A sensing capability in an early common ancestor. In contrast, when interacting with TNF, C. albicans exhibits reduced biofilm metabolism and yeast-to-hyphae transition (Rocha et al. 2017) and also IFN-γ limits its filamentous growth (Kalo-Klein and Witkin 1990).

The ability of microbial pathogens to react to host cytokines and other immune mediators reveals a complex field of host–pathogen interactions. Thereby, specific cytokine signalling facilitates microbial adaptation such as enhanced proliferation, altered virulence factor expression, biofilm formation, morphological changes, cell remodelling, as well as metabolic and transcriptional alterations (Fig. 2D). The observed microbial responses suggest that cytokine sensing is a virulence-associated trait, enabling pathogens to modulate their response to the host immune environment.

Adaptation to in-direct signals of altered immune activation

Microbial interaction with immune cells can elicit host cell death. In the case of C. albicans, the toxin candidalysin induces macrophage lysis and subsequent release of intracellular content as well as NLRP3 inflammasome activation (Kasper et al. 2018). In this context, the release of macrophage cytosolic content and IL-1β through pyroptosis will promote recruitment and activation of further immune cells to overcome the infection (Gross et al. 2009, Drummond et al. 2019). Interestingly, the macrophage-derived protein prothymosin α, found in macrophage lysates, can augment fungal pathogenicity by inducing filamentation (Case et al. 2021). This example showcases that microbes have evolved strategies to cope with the immune activation upon macrophage lysis through recognition of macrophage cytosolic molecules.

Activation of inflammation causes induction of acute-phase proteins. Intriguingly, the acute phase serum protein α1-antitrypsin can induce transcriptional changes in C. albicans characterized by reduced expression of filamentation repressors, also leading to more potent filamentation and thereby impairing fungal clearance by innate immune cells (Jaeger et al. 2024). Inflammation is also associated with epithelial barrier disruption, vascular leakage, and infiltration of serum proteins in infected tissues. In line with this, albumin, the most abundant protein in human serum, was found in the vaginal fluid of vaginal candidiasis patients (Tang et al. 2007). Interestingly, the presence of albumin boosts proliferation and unlocks pathogenic potential, as well as oxidative stress resistance, of the yeast pathogen C. glabrata (Pekmezovic et al. 2021). Albumin was also found to protect group G streptococci against antibacterial peptides released upon epithelial activation. As common commensals of the oropharyngeal tract, albumin-binding capability was hypothesized to have a role in nutrient acquisition under non-inflammatory conditions (Egesten et al. 2011).

Cross-kingdom communication via lipid mediators and hormones

In addition to cytokines that are specific to the mammalian host, the immune system also relies on a network of lipid mediators (eicosanoids) that not only mediate pro-inflammatory effects but also play crucial roles in resolving inflammation (Dennis and Norris 2015). The family of eicosanoids includes prostaglandins, thromboxanes, and leukotrienes, metabolites derived from either cyclooxygenases or lipoxygenases acting on the precursor arachidonic acid (AA), which is released from host membranes (Debeuf 2018). Although synthesis of cytokines has not been described in microbes, most human pathogenic fungi, but not bacteria, can synthesize eicosanoids, especially when free AA is available to them (Noverr et al. 2002). In fungi, different enzymes are involved in the process of prostaglandin synthesis. While Aspergillus spp. possess genes (ppoA, ppoB, and ppoC) that encode for cyclooxygenase-like enzymes (Tsitsigiannis et al. 2005), C. albicans employs a fatty acid desaturase (Ole2) and a multicopper oxidase (Fet3) for PGE2 production (Erb-Downward and Noverr 2007). Nevertheless, compounds commonly used to inhibit PGE2 production in humans are also effectively decreasing the production levels of PGE2 by C. albicans and C. dubliniensis (Ells et al. 2011). This suggests that fungal enzymes not only exhibit functional similarities but also structural resemblances to their human counterparts. Candida albicans and C. neoformans do not only possess the machinery to produce prostaglandins, but their secreted prostaglandins modulate immune responses by increasing the release of the anti-inflammatory cytokine IL-10 (Noverr et al. 2001). Cryptococcus neoformans-derived phospholipase can aid its survival within macrophages by facilitating eicosanoid production and impairing macrophage antifungal defences (Noverr et al. 2003). Candida albicans mutants deficient in PGE2 production exhibited reduced fitness in the gastrointestinal tract of mice, and fungal PGE2 production protected C. albicans from intestinal phagocytes (Tan et al. 2019). These findings imply that PGE2 promotes colonization and survival in the commensal niche (Tan et al. 2019). Fungal eicosanoids also seem to play a key role in pathogenicity mechanisms and fungal survival, as cyclooxygenase inhibitors were observed to reduce the viability of C. albicans and C. neoformans (Noverr et al. 2001). In C. albicans, eicosanoids were observed to regulate the yeast-to-hypha transition (Kalo-Klein and Witkin 1990, Noverr et al. 2001, Noverr and Huffnagle 2004, Ells et al. 2011). Strikingly, the hyphal, but not yeast, morphology of C. albicans is also a strong inducer of eicosanoids in neutrophils and macrophages (Fischer et al. 2021, Schimanski et al. 2024), suggesting an adaptive strategy to anticipate the neutrophil-imposed stress that would follow eicosanoid production.

Host, as well as fungal-derived PGE2, was shown to compromise vaccination efficiency and redirect cytokine responses from a protective Th1- towards a Th2-associated phenotype (Kundu and Noverr 2011). Opportunistic pathogenic fungi may produce lipids to modulate the host immune response, but also exploit host-produced lipids to enhance their pathogenicity. Altogether, recognition and even more so the production of lipid mediators enables pathogenic fungi to directly manipulate signalling processes of the immune network, which might play an important effector role in deploying adaptive pathogenicity. Elucidating how and under which circumstances eicosanoid synthesis is switched on is essential to understand virulence of pathogenic fungi such as Candida spp., and Aspergillus spp.

A special case of lipid mediator communication between host and pathogen is steroid hormones. Steroid hormones, both glucocorticoids and sex hormones, play a role in immunity, reducing inflammation and shaping immune cell differentiation and cytokine production (Bereshchenko et al. 2018). As a commensal of the vaginal mucosa, C. albicans is widely exposed to estrogen and also expresses a high-affinity estrogen-binding protein that partially mediates the inhibitory effect of hyphal formation by 17β-estradiol (Madani et al. 1994, Kurakado et al. 2017, Sherrington et al. 2018). More recently, it was shown that upon exposure to this hormone, C. albicans undergoes further adaptions, and expresses a receptor to bind the complement regulatory protein factor H on its surface to sabotage complement-mediated opsonization and phagocytosis (Kumwenda et al. 2022).

