Abstract
Antimicrobial drug resistance is threatening to take us to the “pre‐antibiotic era”, where people are dying from preventable and treatable diseases and the risk of hospital‐associated infections compromises the success of surgery and cancer treatments. Development of new antibiotics is slow, and alternative approaches to control infections have emerged based on insights into metabolic pathways in host–microbe interactions. Central carbon metabolism of immune cells is pivotal in mounting an effective response to invading pathogens, not only to meet energy requirements, but to directly activate antimicrobial responses. Microbes are not passive players here—they remodel their metabolism to survive and grow in host environments. Sometimes, microbes might even benefit from the metabolic reprogramming of immune cells, and pathogens such as Candida albicans, Salmonella Typhimurium and Staphylococcus aureus can compete with activated host cells for sugars that are needed for essential metabolic pathways linked to inflammatory processes. Here, we discuss how metabolic interactions between innate immune cells and microbes determine their survival during infection, and ways in which metabolism could be manipulated as a therapeutic strategy.
Keywords: bacterial pathogen, fungal pathogen, glycolysis, immunometabolism, macrophage
Subject Categories: Immunology; Metabolism; Microbiology, Virology & Host Pathogen Interaction
Glossary
- AA
amino acids
- ADP
adenosine diphosphate
- ATP
adenosine triphosphate
- BCG
bacillus Calmette–Guérin (vaccine)
- cGAS
GMP‐AMP synthase
- CoA
coenzyme A
- ER
endoplasmatic reticulum
- ETC
electron transport chain
- FA
fatty acids
- GABA
γ‐aminobutyric acid
- GAL4
galactose‐induced gene 4
- GFP
green fluorescent protein
- GlcN
glucosamine
- HDACi
histone deacetylase inhibitors
- HIF
hypoxia‐inducible factor
- ICD
isocitrate dehydrogenase
- ICL
isocitrate lyase
- IDH
isocitrate dehydrogenase
- IDO
indoleamine 2,3‐dioxygenase
- IFN
interferon
- IL
interleukin
- IRF
interferon regulatory factor
- IRG
immune‐responsive gene
- KDM
lysine‐specific demethylase
- LPS
lipopolysaccharide
- MPLA
monophosphoryl lipid A
- mROS
mitochondrial reactive oxygen species
- MS
malate synthase
- mTOR
mechanistic target of rapamycin
- NAD
nicotinamide adenine dinucleotide
- NADPH
nicotinamide adenine dinucleotide phosphate
- NLRP
NACHT, LRR and PYD domain‐containing protein
- NMN
nicotinamide mononucleotide
- NO
nitric oxide
- NOS
nitric oxide synthase
- Nrf2
nuclear factor erythroid 2‐related factor 2
- OXPHOS
oxidative phosphorylation
- PARP
poly(ADP‐ribose) polymerase
- PBMCs
peripheral blood mononuclear cells
- PFK
phosphofructokinase
- PPAR
peroxisome proliferator‐activated receptors
- PYK
pyruvate kinase
- QPRT
quinolinate phosphoribosyltransferase
- ROS
reactive oxygen species
- SDH
succinate dehydrogenase
- SIRT
sirtuin (silent mating type information regulation 2 homolog)
- STING
stimulator of interferon genes
- TCA cycle
tricarboxylic acid cycle
- TF
transcription factor
- TLR
Toll‐like receptor
- UDP‐GlcNAc
uridine diphosphate N‐acetyl glucosamine
Challenges in treating infectious diseases
The discovery of antibiotics in the 20th century is often heralded as one of the major, if not the biggest medical breakthrough in history. Antibiotics save lives from common infections and support medical interventions that require invasive and immunosuppressive approaches, such as surgery, organ and stem cell transplantation and cancer chemotherapy. Unfortunately, less than a 100 years later, we are again in a crisis situation 1. We have overused and misused antibiotics and antifungals not only to treat humans, but also to support food production of animals and plants for our ever‐increasing population. Microbes can rapidly increase cell populations and possess adaptive mechanisms, such as genome rearrangements that promote drug resistance 2 and the appearance of so‐called “mutator” strains 3, 4. This means that the evolution of antimicrobial drug resistance can be fast in all classes of pathogens (bacteria, fungi and parasites) 1, 5, 6, 7. Development of new antimicrobial drugs has not been particularly successful, and pharmaceutical companies are pulling out from developing new antimicrobial agents because of reduced profit margins. A 2016 review sponsored by the UK Department of Health and the Wellcome Trust suggested that in the next three decades, deaths from drug‐resistant infections could be 10 million per year and thereby surpass deaths from cancer 8. While this assertion has been disputed 9, it is nevertheless clear that the threat from infectious diseases is not decreasing, but rather it is increasing. This is not just a problem of the developing world, but concerns all countries where opportunistic infections threaten many procedures of modern medicine, which we take for granted, and which make us feel that we can conquer any disease. Given that developing new antibiotics is challenging, we need to consider diverse facets of the host–microbe interface, to hopefully develop a range of complementary strategies to manipulate this interaction to benefit the host at the expense of the pathogen.
Developing alternative strategies to manage infections critically depends on understanding both sides of the host–microbe interaction. Antimicrobial immunity on the one hand, and immune evasion by microbes on the other, rely on appropriate regulation of transcriptional and signalling networks. These are in turn driven by chemical and physical principles and interactions. Therefore, not surprisingly, it has transpired that host–pathogen interactions are fundamentally regulated by an interplay between host and microbial metabolic pathways and the levels of metabolites in infection microenvironments. Humans face microbes not only in infection but, as is true for any other multicellular organism, they have evolved to coexist commensally with bacteria, fungi or parasites. This is likely to have shaped the metabolic adaptations of host and microbes alike. Commensal microbes generate nutrients essential to maintain human health, but also deplete nutrients to suppress growth of pathogenic microbes. Microbial metabolites also control immunity, by being sensed and metabolised by immune cells. Despite these beneficial interactions with microbiota, commensals and also bona fide pathogens can cause bacteraemia, fungaemia, sepsis, organ failure and mortality. Therefore, a functional epithelial layer and immunity are critically important for maintaining the commensal state of microbiota and defending us from infections. As we will review here, recent advances gave birth to a field of research commonly referred to as “immunometabolism”, which aims to understand how responses of innate immune phagocytes are shaped by cellular metabolism, metabolites and available nutrients. The metabolic adaptations are particularly evident in innate immune cells as they rapidly respond to changing environments, but metabolism also controls responses to infections in T cells and epithelial cells 10. As a counterpart to immunometabolism, microbes reshape their metabolic pathways to match host environments, and could also be employing metabolic tactics to counter immune attack by competing with immune cells for nutrients that are essential for effective antimicrobial responses. Here, we discuss what is known about the reprogramming of host and pathogen metabolic pathways in infection, the plasticity (or otherwise) of host and pathogen metabolism, and how the metabolic interplay controls the host–pathogen interaction. We will focus predominantly on macrophages, as they have been studied extensively in the context of microbial interactions, but several of the mechanisms are also relevant to other immune and non‐immune cell types. Finally, we will assess the potential of nutritional approaches and metabolic manipulations in aiding anti‐infective therapy.