Bacteria are also known to sense and adapt to human sex hormones, which is associated with differences in infection rates and disease outcomes between genders (Garcia-Gomez et al. 2013, Vom Steeg and Klein 2017, Vidaillac et al. 2020). Pseuromonas aeruginosa exposure to different estrogens proved to increase bacterial motility—a key requisite for initiation of biofilm formation—as well as biofilm formation, adherence, and invasiveness of the bronchial epithelium (Tyrrell and Harvey 2020). In female cystic fibrosis patients, exacerbations have been associated with the follicular phase of the menstrual cycle, where estrogen levels are increased. Estrogens (estriol and estradiol) can drive alginate production in P. aeruginosa, a polysaccharide involved in bacterial pathogenicity, parenchymal damage, and the mucoid morphology (Chotirmall et al. 2012). The transition to the mucoid morphology is linked to loss-of-function mutations in the mucA gene, the main regulator of alginate production, which occurs upon estrogen exposure (Chotirmall et al. 2012). Chlamydia trachomatis exposure to sex hormones influences its transcription, and chlamydial infection outcome has been associated with the hormonal status of the epithelium. Estradiol treatment in vitro induces a transcriptional pattern suggestive of a C. trachomatis persister strategy, while progesterone induces a pattern rather resembling an active infection (Amirshahi et al. 2011).

Host amine hormones, such as adrenaline or noradrenaline, can also elicit microbial responses. The ability to sense these neurotransmitters facilitates bacterial adaptation to changing host environments, and is mediated via quorum-sensing systems, which induce a synchronized response to efficiently avoid the host defences (Karavolos et al. 2013). Bacteria were shown to directly bind adrenaline and noradrenaline through the QseC sensor kinase, a part of the autoinducer-3 (AI-3) quorum-sensing system, leading to expression of virulence genes in enterohemorrhagic E. coli O157:H7. Further, qseC-deficient mutants exhibit a reduced virulence, highlighting the importance of this signalling system during host–pathogen interactions. Interestingly, the periplasmic sensing domain of QseC is conserved among several bacterial species, as well as homologue to a fungal protein of unknown function in Aspergillus nidulans (Clarke et al. 2006). Another putative adrenaline receptor, the BasSR two-component signal transduction system, was identified in S. typhimurium. Adrenaline signalling through this receptor reduces antimicrobial peptide resistance of the bacteria. However, sensing of adrenaline also induces expression of oxidative stress resistance genes (such as sodA), suggesting that adrenaline sensing may serve as a cue for upcoming host-imposed stresses (Karavolos et al. 2008). Conversely, sensing of serotonin by the receptor CpxA, commonly found in enteric bacteria, can reduce virulence of enterohemorrhagic E. coli and Citrobacter rodentium (Kumar et al. 2020), possibly serving as a surrogate for a resistant host status, where engagement of virulence would be detrimental for the microorganism.

Conclusions and future perspectives

The success of pathogenic bacteria and fungi can be linked to their ability to adapt to the host and escape host defence mechanisms. Nevertheless, apart from a handful of primary pathogens that can cause infections at low inocula, human pathogens are highly restricted in causing infection due to protective layers of innate and adaptive immune defences. Among viruses, the manipulation of host responses is widespread, yet as outlined in this review, it has become evident that also bacteria and fungi can adapt to and anticipate immune activation by sensing and responding to immune mediators. Interestingly, microbial ability to bind/sense host cytokines is extensively distributed among regular commensal microorganisms, such as E. coli or S. aureus, suggesting that these mechanisms could be a consequence of host–microbe co-evolution. This cross-kingdom sensing facilitates metabolic adaptation to overcome nutritional immunity, PAMP masking to evade immune recognition, defensive and offensive pathogenicity strategies that facilitate the escape from innate immune cells, as well as the formation of biofilms that aid persistence in the host. Microbial adaptations in response to inflammatory mediators may offer an additional explanation to why chronic inflammatory diseases, such as inflammatory bowel disease or diabetes, are associated with drastic changes in the microbiome (Standaert-Vitse et al. 2009, Gosiewski et al. 2014, Wheeler et al. 2017).

However, microbial sensing of the immune status should also be considered with the increasing use of immunotherapeutic and immunomodulatory strategies used in the clinics. A potential example of this is the anti-PD-1 therapy. This immune checkpoint inhibitor boosts levels of IFN-γ, leading to M. tuberculosis reactivation. It is not clear if the described sensing of IFN-γ by M. tuberculosis (Ahmed et al. 2022) plays a direct role, but it is an intriguing possibility. Similarly, PD-1 inhibition also increases Th17 responses (Dulos et al. 2012) and can trigger allergic bronchopulmonary aspergillosis (ABPA) (Pradere et al. 2017, Donato and Krol 2019). Here, it also is unclear whether IL-17 sensing by A. fumigatus contributes (Zelante et al. 2012), yet Th17 responses are also associated with acute ABPA (Bacher et al. 2019).

Adaptation in response to immune mediators provides pathogens with the key advantage of tailoring pathogenicity mechanisms to the host immune status. An in-depth understanding of the molecular mechanisms driving adaptive pathogenicity strategies could reveal potential targets for therapeutic strategies against virulence and pathogenicity (Siscar-Lewin et al. 2019). In contrast to conventional antimicrobial therapy, which often affects the growth and survival of large groups of microorganisms, targeting virulence specifically would circumvent eradicating opportunistic pathogens as long as they are still part of a beneficial microbiome (Siscar-Lewin et al. 2019, Alonso-Monge et al. 2021).

Acknowledgements

The authors acknowledge Sophie Austermeier for feedback on early drafts of the manuscript.

Conflict of interest

The authors have no conflict of interest to declare.

Funding

This work was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) Emmy Noether Program [project no. 434385622/GR 5617/1-1 to M.S.G.]. We further acknowledge funding of this project from the Carl Zeiss Stiftung [to M.S.G.] and by an Exploration Grant of the Boehringer Ingelheim Foundation (BIS) [to M.S.G.].

References

Ahlstrand
 
T
,
Kovesjoki
 
L
,
Maula
 
T
 et al.  
Aggregatibacter actinomycetemcomitans LPS binds human interleukin-8
.
J Oral Microbiol
.
2019
;
11
:
1549931
.

Ahlstrand
 
T
,
Tuominen
 
H
,
Beklen
 
A
 et al.  
A novel intrinsically disordered outer membrane lipoprotein of Aggregatibacter actinomycetemcomitans binds various cytokines and plays a role in biofilm response to interleukin-1beta and interleukin-8
.
Virulence
.
2017
;
8
:
115
34
.

Ahmed
 
M
,
Mackenzie
 
J
,
Tezera
 
L
 et al.  
Mycobacterium tuberculosis senses host interferon-gamma via the membrane protein MmpL10
.
Commun Biol
.
2022
;
5
:
1317
.

Akdis
 
M
,
Aab
 
A
,
Altunbulakli
 
C
 et al.  
Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: receptors, functions, and roles in diseases
.
J Allergy Clin Immunol
.
2016
;
138
:
984
1010
.

Alcami
 
A
,
Koszinowski
 
UH
.
Viral mechanisms of immune evasion
.
Trends Microbiol
.
2000
;
8
:
410
8
.

Ali
 
A
,
Rautemaa
 
R
,
Hietanen
 
J
 et al.  
Expression of interleukin-8 and its receptor IL-8RA in chronic hyperplastic candidosis
.
Oral Microbiol Immunol
.
2006
;
21
:
223
30
.