Metabolism of macrophages
Macrophages come in different forms reflecting their diverse roles during infections, inflammation, wound repair and tissue homeostasis. While each macrophage is likely able to fulfil many, if not all, of these roles, tissue‐resident and recruited macrophages, as well as blood monocytes and neutrophils that accumulate at sites of infection and sterile inflammation, employ distinct immune responses. Macrophage responses may also differ within the same cell population due to concentration gradients of cytokines and pathogens, as well as between different innate immune cells, as all tissue‐resident macrophages likely originate from yolk sac and foetal liver progenitor cells, and are thus distinct from monocyte differentiated macrophages that originate from bone haematopoietic stem cells 11, 12, 13, 14. The former population is believed to be long‐lived and self‐renewing, suggesting different metabolic capabilities. The surrounding tissue cells are also important for macrophage development and function 15. The inflammatory milieu and microbial products further drive the differentiation of macrophages, which impacts their metabolism. For instance, the cytokines IL‐4 and IFN‐γ have long been used to characterise macrophage populations with distinct functions, which are best characterised by their metabolic state. In general, IL‐4 promotes amino acid metabolism via mitochondrial respiration and fatty acid oxidation and upregulates arginase‐1 to generate ornithine and urea for polyamine biosynthesis 16. In contrast, IFN‐γ‐treated mouse macrophages utilise exogenous arginine to generate nitric oxide (NO) radicals via NOS2, thereby inhibiting arginase‐1 activity 17. The bacterial cell surface component lipopolysaccharide (LPS) and IFN‐γ cause a general reduction in amino acid metabolism due to reduced mitochondrial respiration and the upregulation of catabolic enzymes, such as IDO1 that depletes tryptophan levels. Under these conditions, increased glucose import enhances glycolysis and the pentose phosphate pathway to generate energy (ATP) and oxygen radicals (ROS) via the NADPH oxidase. These metabolic responses promote antimicrobial activities: depletion of tryptophan and the generation of ROS prevent the growth of intracellular microbes, which are dependent on host‐derived amino acids and susceptible to ROS. Based on these observations, changes in metabolism were thought to support already committed macrophages to fulfil their diverse functions during infections and tissue homeostasis 17.
More recent studies, however, have demonstrated that the metabolic reprogramming that occurs after LPS exposure not only supports, but directly controls and enables immune responses (Fig 1), as inhibition of glycolysis prevents the expression of the pro‐inflammatory cytokine IL‐1β in primary mouse macrophages 18. This result with LPS was validated with bacterial infection of macrophages with Bordetella pertussis 18. Upregulation of glycolysis in LPS‐activated macrophages leads to the secretion of lactate rather than fuelling mitochondrial respiration despite the presence of oxygen 19. This resembles metabolic change in cancer cells and is commonly referred to as Warburg metabolism, which is utilised by rapidly dividing cells or immune cells that need to quickly respond to environmental cues 20. This is in contrast to naïve macrophages, which utilise both glycolysis and mitochondrial metabolism, whereby the latter generates important metabolites for amino acid and fatty acid synthesis, as well as ATP via oxidative phosphorylation (OXPHOS) via the electron transport chain (ETC) 21, 22.
Figure 1. Metabolic reprogramming of microbes and macrophages in infection.

When challenged with stimuli such as the bacterial ligand LPS, IFN‐γ, the bacterium M. tuberculosis and the fungus C. albicans, macrophages remodel their metabolism so that glucose import and glycolysis are enhanced, and the TCA cycle is compromised. Increased levels of TCA intermediates citrate and succinate play direct roles in promoting antimicrobial responses. Citrate is a precursor of itaconate, an inhibitor of microbial glyoxylate cycle enzyme isocitrate lyase (ICL) and host succinate dehydrogenase (SDH), while succinate stabilises the transcriptional activator of glucose utilisation genes and antimicrobial cytokines, HIF‐1α. Similarly, microbes adapt their metabolism by transcriptionally upregulating the glyoxylate shunt pathway, glycolysis and gluconeogenesis at distinct stages of infection. Reprogramming of microbial metabolism depends on available nutrients, including host‐derived lactate and itaconate. Mitochondria and nucleus (labelled orange and blue, respectively) are present in fungi, but not in bacteria. In fungi, the glyoxylate cycle reactions are further compartmentalised in the peroxisome (indicated by yellow shading). Metabolites and enzymes up‐ and down‐regulated during macrophage–microbe interaction are shown in red and purple, respectively. HIF‐1α, hypoxia‐inducible factor‐1α; ICL, isocitrate lyase; ICT/CCL, succinyl‐CoA:itaconate CoA transferase/(S)‐citramalyl‐CoA lyase; IDH, isocitrate dehydrogenase; IRG1, immune‐responsive gene 1; MS, malate synthase; SDH, succinate dehydrogenase; TF, transcription factor.
After LPS exposure of macrophages, the carbon flux through the TCA cycle is disrupted due to downregulation of isocitrate dehydrogenase (IDH1) and higher levels of the citrate carrier in mitochondria resulting in the accumulation of citrate 21, 23, 24. Increased levels of citrate fuel fatty acid synthesis to generate inflammatory prostaglandins and membrane lipids to promote cytokine secretion 25, 26. In addition, LPS‐activated macrophages upregulate immune‐responsive gene IRG1, which converts citrate into itaconate 21. Itaconate has antimicrobial effects by inhibiting the microbial glyoxylate shunt that generates essential metabolites for intracellular survival of bacteria and fungi 27. Itaconate also regulates host metabolic and immune responses by inhibiting the TCA enzyme succinate dehydrogenase (SDH), and activating transcription factor Nrf2 to control inflammation 28, 29, 30, 31, 32. The increased level of succinate stabilises transcription factor HIF‐1α resulting in the upregulation of IL‐1β 18, 33, 34. The metabolic changes in inflammatory macrophages, and their consequences for antimicrobial responses, are summarised in Fig 1.