Allert
 
S
,
Forster
 
TM
,
Svensson
 
CM
 et al.  
Candida albicans-induced epithelial damage mediates translocation through intestinal barriers
.
mBio
.
2018
;
9
:
e00915
18
.

Almeida
 
RS
.
The hyphal-associated adhesin and invasin Als3 of Candida albicans mediates iron acquisition from host ferritin
.
PLoS Pathog
.
2008
;
4
:
e1000217
.

Almeida
 
RS
.
Candida albicans iron acquisition within the host
.
FEMS Yeast Res
.
2009
;
9
:
1000
12
.

Alonso-Monge
 
R
,
Gresnigt
 
MS
,
Roman
 
E
 et al.  
Candida albicans colonization of the gastrointestinal tract: a double-edged sword
.
PLoS Pathog
.
2021
;
17
:
e1009710
.

Alonso-Monge
 
R
,
Navarro-Garcia
 
F
,
Roman
 
E
 et al.  
The Hog1 mitogen-activated protein kinase is essential in the oxidative stress response and chlamydospore formation in Candida albicans
.
Euk Cell
.
2003
;
2
:
351
61
.

Alvarez-de Miranda
 
FJ
,
Alonso-Sánchez
 
I
,
Alcamí
 
A
 et al.  
TNF decoy receptors encoded by poxviruses
.
Pathogens
.
2021
;
10
:
1065
.

Alves
 
R
,
Barata-Antunes
 
C
,
Casal
 
M
 et al.  
Adapting to survive: how Candida overcomes host-imposed constraints during human colonization
.
PLoS Pathog
.
2020
;
16
:
e1008478
.

Amirshahi
 
A
,
Wan
 
C
,
Beagley
 
K
 et al.  
Modulation of the Chlamydia trachomatis in vitro transcriptome response by the sex hormones estradiol and progesterone
.
BMC Microbiol
.
2011
;
11
:
150
.

Austermeier
 
S
,
Kasper
 
L
,
Westman
 
J
 et al.  
I want to break free—macrophage strategies to recognize and kill Candida albicans, and fungal counter-strategies to escape
.
Curr Opin Microbiol
.
2020
;
58
:
15
23
.

Azevedo
 
MLV
,
Zanchettin
 
AC
,
Vaz de Paula
 
CB
 et al.  
Lung neutrophilic recruitment and IL-8/IL-17A tissue expression in COVID-19
.
Front Immunol
.
2021
;
12
:
656350
.

Bacher
 
P
,
Hohnstein
 
T
,
Beerbaum
 
E
 et al.  
Human anti-fungal Th17 immunity and pathology rely on cross-reactivity against Candida albicans
.
Cell
.
2019
;
176
:
1340
55
.

Baker
 
RE
,
Mahmud
 
AS
,
Miller
 
IF
 et al.  
Infectious disease in an era of global change
.
Nat Rev Micro
.
2022
;
20
:
193
205
.

Bear
 
A
,
Locke
 
T
,
Rowland-Jones
 
S
 et al.  
The immune evasion roles of Staphylococcus aureus protein A and impact on vaccine development
.
Front Cell Infect Microbiol
.
2023
;
13
:
1242702
.

Bereshchenko
 
O
,
Bruscoli
 
S
,
Riccardi
 
C
.
Glucocorticoids, sex hormones, and immunity
.
Front Immunol
.
2018
;
9
:
1332
.

Bertheloot
 
D
.
HMGB1, IL-1α, IL-33 and S100 proteins: dual-function alarmins
.
Cell Mol Immunol
.
2017
;
14
:
43
64
.

Bevins
 
CL
,
Salzman
 
NH
.
Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis
.
Nat Rev Micro
.
2011
;
9
:
356
68
.

Bhattacharya
 
M
,
Horswill
 
AR
.
The role of human extracellular matrix proteins in defining Staphylococcus aureus biofilm infections
.
FEMS Microbiol Rev
.
2024
;
48
:
fuae002
.

Bliska
 
JB
,
Casadevall
 
A
.
Intracellular pathogenic bacteria and fungi—a case of convergent evolution?
.
Nat Rev Micro
.
2009
;
7
:
165
71
.

Born
 
TL
,
Morrison
 
LA
,
Esteban
 
DJ
 et al.  
A poxvirus protein that binds to and inactivates IL-18, and inhibits NK cell response1
.
J Immunol
.
2000
;
164
:
3246
54
.

Brown
 
AJP
,
Gow
 
NAR
,
Warris
 
A
 et al.  
Memory in fungal pathogens promotes immune evasion, colonisation, and infection
.
Trends Microbiol
.
2019
;
27
:
219
30
.

Brunke
 
S
,
Hube
 
B
.
Adaptive prediction as a strategy in microbial infections
.
PLoS Pathog
.
2014
;
10
:
e1004356
.

Caffrey
 
AK
.
Alarmin(g) the innate immune system to invasive fungal infections
.
Curr Opin Microbiol
.
2016
;
32
:
135
43
.

Cao
 
M
,
Goodrich-Blair
 
H
.
Ready or not: microbial adaptive responses in dynamic symbiosis environments
.
J Bacteriol
.
2017
;
199
:
e00883
16
.

Casadevall
 
A
.
‘Ready made’ virulence and ‘dual use’ virulence factors in pathogenic environmental fungi—the Cryptococcus neoformans paradigm
.
Curr Opin Microbiol
.
2003
;
6
:
332
7
.

Casadevall
 
A
.
Climate change brings the specter of new infectious diseases
.
J Clin Invest
.
2020
;
130
:
553
5
.

Casadevall
 
A
.
Environmental Candida auris and the global warming emergence hypothesis
.
mBio
.
2021
;
12
:
1
3
.

Casadevall
 
A
.
Evolution of intracellular pathogens
.
Annu Rev Microbiol
.
2008
;
62
:
19
33
.

Case
 
NT
,
Duah
 
K
,
Larsen
 
B
 et al.  
The macrophage-derived protein PTMA induces filamentation of the human fungal pathogen Candida albicans
.
Cell Rep
.
2021
;
36
:
109584
.

Cebo
 
C
,
Vergoten
 
G
,
Zanetta
 
J-P
.
Lectin activities of cytokines: functions and putative carbohydrate-recognition domains
.
Biochim Biophys Acta
.
2002
;
1572
:
422
34
.

Chen
 
C
,
Itakura
 
E
,
Nelson
 
GM
 et al.  
IL-17 is a neuromodulator of Caenorhabditis elegans sensory responses
.
Nature
.
2017
;
542
:
43
8
.

Childers
 
DS
,
Avelar
 
GM
,
Bain
 
JM
 et al.  
Impact of the environment upon the Candida albicans cell wall and resultant effects upon immune surveillance
.
Curr Top Microbiol Immunol
.
2020
;
425
:
297
330
.

Chotirmall
 
SH
,
Smith
 
SG
,
Gunaratnam
 
C
 et al.  
Effect of estrogen on pseudomonas mucoidy and exacerbations in cystic fibrosis
.
N Engl J Med
.
2012
;
366
:
1978
86
.

Citiulo
 
F
.
Candida albicans scavenges host zinc via Pra1 during endothelial invasion
.
PLoS Pathog
.
2012
;
8
:
e1002777
.

Clark
 
C
,
Drummond
 
RA
.
The hidden cost of modern medical interventions: how medical advances have shaped the prevalence of human fungal disease
.
Pathogens
.
2019
;
8
:
45
.

Clarke
 
MB
,
Hughes
 
DT
,
Zhu
 
C
 et al.  
The QseC sensor kinase: a bacterial adrenergic receptor
.
Proc Natl Acad Sci USA
.
2006
;
103
:
10420
5
.

Corbera-Bellalta
 
M
,
Planas-Rigol
 
E
,
Lozano
 
E
 et al.  
Blocking interferon gamma reduces expression of chemokines CXCL9, CXCL10 and CXCL11 and decreases macrophage infiltration in ex vivo cultured arteries from patients with giant cell arteritis
.
Ann Rheum Dis
.
2016
;
75
:
1177
86
.

Debeuf
 
N
.
Eicosanoid control over antigen presenting cells in asthma
.
Front Immunol
.
2018
;
9
:
2006
.

Denis
 
M
,
Gregg
 
EO
.
Recombinant interleukin-6 increases the intracellular and extracellular growth of Mycobacterium avium
.
Can J Microbiol
.
1991
;
37
:
479
83
.

Denis
 
M
,
Campbell
 
D
,
Gregg
 
EO
.
Interleukin-2 and granulocyte-macrophage colony-stimulating factor stimulate growth of a virulent strain of Escherichia coli
.
Infect Immun
.
1991
;
59
:
1853
6
.

Dennis
 
EA
,
Norris
 
PC
.
Eicosanoid storm in infection and inflammation
.
Nat Rev Immunol
.
2015
;
15
:
511
23
.

Donato
 
AA
,
Krol
 
R
.
Allergic bronchopulmonary aspergillosis presumably unmasked by PD-1 inhibition
.
BMJ Case Rep
.
2019
;
12
:
e227814
.

Drummond
 
RA
,
Swamydas
 
M
,
Oikonomou
 
V
 et al.  
CARD9(+) microglia promote antifungal immunity via IL-1β- and CXCL1-mediated neutrophil recruitment
.
Nat Immunol
.
2019
;
20
:
559
70
.

Duensing
 
TD
,
Wing
 
JS
,
van Putten
 
JP
.
Sulfated polysaccharide-directed recruitment of mammalian host proteins: a novel strategy in microbial pathogenesis
.
Infect Immun
.
1999
;
67
:
4463
8
.

Dulos
 
J
,
Carven
 
GJ
,
van Boxtel
 
SJ
 et al.  
PD-1 blockade augments Th1 and Th17 and suppresses Th2 responses in peripheral blood from patients with prostate and advanced melanoma cancer
.
J Immunother
.
2012
;
35
:
169
78
.

Dyakov
 
IN
,
Mavletova
 
DA
,
Chernyshova
 
IN
 et al.  
FN3 protein fragment containing two type III fibronectin domains from B. longum GT15 binds to human tumor necrosis factor alpha in vitro
.
Anaerobe
.
2020
;
65
:
102247
.

Egesten
 
A
,
Frick
 
IM
,
Morgelin
 
M
 et al.  
Binding of albumin promotes bacterial survival at the epithelial surface
.
J Biol Chem
.
2011
;
286
:
2469
76
.

Ells
 
R
,
Kock
 
JL
,
Albertyn
 
J
 et al.  
Effect of inhibitors of arachidonic acid metabolism on prostaglandin E(2) production by Candida albicans and Candida dubliniensis biofilms
.
Med Microbiol Immunol
.
2011
;
200
:
23
28
.

Engelsoy
 
U
,
Rangel
 
I
,
Demirel
 
I
.
Impact of proinflammatory cytokines on the virulence of uropathogenic Escherichia coli
.
Front Microbiol
.
2019
;
10
:
1051
.

Erb-Downward
 
JR
,
Noverr
 
MC
.
Characterization of prostaglandin E2 production by Candida albicans
.
Infect Immun
.
2007
;
75
:
3498
505
.

Erwig
 
LP
,
Gow
 
NA
.
Interactions of fungal pathogens with phagocytes
.
Nat Rev Micro
.
2016
;
14
:
163
76
.

Fischer
 
J
,
Gresnigt
 
MS
,
Werz
 
O
 et al.  
Candida albicans-induced leukotriene biosynthesis in neutrophils is restricted to the hyphal morphology
.
FASEB J
.
2021
;
35
:
e21820
.

Foster
 
TJ
.
Immune evasion by staphylococci
.
Nat Rev Micro
.
2005
;
3
:
948
58
.

Frick
 
I-M
.
Constitutive and inflammation-dependent antimicrobial peptides produced by epithelium are differentially processed and inactivated by the commensal Finegoldia magna and the pathogen Streptococcus pyogenes
.
J Immunol
.
2011
;
187
:
4300
9
.

Fukushima
 
K
,
Hara-Kuge
 
S
,
Ohkura
 
T
 et al.  
Lectin-like characteristics of recombinant human interleukin-1beta recognizing glycans of the glycosylphosphatidylinositol anchor
.
J Biol Chem
.
1997
;
272
:
10579
84
.

Garcia-Gomez
 
E
,
Gonzalez-Pedrajo
 
B
,
Camacho-Arroyo
 
I
.
Role of sex steroid hormones in bacterial-host interactions
.
Biomed Res Int
.
2013
;
2013
:
1
.

Gendrin
 
C
,
Sarrazin
 
S
,
Bonnaffe
 
D
 et al.  
Hijacking of the pleiotropic cytokine interferon-gamma by the type III secretion system of Yersinia pestis
.
PLoS One
.
2010
;
5
:
e15242
.

Gosiewski
 
T
,
Salamon
 
D
,
Szopa
 
M
 et al.  
Quantitative evaluation of fungi of the genus Candida in the feces of adult patients with type 1 and 2 diabetes—a pilot study
.
Gut Pathog
.
2014
;
6
:
43
.

Gross
 
O
,
Poeck
 
H
,
Bscheider
 
M
 et al.  
Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence
.
Nature
.
2009
;
459
:
433
6
.

Gunn
 
MD
,
Nelken
 
NA
,
Liao
 
X
 et al.  
Monocyte chemoattractant protein-1 is sufficient for the chemotaxis of monocytes and lymphocytes in transgenic mice but requires an additional stimulus for inflammatory activation
.
J Immunol
.
1997
;
158
:
376
83
.

Gutierrez Jauregui
 
R
,
Fleige
 
H
,
Bubke
 
A
 et al.  
IL-1beta promotes Staphylococcus aureus biofilms on implants in vivo
.
Front Immunol
.
2019
;
10
:
1082
.

Hernaez
 
B
,
Alcamí
 
A
.
Virus-encoded cytokine and chemokine decoy receptors
.
Curr Opin Immunol
.
2020
;
66
:
50
6
.

Hmama
 
Z
,
Pena-Diaz
 
S
,
Joseph
 
S
 et al.  
Immunoevasion and immunosuppression of the macrophage by Mycobacterium tuberculosis
.
Immunol Rev
.
2015
;
264
:
220
32
.