Oxidation of succinate via SDH further triggers inflammatory responses via the generation of mitochondrial ROS (mROS), likely due to reverse electron transport. mROS has multiple functions. Firstly, mROS is antimicrobial, delivered by mitochondria that traffic to bacteria‐containing phagosomes 35, 36. Secondly, mROS further stabilises HIF‐1α, promoting inflammation 33. Thirdly, mROS may activate the NLRP3 inflammasome 37. Finally, excessive mROS may lead to irreversible mitochondria damage resulting in the release of DNA, which can lead to NLRP3 and cGAS/STING‐mediated inflammation 38, 39, 40, 41. LPS treatment has also recently been shown to cause depletion of NAD+ due to increased activity of NAD‐consuming pathways, such poly(ADP‐ribose) biosynthesis by PARPs and sirtuins, and downregulation of the NAD biosynthesis gene QPRT, despite upregulation of the initial enzyme IDO1 42. Loss of cellular NAD+ and mitochondrial deacetylase activity of SIRT3 results in the acetylation and inhibition of complex I, thereby reducing oxidative phosphorylation, and triggering increased pro‐inflammatory responses in macrophages and mice challenged with LPS 42. We note that many of the studies that we discussed used LPS stimulation as a proxy for bacterial infection, with validation with whole bacteria rarely performed. Nevertheless, challenge of primary mouse macrophages and human peripheral blood mononuclear cells (PBMCs) with the yeast pathogen Candida albicans resulted in similar metabolic changes to those seen with LPS 43, 44. However, differences compared to LPS have been reported in other cases. Specifically, challenge of human monocytes with the synthetic TLR2 receptor ligand Pam3CSK4, or whole‐cell lysates prepared from the bacterium Mycobacterium tuberculosis, Staphylococcus aureus or Escherichia coli resulted in upregulation of both glycolysis and mitochondrial oxidative phosphorylation 45. Similarly, both glycolysis and oxidative phosphorylation were increased (while fatty oxidation genes were mostly repressed) in human blood samples from people suffering from bacterial or fungal infection 43. Future experiments using live infections of immune cells with diverse microbial pathogens should broaden our understanding of which metabolic changes in innate immune cells are common, and which are specific to distinct infection agents.
In contrast to the situation described above for LPS and IFN‐γ, IL‐4‐mediated transcriptional programmes increase mitochondrial metabolism and respiration in macrophages to promote wound healing mechanisms 17. For instance, inhibiting the reprogramming of mitochondrial respiration prevents anti‐inflammatory phenotype and the upregulation of arginase‐1, which is required to increase proline levels for collagen synthesis to support the extracellular matrix in wound repair 46, 47. Increased rates of fatty acid oxidation and mitochondrial respiration may directly drive IL‐4 activation of macrophages 46, 48, 49. In addition, IL‐4 treatment results in increased hexosamine biosynthesis, generating UDP‐GlcNAc, which is essential for N‐glycosylation of cell surface receptors that are upregulated in these macrophages 21. Hexosamine biosynthesis depends on glucose and glutamine utilisation, and both metabolites are critical to induce the expression of IL‐4‐dependent genes 21, 48, 50. Besides hexosamine biosynthesis, glucose also feeds into glycolysis, which is upregulated in IL‐4‐activated macrophages depending on the transcription factor IRF4 48, 50. It is thought that increased glycolysis enables fatty acid biosynthesis, which is upregulated in IL‐4‐activated macrophages to promote oxidative metabolism. Inhibition of glycolysis and fatty acid biosynthesis prevents the expression of several IL‐4‐dependent markers 48. However, more recent genetic studies suggest that coenzyme A (CoA) homeostasis, rather than mitochondrial respiration, controls IL‐4 activation of macrophages 51. CoA is involved in many cellular processes, ranging from fatty acid synthesis to post translational modification of histones and other proteins. Treatment with IL‐4 causes accumulation of acetyl‐CoA partly due to citrate cleavage and increased uptake of glucose, glutamine and fatty acids, which are catabolised to acetyl‐CoA 50. Increased acetyl‐CoA levels promote histone acetylation, mediating IL‐4‐dependent immune responses and arginase‐1 expression 50.
IL‐4‐treated macrophages remain metabolically flexible, as they can utilise fatty acids or glucose to fulfil their roles, likely because IRF4‐dependent transcription increases mitochondrial metabolism and glycolysis. As such, IL‐4‐treated macrophages can be reprogrammed by LPS/IFN‐γ to express inflammatory markers 49. In contrast, LPS‐ and IFN‐γ‐treated macrophages primarily utilise glucose to fuel metabolic pathways and immune responses, are metabolically inflexible and fail to respond to IL‐4 48, 49.
Immune cell metabolism in sepsis patients
Blood‐derived immune cells of sepsis patients that are exposed to circulating LPS and other microbial products provide unique opportunities to study the role of metabolic reprogramming during and after human infections. Transcriptional profiling of blood‐derived lymphocytes isolated from sepsis patients with acute hyper‐inflammation or LPS‐induced experimental endotoxemia showed differential expression of glycolysis and mitochondrial respiration genes, with either up‐ or downregulation of these pathways detected relative to healthy people depending on the condition 43. Several metabolites are increased in the blood of sepsis patients, including lactate, raising the possibility that the metabolic reprogramming of leucocytes during sepsis contributes to the hyperlactaemia 52. Hyperglycaemia is also strongly associated with severe sepsis, due to insulin resistance and impaired degradation of the insulin receptor 53. Increased blood glucose levels trigger glucose transporter‐mediated transcriptional reprogramming of epithelial cells, which causes loss of gut barrier integrity and infections 54. Whether increased blood glucose levels are critical to support inflammatory responses of activated macrophages and monocytes remains unknown. The plasma concentrations of most amino acids are altered in sepsis patients compared to healthy controls and may indicate disease severity 52. For instance, glutamine levels are increased, and may fuel succinate synthesis via the GABA shunt and thus promote inflammatory responses 55. Also, sepsis patients show increased arginine levels that may support NO production via NOS2 in inflammatory macrophages. Compared to survivors, arginine levels were reduced in sepsis patients that failed to recover 55. Similarly, several plasma lipid levels, particularly carnitine esters, are increased in sepsis non‐survivors relative to controls, together with decreased expression of fatty acid transporters 56. This likely reflects defects in mitochondrial import of fatty acids, which requires carnitine palmitoyltransferase and reduced rates of β‐oxidation. Consistent with this, the expression of peroxisome proliferator‐activated receptors (PPAR) α, β and δ, which regulate several aspects of β‐oxidation, is reduced in severe sepsis patients 57, 58. Collectively, these data support in vitro observations in LPS‐treated macrophages showing that increased glycolysis fuels fatty acid synthesis. Conversely, uptake of fatty acid and their catabolism via oxidation into acetate may further enable the accumulation of citrate and succinate in the TCA cycle to drive inflammatory responses. Of note and as mentioned previously, the reduction of mitochondrial respiration, as occurs in LPS‐treated macrophages, is not always observed in sepsis patients 43, 52, possibly reflecting the infection site and/or microbes involved, as in vitro experiments showed that activation with ligands such as Pam3CSK4, E. coli and S. aureus does not induce a reduction of mitochondrial respiration in monocytes 45.