Hogbom
 
M
,
Ihalin
 
R
.
Functional and structural characteristics of bacterial proteins that bind host cytokines
.
Virulence
.
2017
;
8
:
1592
601
.

Hood
 
MI
.
Nutritional immunity: transition metals at the pathogen–host interface
.
Nat Rev Micro
.
2012
;
10
:
525
37
.

Hopke
 
A
,
Brown
 
AJP
,
Hall
 
RA&
 et al.  
Dynamic fungal cell wall architecture in stress adaptation and immune evasion
.
Trends Microbiol
.
2018
;
26
:
284
95
.

Hopke
 
A
,
Scherer
 
A
,
Kreuzburg
 
S
 et al.  
Neutrophil swarming delays the growth of clusters of pathogenic fungi
.
Nat Commun
.
2020
;
11
:
2031
.

Hovingh
 
ES
,
van den Broek
 
B
,
Jongerius
 
I
.
Hijacking complement regulatory proteins for bacterial immune evasion
.
Front Microbiol
.
2016
;
7
:
2004
.

Huang
 
J
,
Hu
 
P
,
Ye
 
L
 et al.  
Pan-drug resistance and hypervirulence in a human fungal pathogen are enabled by mutagenesis induced by mammalian body temperature
.
Nat Microbiol
.
2024
;
9
:
1686
99
.

Hube
 
B
.
Fungal adaptation to the host environment
.
Curr Opin Microbiol
.
2009
;
12
:
347
9
.

Hultgren
 
O
,
Kopf
 
M
,
Tarkowski
 
A
.
Staphylococcus aureus-induced septic arthritis and septic death is decreased in IL-4-deficient mice: role of IL-4 as promoter for bacterial growth
.
J Immunol
.
1998
;
160
:
5082
7
.

Jaeger
 
M
,
Dietschmann
 
A
,
Austermeier
 
S
 et al.  
Alpha1-antitrypsin impacts innate host–pathogen interactions with Candida albicans by stimulating fungal filamentation
.
Virulence
.
2024
;
15
:
2333367
.

Kalo-Klein
 
A
,
Witkin
 
SS
.
Prostaglandin E2 enhances and gamma interferon inhibits germ tube formation in Candida albicans
.
Infect Immun
.
1990
;
58
:
260
2
.

Kamada
 
N
,
Kim
 
YG
,
Sham
 
HP
 et al.  
Regulated virulence controls the ability of a pathogen to compete with the gut microbiota
.
Science
.
2012
;
336
:
1325
9
.

Kammer
 
P
,
McNamara
 
S
,
Wolf
 
T
 et al.  
Survival strategies of pathogenic Candida species in human blood show independent and specific adaptations
.
mBio
.
2020
;
11
:
e02435
20
.

Kanangat
 
S
,
Postlethwaite
 
A
,
Cholera
 
S
 et al.  
Modulation of virulence gene expression in Staphylococcus aureus by interleukin-1beta: novel implications in bacterial pathogenesis
.
Microbes Infect
.
2007
;
9
:
408
15
.

Kanda
 
N
,
Shimizu
 
T
,
Tada
 
Y
 et al.  
IL-18 enhances IFN-gamma-induced production of CXCL9, CXCL10, and CXCL11 in human keratinocytes
.
Eur J Immunol
.
2007
;
37
:
338
50
.

Karavolos
 
MH
,
Spencer
 
H
,
Bulmer
 
DM
 et al.  
Adrenaline modulates the global transcriptional profile of Salmonella revealing a role in the antimicrobial peptide and oxidative stress resistance responses
.
BMC Genom
.
2008
;
9
:
458
.

Karavolos
 
MH
,
Winzer
 
K
,
Williams
 
P
 et al.  
Pathogen espionage: multiple bacterial adrenergic sensors eavesdrop on host communication systems
.
Mol Microbiol
.
2013
;
87
:
455
65
.

Kasper
 
L
,
Konig
 
A
,
Koenig
 
PA
 et al.  
The fungal peptide toxin candidalysin activates the NLRP3 inflammasome and causes cytolysis in mononuclear phagocytes
.
Nat Commun
.
2018
;
9
:
4260
.

Kaya
 
E
,
Grassi
 
L
,
Benedetti
 
A
 et al.  
In vitro interaction of Pseudomonas aeruginosa biofilms with human peripheral blood mononuclear cells
.
Front Cell Infect Microbiol
.
2020
;
10
:
187
.

Kotwal
 
GJ
.
Poxviral mimicry of complement and chemokine system components: what’s the end game?
.
Immunol Today
.
2000
;
21
:
242
8
.

Kreitmann
 
L
,
Helms
 
J
,
Martin-Loeches
 
I
 et al.  
ICU-acquired infections in immunocompromised patients
.
Intensive Care Med
.
2024
;
50
:
332
49
.

Krupa
 
A
,
Fol
 
M
,
Dziadek
 
BR
 et al.  
Binding of CXCL8/IL-8 to Mycobacterium tuberculosis modulates the innate immune response
.
Mediators Inflamm
.
2015
;
2015
:
124762
.

Kullberg
 
BJ
,
Arendrup
 
MC
.
Invasive candidiasis
.
N Engl J Med
.
2015
;
373
:
1445
56
.

Kumamoto
 
CA
,
Gresnigt
 
MS
,
Hube
 
B
.
The gut, the bad and the harmless: Candida albicans as a commensal and opportunistic pathogen in the intestine
.
Curr Opin Microbiol
.
2020
;
56
:
7
15
.

Kumar
 
A
,
Russell
 
RM
,
Pifer
 
R
 et al.  
The serotonin neurotransmitter modulates virulence of enteric pathogens
.
Cell Host Microbe
.
2020
;
28
:
41
53
.

Kumwenda
 
P
,
Cottier
 
F
,
Hendry
 
AC
 et al.  
Estrogen promotes innate immune evasion of Candida albicans through inactivation of the alternative complement system
.
Cell Rep
.
2022
;
38
:
110183
.

Kundu
 
G
,
Noverr
 
MC
.
Exposure to host or fungal PGE(2) abrogates protection following immunization with Candida-pulsed dendritic cells
.
Med Mycol
.
2011
;
49
:
380
94
.

Kurakado
 
S
,
Kurogane
 
R
,
Sugita
 
T
.
17β-Estradiol inhibits estrogen binding protein-mediated hypha formation in Candida albicans
.
Microb Pathog
.
2017
;
109
:
151
5
.

Lange
 
T
,
Kasper
 
L
,
Gresnigt
 
MS
 et al.  
“Under Pressure”—how fungi evade, exploit, and modulate cells of the innate immune system
.
Semin Immunol
.
2023
;
66
:
101738
.

Lee
 
JH
,
Del Sorbo
 
L
,
Khine
 
AA
 et al.  
Modulation of bacterial growth by tumor necrosis factor-alpha in vitro and in vivo
.
Am J Respir Crit Care Med
.
2003a
;
168
:
1462
70
.

Lee
 
WL
,
Harrison
 
RE
,
Grinstein
 
S
.
Phagocytosis by neutrophils
.
Microbes Infect
.
2003b
;
5
:
1299
306
.

Lionakis
 
MS
,
Lim
 
JK
,
Lee
 
CC&
 et al.  
Organ-specific innate immune responses in a mouse model of invasive candidiasis
.
J Innate Immun
.
2011
;
3
:
180
99
.

Lopes
 
JP
,
Lionakis
 
MS
.
Pathogenesis and virulence of Candida albicans
.
Virulence
.
2022
;
13
:
89
121
.

Lu
 
LL
.
Beyond binding: antibody effector functions in infectious diseases
.
Nat Rev Immunol
.
2018
;
18
:
46
61
.

Luo
 
G
,
Niesel
 
DW
,
Shaban
 
RA
 et al.  
Tumor necrosis factor alpha binding to bacteria: evidence for a high-affinity receptor and alteration of bacterial virulence properties
.
Infect Immun
.
1993
;
61
:
830
5
.

Ma
 
J
,
Zhang
 
YG
,
Xia
 
Y
 et al.  
The inflammatory cytokine tumor necrosis factor modulates the expression of Salmonella typhimurium effector proteins
.
J Inflamm
.
2010
;
7
:
42
.

McLaughlin
 
RA
,
Hoogewerf
 
AJ
.
Interleukin-1beta-induced growth enhancement of Staphylococcus aureus occurs in biofilm but not planktonic cultures
.
Microb Pathog
.
2006
;
41
:
67
79
.

McLoughlin
 
RM
,
Lee
 
JC
,
Kasper
 
DL
 et al.  
IFN-gamma regulated chemokine production determines the outcome of Staphylococcus aureus infection
.
J Immunol
.
2008
;
181
:
1323
32
.

Madani
 
ND
,
Malloy
 
PJ
,
Rodriguez-Pombo
 
P
 et al.  
Candida albicans estrogen-binding protein gene encodes an oxidoreductase that is inhibited by estradiol
.
Proc Natl Acad Sci USA
.
1994
;
91
:
922
6
.

Mahdavi
 
J
,
Royer
 
PJ
,
Sjolinder
 
HS
 et al.  
Pro-inflammatory cytokines can act as intracellular modulators of commensal bacterial virulence
.
Open Biol
.
2013
;
3
:
130048
.

Mantovani
 
A
,
Dinarello
 
CA
,
Molgora
 
M
 et al.  
Interleukin-1 and related cytokines in the regulation of inflammation and immunity
.
Immunity
.
2019
;
50
:
778
95
.

Martins
 
R
,
Maier
 
J
,
Gorki
 
AD
 et al.  
Heme drives hemolysis-induced susceptibility to infection via disruption of phagocyte functions
.
Nat Immunol
.
2016
;
17
:
1361
72
.

Matsushima
 
K
,
Yang
 
D
,
Oppenheim
 
JJ
.
Interleukin-8: an evolving chemokine
.
Cytokine
.
2022
;
153
:
155828
.

Meduri
 
GU
,
Kanangat
 
S
,
Stefan
 
J
 et al.  
Cytokines IL-1beta, IL-6, and TNF-alpha enhance in vitro growth of bacteria
.
Am J Respir Crit Care Med
.
1999
;
160
:
961
7
.

Meri
 
T
,
Amdahl
 
H
,
Lehtinen
 
MJ
 et al.  
Microbes bind complement inhibitor factor H via a common site
.
PLoS Pathog
.
2013
;
9
:
e1003308
.

Mitchell
 
A
,
Romano
 
GH
,
Groisman
 
B
 et al.  
Adaptive prediction of environmental changes by microorganisms
.
Nature
.
2009
;
460
:
220
4
.

Montano
 
DE
,
Hartung
 
S
,
Wich
 
M
 et al.  
The TLR-NF-kB axis contributes to the monocytic inflammatory response against a virulent strain of Lichtheimia corymbifera, a causative agent of invasive mucormycosis
.
Front Immunol
.
2022
;
13
:
882921
.

Moore
 
SR
,
Menon
 
SS
,
Cortes
 
C
 et al.  
Hijacking factor H for complement immune evasion
.
Front Immunol
.
2021
;
12
:
602277
.

Moriel
 
DG
,
Heras
 
B
,
Paxman
 
JJ
 et al.  
Molecular and structural characterization of a novel Escherichia coli interleukin receptor mimic protein
.
mBio
.
2016
;
7
:
e02046
.

Moyes
 
DL
,
Wilson
 
D
,
Richardson
 
JP
 et al.  
Candidalysin is a fungal peptide toxin critical for mucosal infection
.
Nature
.
2016
;
532
:
64
8
.

Mylonakis
 
E
.
Killing of Caenorhabditis elegans by Cryptococcus neoformans as a model of yeast pathogenesis
.
Proc Natl Acad Sci USA
.
2002
;
99
:
15675
80
.

Naglik
 
JR
,
Konig
 
A
,
Hube
 
B
 et al.  
Candida albicans-epithelial interactions and induction of mucosal innate immunity
.
Curr Opin Microbiol
.
2017
;
40
:
104
12
.

Naglik
 
JR
,
Moyes
 
D
.
Epithelial cell innate response to Candida albicans
.
Adv Dent Res
.
2011
;
23
:
50
5
.

Nakajima
 
R
,
Brubaker
 
RR
.
Association between virulence of Yersinia pestis and suppression of gamma interferon and tumor necrosis factor alpha
.
Infect Immun
.
1993
;
61
:
23
31
.

Nakajima
 
R
,
Motin
 
VL
,
Brubaker
 
RR
.
Suppression of cytokines in mice by protein A-V antigen fusion peptide and restoration of synthesis by active immunization
.
Infect Immun
.
1995
;
63
:
3021
9
.

Nenciarini
 
S
,
Renzi
 
S
,
di Paola
 
M
 et al.  
The yeast–human coevolution: fungal transition from passengers, colonizers, and invaders
.
WIREs Mech Dis
.
2024
;
16
:
e1639
.

Neutra
 
MR
,
Pringault
 
E
,
Kraehenbuhl
 
JP
.
Antigen sampling across epithelial barriers and induction of mucosal immune responses
.
Annu Rev Immunol
.
1996
;
14
:
275
300
.

Noverr
 
MC
,
Cox
 
GM
,
Perfect
 
JR
 et al.  
Role of PLB1 in pulmonary inflammation and cryptococcal eicosanoid production
.
Infect Immun
.
2003
;
71
:
1538
47
.

Noverr
 
MC
,
Huffnagle
 
GB
.
Regulation of Candida albicans morphogenesis by fatty acid metabolites
.
Infect Immun
.
2004
;
72
:
6206
10
.

Noverr
 
MC
,
Phare
 
SM
,
Toews
 
GB
 et al.  
Pathogenic yeasts Cryptococcus neoformans and Candida albicans produce immunomodulatory prostaglandins
.
Infect Immun
.
2001
;
69
:
2957
63
.

Noverr
 
MC
,
Toews
 
GB
,
Huffnagle
 
GB
.
Production of prostaglandins and leukotrienes by pathogenic fungi
.
Infect Immun
.
2002
;
70
:
400
2
.

Núñez
 
G
,
Sakamoto
 
K
,
Soares
 
MP
.
Innate nutritional immunity
.
J Immunol
.
2018
;
201
:
11
8
.