Current efforts are aimed at identifying the role of individual metabolites in promoting survival of sepsis patients, and how this knowledge could be applied to manipulate metabolism for treatment. The role of metabolism to control macrophage immune responses, however, has largely been studied in ex vivo immune cells or cell lines treated with defined stimuli in rich culture media. Determining the metabolism of tissue‐resident macrophages and monocyte‐derived macrophages within infection microenvironments remains technically challenging. It is equally difficult to quantify the levels of key immune‐related metabolite levels in these niches. While inflammatory responses are critically important to eliminate microbes during sepsis, excessive inflammation contributes to tissue damage, dissemination and increased mortality. As such, timing of nutrient supplementation during sepsis may be critical. Inflammatory macrophages also secrete cytokines to dampen inflammation, including IL‐4 59. Consequently, tissue macrophages may not strictly adhere to an IFN‐γ/IL‐4 dichotomy, but rather follow a broad spectrum of activation states 60. This warrants further studies to determine the metabolism of other activated macrophages and individual lymphocytes isolated from sepsis patients 45.
Immune cell tolerance and paralysis during severe sepsis
In severe sepsis patients, macrophages and monocytes often fail to respond appropriately to microbial and inflammatory signals, resulting in an immune tolerant state that can develop into immune paralysis. The absence of a functional innate immune system may lead to increased infection and mortality rates commonly observed in severe sepsis patients. The underlying mechanisms that lead to immune tolerance, however, remain poorly understood. In vitro studies have shown that LPS‐treated macrophages fail to respond to subsequent LPS exposure, and this is likely driven by epigenetic modifications of histones and changes to gene transcription. In particular, prolonged LPS exposure of macrophages increases cellular levels of NAD+, and NAD+‐dependent sirtuins (SIRT1 and SIRT6) downregulate glycolysis and increase β‐oxidation 61. SIRT6 targets HIF‐1α promoter sites to decrease acetylation of lysine 9 of histone H3 (H3K9Ac), causing reduced expression of genes encoding glycolytic enzymes 62.
NAD+ levels are also increased in immunotolerant monocytes from sepsis patients 43. Here, epigenetic changes at H3K9 similarly correlate with reduced rates of glycolysis in immunotolerant monocytes 43. Tolerant monocytes show reduced mitochondrial and fatty acid metabolism, further compounding their inability to respond to LPS. Nevertheless, immunotolerant monocytes secrete 10‐fold more IL‐10 compared to LPS‐treated control cells 43. IL‐10 is thought to contribute to immunotolerance in macrophages by inhibiting glycolysis and inducing the degradation of damaged mitochondria (mitophagy) via mTOR signalling 63. Loss of IL‐10 increases inflammation, partly due to activation of the NLRP3 inflammasome mediated by increased glycolysis and mitochondrial ROS 63. IL‐10 secretion, as well as defects in metabolism and immune response, is reversed in patients who have recovered from severe sepsis 43. The molecular pathways involved in the resolution of immunotolerance, however, remain to be described.
Metabolic aspects of trained immunity
Immunotolerance likely acts as a mechanism to dampen excessive inflammation and appears to be chiefly mediated by reducing leucocyte metabolism. In a contrasting scenario, innate immune cells can be reprogrammed to respond to a secondary signal more quickly and vigorously compared to the first time. This suggests that innate immune cells are capable of memory, which is referred to as “innate immune memory” or “trained immunity” 64. Trained immunity is not restricted to a particular stimulus, and it confers cross‐protection against diverse infections 65. Similar to macrophages activated by the bacterial cell surface component LPS, the cell wall component from fungal cells, β‐glucan, increases macrophage glycolysis and reduces mitochondrial respiration. Inhibiting glycolysis reduces macrophage immune responses to β‐glucan, as described for LPS 66. In contrast, however, β‐glucan‐mediated metabolic reprogramming lasts for weeks and months, whereas LPS activation leads to metabolic dysfunction and immune tolerance within 6 days 66, 67. It is thought that these different metabolic responses are due to changes in the epigenetic state of chromatin and therefore gene expression, including histone acetylation and methylation 66, 67, 68. Epigenetic modifications are triggered by cellular metabolites, which in turn enhance the expression of metabolic pathways. For instance, β‐glucan is sensed by the cell surface receptor dectin‐1 that signals via Akt/mTOR/HIF‐1α to induce metabolic reprogramming due changes in the TCA cycle and the accumulation of fumarate 66, 68. Fumarate inhibits the KDM5 demethylase and causes the accumulation of the H3K4 trimethylation mark 69. It also has a further effect on H3K27 acetylation and HIF‐1α levels and was associated with increased expression of glycolytic enzymes 69. Epigenetic modifications underpin the long‐lasting changes in the metabolism of innate immune cells, as activation of histone deacetylases reduces trained immunity and promotes immunotolerance 66. This is observed in animals and humans, whereby clearance of initial fungal infections or vaccinations induce permanent changes in the cellular metabolism of innate immune cells 64.
Activated monocytes are thought to be short‐lived whereas trained immunity can last for weeks and months. β‐glucan and other stimuli including the BCG vaccine and Western diets trigger reprogramming of haematopoietic stem cells within the bone marrow that give rise to circulating trained monocytes, explaining some aspects of the long‐lasting effect of trained immunity 70, 71. However, several tissue‐resident phagocytes, including alveolar macrophages, are of embryonic origin and self‐renew independently of the bone marrow 72. Despite this, viral lung infections trigger long‐lasting memory in alveolar macrophages depending on INF‐γ priming by T cells, which increases glycolysis and enables protection from bacterial pathogens 73. This suggests that many microbes encounter innate immune cells that already have a reprogrammed metabolism and display a heightened immune response compared to the naïve monocytes/macrophages commonly utilised in infection studies. This raises the prospect that microbes, such as commensals which regularly interact with host immunity, may have evolved mechanisms to combat reprogrammed innate immune cells to survive, proliferate and establish infections.
Microbial metabolism in infection
Depending on their mode of host interaction (commensal/pathogen) and/or route of infection, microbes encounter diverse host sites, including the bloodstream, skin, gut, mucosal surfaces, organs and tissues, and intracellular host environments when phagocytosed by innate immune cells. These environments are nutritionally heterogeneous, and microbial metabolism has therefore evolved to be adapted to their particular lifestyle, and respond rapidly when nutrient level change. Specifically, the immunometabolic shift in activated macrophages directly “talks” to the metabolic pathways of microbes (Fig 1), and this crosstalk modulates host–pathogen interactions. While immunometabolic shifts are meant to help the host to defeat the infection, in some instances microbes might benefit from the changes in metabolite levels that result from host metabolic reprogramming 74, 75, 76, 77. Furthermore, it is also possible for the microbial pathogen to seize the opportunity and turn the immunometabolic reprogramming of host cells into a nutritional disadvantage 44. Below we review the metabolic strategies of key microbial pathogens and discuss the concept of microbes turning the tables on immune cells during their metabolic interactions.