Paino
 
A
,
Ahlstrand
 
T
,
Nuutila
 
J
 et al.  
Identification of a novel bacterial outer membrane interleukin-1Beta-binding protein from Aggregatibacter actinomycetemcomitans
.
PLoS One
.
2013
;
8
:
e70509
.

Pardini
 
G
,
De Groot
 
PW
,
Coste
 
AT
 et al.  
The CRH family coding for cell wall glycosylphosphatidylinositol proteins with a predicted transglycosidase domain affects cell wall organization and virulence of Candida albicans
.
J Biol Chem
.
2006
;
281
:
40399
411
.

Parnian
 
R
,
Heydarifard
 
F
,
Mousavi
 
FS
 et al.  
Innate immune response to monkeypox virus infection: mechanisms and immune escape
.
J Innate Immun
.
2024
;
16
:
413
24
.

Pauwels
 
A-M
.
Patterns, receptors, and signals: regulation of phagosome maturation
.
Trends Immunol
.
2017
;
38
:
407
22
.

Peck
 
JR
,
Waxman
 
D
.
What is adaptation and how should it be measured?
.
J Theor Biol
.
2018
;
447
:
190
8
.

Pekmezovic
 
M
,
Kaune
 
AK
,
Austermeier
 
S
 et al.  
Human albumin enhances the pathogenic potential of Candida glabrata on vaginal epithelial cells
.
PLoS Pathog
.
2021
;
17
:
e1010037
.

Pittman
 
K
.
Damage-associated molecular patterns control neutrophil recruitment
.
J Innate Immun
.
2013
;
5
:
315
23
.

Porat
 
R
,
Clark
 
BD
,
Wolff
 
SM
 et al.  
Enhancement of growth of virulent strains of Escherichia coli by interleukin-1
.
Science
.
1991
;
254
:
430
2
.

Potian
 
JA
.
Preexisting helminth infection induces inhibition of innate pulmonary anti-tuberculosis defense by engaging the IL-4 receptor pathway
.
J Exp Med
.
2011
;
208
:
1863
74
.

Pradere
 
P
,
Michot
 
JM
,
Champiat
 
S
 et al.  
Allergic broncho-pulmonary aspergillosis following treatment with an anti-programmed cell death protein 1 monoclonal antibody therapy
.
Eur J Cancer
.
2017
;
75
:
308
9
.

Pradhan
 
A
,
Avelar
 
GM
,
Bain
 
JM
 et al.  
Non-canonical signalling mediates changes in fungal cell wall PAMPs that drive immune evasion
.
Nat Commun
.
2019
;
10
:
5315
.

Quinn
 
GA
,
Tarwater
 
PM
,
Cole
 
AM
.
Subversion of interleukin-1-mediated host defence by a nasal carrier strain of Staphylococcus aureus
.
Immunology
.
2009
;
128
:
e222
229
.

Ramesh
 
G
,
MacLean
 
AG
,
Philipp
 
MT
.
Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration, and neuropathic pain
.
Mediators Inflamm
.
2013
;
2013
:
1
.

Ray
 
SC
,
Rappleye
 
CA
.
Flying under the radar: Histoplasma capsulatum avoidance of innate immune recognition
.
Semin Cell Dev Biol
.
2019
;
89
:
91
8
.

Reading
 
PC
,
Smith
 
GL
.
Vaccinia virus interleukin-18-binding protein promotes virulence by reducing gamma interferon production and natural killer and T-cell activity
.
J Virol
.
2003
;
77
:
9960
8
.

Reuter
 
M
,
Caswell
 
CC
,
Lukomski
 
S
 et al.  
Binding of the human complement regulators CFHR1 and factor H by streptococcal collagen-like protein 1 (Scl1) via their conserved C termini allows control of the complement cascade at multiple levels
.
J Biol Chem
.
2010
;
285
:
38473
85
.

Ristow
 
LC
,
Welch
 
RA
.
Hemolysin of uropathogenic Escherichia coli: a cloak or a dagger?
.
Biochim Biophys Acta
.
2016
;
1858
:
538
45
.

Rocha
 
FAC
,
Alves
 
A
,
Rocha
 
MFG
 et al.  
Tumor necrosis factor prevents Candida albicans biofilm formation
.
Sci Rep
.
2017
;
7
:
1206
.

Rodaki
 
A
,
Bohovych
 
IM
,
Enjalbert
 
B
 et al.  
Glucose promotes stress resistance in the fungal pathogen Candida albicans
.
MBoC
.
2009
;
20
:
4845
55
.

Rollauer
 
SE
,
Sooreshjani
 
MA
,
Noinaj
 
N
 et al.  
Outer membrane protein biogenesis in Gram-negative bacteria
.
Phil Trans R Soc B
.
2015
;
370
:
20150023
.

Satorres
 
SE
,
Alcaraz
 
LE
,
Cargnelutti
 
E
 et al.  
IFN-gamma plays a detrimental role in murine defense against nasal colonization of Staphylococcus aureus
.
Immunol Lett
.
2009
;
123
:
185
8
.

Savageau
 
MA
.
Demand theory of gene regulation. II. Quantitative application to the lactose and maltose operons of Escherichia coli
.
Genetics
.
1998
;
149
:
1677
91
.

Schimanski
 
J
,
Gresnigt
 
MS
,
Brunner
 
E
 et al.  
Hyphal-associated protein expression is crucial for Candida albicans-induced eicosanoid biosynthesis in immune cells
.
Eur J Immunol
.
2024
;
54
:
e2350743
.

Sherblom
 
AP
,
Sathyamoorthy
 
N
,
Decker
 
JM
 et al.  
IL-2, a lectin with specificity for high mannose glycopeptides
.
J Immunol
.
1989
;
143
:
939
44
.

Sherrington
 
SL
,
Kumwenda
 
P
,
Kousser
 
C
 et al.  
Host sensing by pathogenic fungi
.
Adv Appl Microbiol
.
2018
;
102
:
159
221
.

Shi
 
H
,
Tang
 
J
,
An
 
C
 et al.  
Protein A of Staphylococcus aureus strain NCTC8325 interacted with heparin
.
Arch Microbiol
.
2021
;
203
:
2563
73
.

Shinu
 
P
,
Mouslem
 
AKA
,
Nair
 
AB
 et al.  
Progress report: antimicrobial drug discovery in the resistance era
.
Pharmaceuticals
.
2022
;
15
:
413
.

Siscar-Lewin
 
S
,
Hube
 
B
,
Brunke
 
S
.
Antivirulence and avirulence genes in human pathogenic fungi
.
Virulence
.
2019
;
10
:
935
47
.

Smith
 
VP
,
Bryant
 
NA
,
Alcamí
 
A
.
Ectromelia, vaccinia and cowpox viruses encode secreted interleukin-18-binding proteins
.
J Gen Virol
.
2000
;
81
:
1223
30
.

Spaan
 
AN
,
Reyes-Robles
 
T
,
Badiou
 
C
 et al.  
Staphylococcus aureus targets the Duffy antigen receptor for chemokines (DARC) to lyse erythrocytes
.
Cell Host Microbe
.
2015
;
18
:
363
70
.