Extracellular pathogens
Pathogens that persist in the blood as extracellular microbes, such as Trypanosoma brucei, or within red blood cells, such as Plasmodium falciparum, will invariably utilise glucose as the primary carbon source 78. In these microbes, mitochondrial metabolism is thought to be reduced, altered or absent, although more recent genetic and metabolic profiling approaches have identified additional pathways and alternative carbon sources that promote survival of T. brucei and P. falciparum 79, 80. These additional pathways either replenish metabolites otherwise depleted by glycolysis or support microbial survival when glucose levels are limited.
Mycobacterium tuberculosis
In contrast to extracellular pathogens, intracellular microbes frequently encounter environmental niches that contain low glucose levels. This is particularly evident in microbes that reside in specialised vacuoles of macrophages, as these host cells rapidly alter their own metabolism during infections and thus available nutrients. As discussed above, macrophages infected with BCG/killed Mycobacteria tuberculosis upregulate glycolysis to trigger inflammatory responses and trained immunity. In contrast, macrophages harbouring virulent M. tuberculosis show reduced rates of glycolysis and mitochondrial respiration, but increased uptake of exogenous fatty acids 81. Perhaps as a consequence of this, M. tuberculosis fails to gain access to sufficient levels of glucose and uses fatty acids a primary carbon source within macrophages. While determining metabolite concentrations within organelles remains challenging, M. tuberculosis mutants that are no longer able to utilise glucose are still able to establish lung infections in mice, suggesting that the levels of glucose are low in infection microenvironments and that intracellular bacteria utilise alternative carbon sources 82. However, mutants that cannot utilise glucose fail to maintain viability during chronic infections, indicating that the bacteria have access to at least some pools of glucose and that intracellular glucose levels likely fluctuate during the course of infection 82.
Given the increased levels of fatty acids within infected macrophages, it was thought that M. tuberculosis utilises lipids to maintain growth and viability. Unlike humans, bacteria are able to utilise 2‐carbon acetyl‐CoA derived from fatty acid oxidation, to generate 6‐carbon glucose via TCA intermediates and gluconeogenesis. This is only possible because of the glyoxylate shunt, and enzymes isocitrate lyase (ICL) and malate synthase (MS) that convert the TCA intermediate isocitrate to glyoxylate and then to malate. This metabolic pathway enables bypassing of the oxidative decarboxylation steps within the TCA cycle and replenishes 4‐carbon metabolites important for amino acid synthesis and generation of the gluconeogenic substrate oxaloacetate. Deletion of either enzyme, ICL or ML, reduces the ability of M. tuberculosis to establish and maintain lung infections in mice 83, 84. As other TCA intermediates are likely required to maintain energy bioenergetics and for biosynthesis of important metabolites, bacteria must ensure that isocitrate remains available for isocitrate dehydrogenase (ICD). In M. tuberculosis, this is regulated by glyoxylate itself, which activates ICD, thus establishing a rheostat so that both pathways, TCA cycle and the glyoxylate shunt, have access to sufficient levels of isocitrate 85. As such, the flux of metabolic pathways determines survival of intracellular microbes, but also enables metabolic responses to resist immune attack, because the glyoxylate shunt is primarily protective in INF‐γ‐activated macrophages 86.
Candida albicans
As a human commensal, the fungus C. albicans colonises the gut, oral and genital mucosa, while the transition to pathogenesis is associated with an increased load and, in systemic disease, bloodstream location and invasion of organs. Although C. albicans is phagocytosed by macrophages as a host defence mechanism, this yeast is not an intracellular pathogen. In vitro experiments have shown that C. albicans is equipped with effective mechanisms for escape from macrophages via a switch from yeast to hyphal cell morphology, which in turn activates pyroptosis, a programmed cell death of macrophages that depends on the NLRP3/caspase‐1 inflammasome 87, 88. Since it only occupies the macrophage environment transiently, and it therefore sees both intracellular and extracellular environments, C. albicans displays dynamic regulation of its metabolic pathways.
Analysis of C. albicans mutants is consistent with non‐glucose carbon sources being important during infection, as mutations in the glyoxylate cycle or gluconeogenesis modulate in host fitness in mouse infections 89, 90. The situation with fatty acid oxidation is less clear 91, 92. At least in part, utilisation of non‐glucose carbon sources by C. albicans stems from the necessity to survive and grow within macrophages, because phagocytosed C. albicans upregulates enzymes of the glyoxylate shunt, gluconeogenesis and fatty acid oxidation 44, 93. Transcription of enzymes needed to utilise non‐glucose carbon source was also detected in C. albicans facing human blood 94, 95, in a model of epithelial infection 96, in vivo in mouse infections 97, 98, 99 and in isolates obtained from AIDS patients suffering from mucosal candidiasis 96.
Glucose is the preferred carbon source for C. albicans in vitro, but is it important in vivo? Yes, it is, for pathogenesis 90, and also likely for commensal gut colonisation 100. A C. albicans deletion mutant in the glycolytic enzyme pyruvate kinase (PYK1) shows reduced virulence in mouse infections 90, and so does a mutant in which the two transcription factors needed to transcribe glycolytic and other carbohydrate metabolism genes, TYE7 and GAL4, are deleted 101. But, if alternative carbon source utilisation is “on” in host environments (as suggested by studies discussed above), shouldn't glycolysis be “off”? While fungal glycolysis and glucose utilisation genes are repressed when facing host conditions 93, 95, 98, 102, it is worth noting that several studies compared the transcriptome of C. albicans in the host to in vitro growth in glucose‐rich medium. This means that upregulation of alternative carbon utilisation in host environments is easily detected over the reference condition, but the expression of glycolysis and glucose utilisation genes appears repressed or not changed in this comparison. Indeed, imaging of GFP driven by promoters of phosphofructokinase PFK2 and pyruvate kinase PYK1 showed they were expressed when C. albicans was intracellular in macrophages 90, and the pyk1 mutant of C. albicans cannot form hyphae or kill macrophages in vitro 44. Glycolytic gene expression has also been reported in C. albicans in transcriptomic experiments in mouse infections 97, using GFP fusions to glycolytic gene promoters in C. albicans disseminated to the kidney 90, and during incubation with human blood 94. Glycolytic growth could be needed by C. albicans to proliferate in the host and could further represent an immune evasion mechanism, as activated inflammatory phagocytes have high needs for glucose 44.
Candida albicans has evolved regulatory mechanisms to have metabolic flexibility and consume multiple carbon sources at the same time 103. It is also possible that host–pathogen interactions change over the time frame of infection, and therefore, regulation of carbon source utilisation by C. albicans is dynamic. This is most clearly seen during infection of macrophages by C. albicans in vitro, where the initial repression of fungal glycolysis is reversed coinciding with C. albicans egressing from macrophages and finding glucose again 44, 93. This dynamic regulation is mediated by transcription factors Tye7 and Gal4 that activate glycolytic gene expression in a timely manner as C. albicans escapes macrophages 44. Whether these mechanisms of macrophage escape through hyphal formation and dynamic reprogramming of fungal metabolism also operate in vivo remains to be understood. Finally, lactate is thought to be an important carbon source for C. albicans in humans, where it is produced by other microbiota and by activated inflammatory phagocytes that have switched to Warburg metabolism. Again, utilisation of lactate might serve a few different functions for C. albicans, enabling the fungus not only to proliferate, but also to evade immunity by impacting on cell surface structure and immune cell interactions 104.
Leishmania parasites
Not all intracellular microbes express glyoxylate shunt enzymes, including Leishmania parasites, and may thus rely on sugar uptake within macrophages. Because glucose levels are limited, Leishmania also scavenges lipids and amino acids from its macrophage host to fuel mitochondrial respiration 105, 106, 107. This indicates that some microbes have the ability to utilise different carbon sources to promote growth and expression of cell surface virulence factors. Surprisingly, however, Leishmania parasites suppress sugar and amino acid uptake by inducing the degradation of cell surface transporters, resulting in reduced rates of glycolysis and mitochondrial metabolism 105. The minimal metabolism is thought to prevent depletion of important nutrients, but also reduces mitochondria‐mediated oxidative stress that is likely detrimental in an oxidative environment created by antimicrobial responses of macrophages. The stringent metabolism of Leishmania is also associated with reduced expression of cell surface carbohydrates, typically considered virulence factors, and a slow growth rate (doubling time: ~12 days) 16. This raises the prospect that Leishmania parasites evade immune attack by reducing metabolism and the expression of cell surface molecules that are sensed by macrophages. Alternatively, intracellular microbes that rely on host cell for survival may forfeit glucose to support host metabolism. This would also minimise the metabolic stress on the host, which can be sensed to mount potent immune responses 108. In support of this, Leishmania scavenges amino sugars that are nutritionally less important for macrophages to replenish its metabolic pathways during intracellular growth 109, 110. This suggests that microbes adapt their metabolic capabilities depending on the environmental niche, but also indicates that some microbes may have been driven into hostile territory, such as lysosomes, in search for essential nutrients and to evade immune attack 16.
Turning the tables on immune cells: how microbial pathogens might benefit from immunometabolic shifts
From everything that we discussed so far, the consensus is that the metabolic shifts in immune cells upon infection, i.e. heightened glycolysis, disruptions to the TCA cycle and changes to mitochondrial respiration, drive antimicrobial inflammation and the production of direct antimicrobial effectors, such as itaconate and ROS. To launch a productive infection, microbes must either evade immune cell activation or, alternatively, adapt to, or even benefit from, the metabolic shifts in immune cells.
Immunometabolic shifts cause increased concentrations of glucose and lactate in host cells that could promote intracellular survival of Listeria monocytogenes, Legionella pneumophila, Brucella abortus and M. tuberculosis 74, 75, 76, 77, 111. A glucose‐consuming microbe would potentially also interfere with inflammatory responses, similar to the glycolytic inhibitor 2‐deoxy‐glucose that is commonly used in studying Warburg metabolism in immune cells. Furthermore, LPS‐/IFN‐γ‐activated or C. albicans‐activated macrophages are metabolically inflexible and rely on glucose due to the downregulation of mitochondrial oxidative phosphorylation 44, 49. Not only that, activated macrophages rapidly lose viability in the absence of glucose, which means that glucose‐consuming microbes could perturb host glucose homeostasis with potentially catastrophic consequences, as we have recently described for macrophages infected ex vivo with C. albicans 44 (Fig 2).
Figure 2. Glucose competition between microbes and host cells determines host–pathogen responses and outcomes.

(A) Upon C. albicans infection, macrophage glycolysis is turned on rendering them metabolically inflexible and necessitating glucose. Once internalised by macrophages, C. albicans switches from yeast to filamentous hyphae supported by amino and fatty acid (AA, FA) metabolism. Initial hyphal escape is mediated in part by NLRP3 inflammasome‐dependent pyroptosis 87, 88. Fungal hyphae then reactivate glycolysis following egress from macrophages 44, 93, which leads to depletion of glucose by the fungus 44. Glucose starvation triggers death of macrophage, as these immune cells are metabolically inflexible and become dependent on glucose as the sole carbon source 44. Once glucose levels drop, C. albicans may utilise macrophage‐derived lactate and other carbon sources, such as amino and fatty acids, but this remains to be firmly shown. (B) S. Typhimurium competes for glucose with macrophages, disrupting host glucose homeostasis and leading to increased levels of ROS and decreased levels of NADH 112. This metabolic stress causes activation of the inflammasome, with consequent production of cytokine (IL‐1β) and pyroptotic lysis. Pyroptosis is a host inflammatory response, but also enables bacterial egress and dissemination. C) During skin infection, S. aureus consumes glucose, triggering glucose stress in keratinocytes 113. This contributes the signal for Warburg reprogramming in keratinocytes, and consequent activation of transcription factor HIF‐1α. HIF‐1α mediates pro‐inflammatory cytokine transcription, and also transcription of genes related to glycolysis and glucose consumption. D) While Leishmania preferentially uses glucose as carbon source, in intracellular stages it relies on alternative carbon sources including glucosamine (GlcN), amino acids (AA) and fatty acids (FA) 105, 106, 107, 110. Glucose uptake is reduced, likely promoting macrophage viability and parasite growth.
Glucose competition between host cells and microbes has also been suggested for Salmonella enterica serovar Typhimurium infecting macrophages and Staphylococcus aureus infecting skin keratinocytes 112, 113 (Fig 2). Glucose consumption by intracellular S. Typhimurium disrupts host glycolysis, which creates a metabolic signal in the form of lower levels of NADH and higher ROS, activating the NLRP3 inflammasome 112. In this scenario the consequence of competition for glucose is an inflammatory response, which includes maturation and secretion of IL‐1β cytokine and lysis of macrophages by pyroptotic cell death. Pyroptosis contributes to inflammation, but also releases intracellular bacteria to spread infection, potentially benefitting the pathogen. In the case of S. aureus, the glycolysis mutant pyk1 was less able to activate metabolic reprogramming in keratinocytes towards high glycolysis, and it triggered less IL‐1β cytokine and reduced lysis of keratinocytes and was less virulent in an animal model of skin infection 113. The authors proposed that glucose consumption by S. aureus causes nutritional stress in keratinocytes, leading to upregulation of glycolysis and driving inflammatory processes 113 (Fig 2). Staphylococcus aureus can replicate and grow extracellularly. Therefore, here also, glucose competition could be benefitting the pathogen by contributing to host cell lysis and microbial release. In both bacterial and fungal interactions with macrophages, it has been shown that the consequences of glucose competition can be reversed by restoring host metabolic homeostasis. Macrophage cell death following C. albicans infection can be delayed by increasing the availability of glucose 44, and inflammasome activation by S. Typhimurium can be suppressed by addition of pyruvate, the final metabolite produced by glycolysis 112.