Stadnyk
 
AW
.
Intestinal epithelial cells as a source of inflammatory cytokines and chemokines
.
Can J Gastroenterol
.
2002
;
16
:
241
6
.

Standaert-Vitse
 
A
,
Sendid
 
B
,
Joossens
 
M
 et al.  
Candida albicans colonization and ASCA in familial Crohn’s disease
.
Am J Gastroenterol
.
2009
;
104
:
1745
53
.

Steenbergen
 
JN
,
Shuman
 
HA
,
Casadevall
 
A
.
Cryptococcus neoformans interactions with amoebae suggest an explanation for its virulence and intracellular pathogenic strategy in macrophages
.
Proc Natl Acad Sci USA
.
2001
;
98
:
15245
50
.

Taherkhani
 
S
,
Suzuki
 
K
,
Castell
 
L
.
A short overview of changes in inflammatory cytokines and oxidative stress in response to physical activity and antioxidant supplementation
.
Antioxidants (Basel)
.
2020
;
9
:
886
.

Tan
 
TG
,
Lim
 
YS
,
Tan
 
A
 et al.  
Fungal symbionts produce prostaglandin E(2) to promote their intestinal colonization
.
Front Cell Infect Microbiol
.
2019
;
9
:
359
.

Tang
 
LJ
,
De Seta
 
F
,
Odreman
 
F
 et al.  
Proteomic analysis of human cervical-vaginal fluids
.
J Proteome Res
.
2007
;
6
:
2874
83
.

Tekaia
 
F
.
Aspergillus fumigatus: saprophyte or pathogen?
.
Curr Opin Microbiol
.
2005
;
8
:
385
92
.

Thieme
 
N
,
Wu
 
VW
,
Dietschmann
 
A
 et al.  
The transcription factor PDR-1 is a multi-functional regulator and key component of pectin deconstruction and catabolism in Neurospora crassa
.
Biotechnol Biofuels
.
2017
;
10
:
149
.

Tomlin
 
H
,
Piccinini
 
AM
.
A complex interplay between the extracellular matrix and the innate immune response to microbial pathogens
.
Immunology
.
2018
;
155
:
186
201
.

Treseler
 
CB
,
Maziarz
 
RT
,
Levitz
 
SM
.
Biological activity of interleukin-2 bound to Candida albicans
.
Infect Immun
.
1992
;
60
:
183
8
.

Tsitsigiannis
 
DI
,
Bok
 
JW
,
Andes
 
D
 et al.  
Aspergillus cyclooxygenase-like enzymes are associated with prostaglandin production and virulence
.
Infect Immun
.
2005
;
73
:
4548
59
.

Tyrrell
 
J
,
Harvey
 
BJ
.
Sexual dimorphism in the microbiology of the CF ‘Gender Gap’: estrogen modulation of Pseudomonas aeruginosa virulence
.
Steroids
.
2020
;
156
:
108575
.

Vallon-Eberhard
 
A
,
Landsman
 
L
,
Yogev
 
N
 et al.  
Transepithelial pathogen uptake into the small intestinal lamina propria
.
J Immunol
.
2006
;
176
:
2465
9
.

van de Veerdonk
 
FL
,
Gresnigt
 
MS
,
Romani
 
L
 et al.  
Aspergillus fumigatus morphology and dynamic host interactions
.
Nat Rev Micro
.
2017
;
15
:
661
74
.

Vidaillac
 
C
,
Yong
 
VFL
,
Aschtgen
 
MS
 et al.  
Sex steroids induce membrane stress responses and virulence properties in Pseudomonas aeruginosa
.
mBio
.
2020
;
11
:
e01774
20
.

Vom Steeg
 
LG
,
Klein
 
SL
.
Sex steroids mediate bidirectional interactions between hosts and microbes
.
Horm Behav
.
2017
;
88
:
45
51
.

Wei
 
XQ
,
Leung
 
BP
,
Niedbala
 
W
 et al.  
Altered immune responses and susceptibility to Leishmania major and Staphylococcus aureus infection in IL-18-deficient mice
.
J Immunol
.
1999
;
163
:
2821
8
.

Wheeler
 
ML
,
Limon
 
JJ
,
Underhill
 
DM
.
Immunity to commensal fungi: detente and disease
.
Annu Rev Pathol Mech Dis
.
2017
;
12
:
359
85
.

Woytschak
 
J
.
Type 2 interleukin-4 receptor signaling in neutrophils antagonizes their expansion and migration during infection and inflammation
.
Immunity
.
2016
;
45
:
172
84
.

Wu
 
L
,
Estrada
 
O
,
Zaborina
 
O
 et al.  
Recognition of host immune activation by Pseudomonas aeruginosa
.
Science
.
2005
;
309
:
774
7
.

Wu
 
S
,
Zhang
 
T
,
Qiang
 
W
 et al.  
Modulation of immune responses in the central nervous system by Zika virus, West Nile virus, and dengue virus
.
Rev Med Virol
.
2024
;
34
:
e2535
.

Wu
 
VW
,
Thieme
 
N
,
Huberman
 
LB
 et al.  
The regulatory and transcriptional landscape associated with carbon utilization in a filamentous fungus
.
Proc Natl Acad Sci USA
.
2020
;
117
:
6003
13
.

Xiang
 
Y
,
Moss
 
B
.
IL-18 binding and inhibition of interferon γ induction by human poxvirus-encoded proteins
.
Proc Natl Acad Sci USA
.
1999
;
96
:
11537
42
.

Yung
 
SC
,
Parenti
 
D
,
Murphy
 
PM
.
Host chemokines bind to Staphylococcus aureus and stimulate protein A release
.
J Biol Chem
.
2011
;
286
:
5069
77
.

Zanetta
 
JP
,
Bonaly
 
R
,
Maschke
 
S
 et al.  
Differential binding of lectins IL-2 and CSL to Candida albicans and cancer cells
.
Glycobiology
.
1998
;
8
:
221
5
.

Zav'yalov
 
VP
,
Chernovskaya
 
TV
,
Navolotskaya
 
EV
 et al.  
Specific high affinity binding of human interleukin 1 beta by Caf1A usher protein of Yersinia pestis
.
FEBS Lett
.
1995
;
371
:
65
8
.

Zelante
 
T
,
Iannitti
 
RG
,
De Luca
 
A
 et al.  
Sensing of mammalian IL-17A regulates fungal adaptation and virulence
.
Nat Commun
.
2012
;
3
:
683
.

Zha
 
L
,
Garrett
 
S&
,
Sun
 
J
.
Salmonella infection in chronic inflammation and gastrointestinal cancer
.
Diseases
.
2019
;
7
:
28
.

Zhang
 
YJ
,
Rutledge
 
BJ&
,
Rollins
 
BJ
.
Structure/activity analysis of human monocyte chemoattractant protein-1 (MCP-1) by mutagenesis. Identification of a mutated protein that inhibits MCP-1-mediated monocyte chemotaxis
.
J Biol Chem
.
1994
;
269
:
15918
24
.

Author notes

Sophia U J Hitzler and Candela Fernández-Fernández authors contributed equally to this work

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact [email protected]