Does infection disrupt host glucose homeostasis in vivo? While sepsis, as many other severe illnesses, is associated with increased blood glucose levels (so‐called “stress hyperglycaemia”), sepsis patients might be hyper‐susceptible to insulin interventions aimed at reducing glucose levels, which can cause life‐threatening hypoglycaemia 114. Interestingly, we have seen an equivalent mechanism in the mouse candidaemia model, whereby C. albicans‐infected mice developed severe hypoglycaemia following administration of the glucose‐lowering drug metformin, whereas in the absence of infection mice could maintain blood glucose levels when injected with the same dose of metformin 44. In another mouse study, injection of LPS or infection with S. Typhimurium caused hypoglycaemia when co‐administered with a low dose of insulin 53. It is worth noting that in both infection models, with C. albicans and with S. Typhimurium, the microbial infection alone was also causing some hypoglycaemia, which was dramatically increased with glucose‐lowering interventions (metformin or insulin) 44, 53. A transient reduction in host blood glucose was also reported in a mouse model of polymicrobial bacterial septic infection 115.
At present, it is not clear whether, during infection, microbes directly perturb host glucose homeostasis (by competing with host cells for glucose), or whether the observed hypoglycaemia results from dysfunction of physiological processes during sepsis. These two scenarios are not mutually exclusive. Infection can impact on insulin clearance 53 and reduce glucose biosynthesis by gluconeogenesis in the liver 115. This is consistent with broad physiological effects of infection on glucose metabolism and uptake. Supporting direct effects of microbial competition for glucose, glycolytic mutants of C. albicans have reduced ability to trigger hypoglycaemia in response to metformin 44. The interpretation of these data is however complicated: do the C. albicans glycolytic mutants maintain host glucose homeostasis because they consume less glucose? Or is it because they are hypovirulent, and therefore trigger distinct physiological responses by the host compared to fully virulent strains? Testing microbial mutants in a variety of pathways required for virulence (metabolic and non‐metabolic) should resolve this question.
Therapeutic options for infections based on nutrients and metabolism
The prospect that inhibiting metabolic enzymes or boosting metabolic pathways may be beneficial in infectious diseases is gaining traction (Table 1). These anti‐infective approaches would target the host, compared to traditional antimicrobial therapy, which targets the pathogen. Given the difficulties in developing new antimicrobial therapeutics, host‐targeted approaches should be considered, while keeping in mind potential side effects of metabolic therapies with broad effects on multiple host cell types.
Table 1.
Possible metabolic strategies in infection
| Strategy | Target | Compound | Effect |
|---|---|---|---|
| I. Target metabolic enzymes | Glycolysis | 2‐Deoxy glucose (2‐DG) | Repressed inflammation via HIF‐1α |
| GABA transaminase of the GABA shunt; production of succinate | Vigabatrin | Repressed inflammation via HIF‐1α | |
| II. Target chromatin and transcription factors that drive metabolic reprogramming | Demethylase | Fumarate | Induction of trained phenotype in monocytes. Induction of HIF‐1α‐dependent metabolic genes |
| SIRT1 | EX‐527 | Reactivation of immune response in tolerant immune cells. Inhibition of SIRT1 inhibits metabolism of fatty acids. Acts on transcription factors NF‐kappa Β and PGC‐1 | |
| Histone lysine deacetylases (HDACs) |
Panobinostat Entinostat RGFP966 |
Reduction of inflammatory and anti‐inflammatory cytokine production | |
| III. Modulate signalling pathways that control immunometabolism |
AMPK (mTOR indirectly) Mitochondrial respiration |
Metformin | Repress inflammation by inhibiting metabolic reprogramming of immune cells. |
| Dectin‐1 signalling | β‐Glucan | Promote trained immunity and reverse immunoparalysis by triggering epigenetic changes in metabolic gene expression. | |
| TLR4 signalling | Monophosphoryl lipid A (MPLA) | Triggers reversible reprogramming on macrophages to Warburg effect and the reactivation of mitochondrial functions | |
| IFN‐γ signalling | IFN‐γ | Induces glycolysis and reactivates immunotolerant monocytes. | |
| NAD+ levels | β‐Nicotinamide mononucleotide (NMN) | Reduce inflammation by promoting SIRT3‐dependent deacetylation of substrates in mitochondria, to increase mitochondrial respiration and metabolism. | |
| IV. Supplement metabolites | Various cells | Glucose |
Maintain homeostatic blood glucose levels Maintain immune cell function Maintain tissue homeostasis, reduce ER stress and protect brain function |
Several clinical studies have focused on inhibiting inflammatory pathways during sepsis, as uncontrolled cytokine secretion is thought to lead to organ and tissue damage. One approach to this is to control metabolic enzymes. Given the importance of glycolysis and its enzymes for inflammatory responses in innate immune cells, glycolysis blockers, such as 2‐deoxy glucose, prevent secretion of IL‐1β during LPS administration in mice 18. Another possible target is GABA transaminase, which fuels the TCA cycle to promote accumulation of succinate. Its inhibition with vigabatrin reduced IL‐1β secretion after LPS administration or Salmonella infections in mice 18. Vigabatrin is currently used in the clinic to treat epilepsy and is associated with increased risks of infections, possibly due to its anti‐inflammatory properties 116.
Another approach is to control chromatin modification enzymes. Histone deacetylase inhibitors (HDACi), some of which are approved for cancer treatment, also reduce inflammation in fungal and bacterial infections 117. Whether this is solely due to changes in histone acetylation and gene expression, or also involves the many non‐histone targets of HDACs, awaits confirmation. It is worth keeping in mind that bacterial metabolites, such as the short‐chain fatty acid butyrate, are potent HDAC inhibitors. Moreover, the TCA metabolite fumarate or derivatives that are less likely to be further metabolised inhibit histone demethylation, and their administration may be beneficial in modulating inflammatory responses to infection 69.
Signalling pathways that control metabolism and inflammatory responses could also be targeted. For instance, metformin, which is commonly used to treat diabetes via activation of AMP kinase to lower blood glucose levels, dampens inflammation. This is mediated via reduced mTOR activity, which in turn dampens HIF‐1α‐dependent secretion of IL‐1β 43. Metformin treatment is associated with reduced risk of developing sepsis in humans 118; however, studies in mice suggest that it may increase opportunistic fungal infections by lowering immune responses to C. albicans and causing hypoglycaemia 43, 44. LPS triggers loss of NAD+, and restoring NAD levels by administration of the precursor metabolite, β‐nicotinamide mononucleotide (NMN), prevents inflammatory responses in macrophages after LPS challenge 42. Interestingly, macrophages show increased inflammatory responses in the elderly, which are known to have altered NAD homeostasis. Again, NMN administration reverted those inflammatory responses to levels typically associated with younger individuals 42. Monophosphoryl lipid A (MPLA), which activates the macrophage receptor TLR4 and is an FDA‐approved vaccine adjuvant, is also thought to have promise as an anti‐infective agent in bacterial (S. aureus) as well as fungal (C. albicans) infections 119. In vitro experiments with bone marrow‐derived mouse macrophages showed that while initially MPLA treatment triggered a Warburg effect, 3 days later macrophages reactivated mitochondrial oxidative phosphorylation 119. This result is interesting in the light of our work with C. albicans that showed that bone marrow‐derived macrophages cannot reactivate mitochondrial metabolism in time to counter glucose depletion by the fungus 44. Making macrophages able to reactivate mitochondrial functions could protect them under glucose starvation conditions. We note that under our experimental conditions (multiplicity of infection of 3 Candida to 1 macrophage and standard tissue culture medium RPMI that contains ≈ 10 mM glucose), the time frame for the glucose depletion and catastrophic consequence on host mitochondria was a matter of hours, rather than days. The precise conditions of infection in vivo, including multiplicity of infection, glucose concentrations and levels of other metabolites, will all impact on these processes. Future work will need to understand how we can translate the in vitro data to in vivo effects.
Given that sepsis can progress to an immune‐tolerance state, metabolic reprogramming of innate immune cells to restore inflammatory responses and prevent infections is also currently being explored experimentally. For instance, fungal β‐glucan induces trained immunity characterised by increased glycolysis and partially reverses LPS‐induced immunoparalysis of human monocytes after experimental endotoxemia 67. This was largely due to induced histone modifications that restored the expression of several inflammatory cytokines. Administration of glucans showed protection not only against C. albicans, but also against S. aureus infections, in keeping with the observation that trained immunity is not a pathogen‐specific response 120. While β‐glucan administration has not been validated in human sepsis patients, it is currently being trialled to enhance anti‐cancer immunotherapies 121. β‐glucan as well as the BCG vaccine also protect from unrelated infections by enhancing pro‐inflammatory responses, suggesting the possibility to stimulate protective mechanisms against infectious diseases and also cancers 121. Both β‐glucans and BCG cause epigenetic changes on key glycolytic genes to maintain their increased expression 66, 122. Targeting these epigenetic mechanisms directly has emerged as a potential mechanism to reactivate immunotolerant leucocytes, as specific histone marks have been associated with reduced expression of pro‐inflammatory cytokines. This is partly driven by the NAD+‐dependent deacetylase, SIRT1 61. The SIRT1 inhibitor EX‐527 was able to reverse the tolerance phenotype of septic mice, resulting in improved mouse survival rates, bacterial clearance and innate immune responses 123. Presumably this also reflects increased metabolic rates of leucocytes to promote inflammatory response, but this has not been investigated so far. It has also become apparent that cytokines themselves affect the metabolism of leucocytes, including autocrine signalling. For instance, IFN‐γ was able to restore the immunotolerant phenotype of human monocytes isolated from sepsis patients, which includes increased glycolytic flux as determined by lactate secretion 43. This suggests that reactivation of inflammatory metabolism is feasible and that this restores immunoparalysis in fungal sepsis patients 43, 124.
Lastly, several studies have shown that administration of metabolites can be beneficial in diverse infections. Blood glucose levels are tightly controlled to maintain tissue homeostasis, but likely also contribute to maintaining immune function. During sepsis, blood glucose levels initially increase but can become hypoglycaemic in patients where glucose control is implemented, which is associated with poor clinical outcomes 114. In murine infection models with diverse pathogens (bacteria, fungi and viruses), glucose has been shown to have either positive or negative effects on host outcomes (Fig 3). These contrasting results highlight the need for deeper understanding of how glucose regulates host physiology depending on the nature of infection. It will also be essential to study how this research in experimental animals translates to humans. Glucose supplementation rescued mice that failed to maintain blood glucose levels after polymicrobial infection 115. Glucose administration is also protective in experimental influenza infections by promoting protein glycosylation and reducing ER stress to maintain tissue homeostasis in mice 125. Loss of blood glucose is also partly due the reduced nutritional intake, termed sickness behaviour, during infections. Reduced food intake, and thus reduced blood glucose levels, has been shown to be protective in some bacterial infections and LPS sepsis model, although food restrictions negatively affect host outcomes in many other infections 125, 126. Not surprisingly then, microbes themselves target host metabolism as Salmonella prevents reduced food intake of its host to increase survival of bacteria and host 126. Similarly, host metabolism affects microbial virulence, as decreased glucose absorption due to insulin resistance increases gut glucose levels, which then suppresses expression of a virulence factor in pathogenic E. coli 127. Consequently, oral glucose administration attenuates gut infections and increases mouse survival 127. Moreover, feeding infected mice glucose improved animal survival rates in systemic C. albicans infection (precisely why glucose was beneficial in this context remains to be determined) 44. Glucose also rescued animals from the severe hypoglycaemia that developed when metformin and C. albicans were administered in conjunction 44.
Figure 3. Diverse effects of glucose metabolism on host outcomes in animal infections.

Glucose administration prevents hypoglycaemia (loss of blood glucose) in Candida albicans‐infected mice treated with metformin and in polymicrobial infections in the absence of liver gluconeogenesis, thereby promoting mouse survival 44, 115. Glucose is also beneficial in mice during lung infections with influenza by preventing neuronal death due to increased glucose demand, although loss of blood glucose levels was not restored 125. In contrast, glucose administration accelerates accumulation of reactive oxygen species (ROS) in the brain after Listeria monocytogenes infection and decreases mouse survival 125.
Conclusions and outlook
In the era of raising concerns regarding antimicrobial drug resistance and lack of investment to develop new antibiotics and antifungals, it is paramount that we develop complementary strategies for the treatment of infectious diseases. As discussed in this review, metabolic interactions of innate immune cells and pathogens present a range of opportunities for interventions that will benefit the host by assisting immune cells in their antimicrobial activities, and by controlling inflammation so that it is helpful, but not toxic. However, several challenges and open questions remain (see Box 1). In particular, we need to understand “local versus global issues”, to borrow an expression from politics. For example, we need to be able to know how the levels of metabolites and nutrients fluctuate in the infection foci in local microenvironments in infected tissues and organs, which could be achieved by applying new imaging methods that are able to capture metabolites, such as NMR microscopy. Global changes in metabolites affect the physiology that accompanies infections, and understanding how metabolism affects sickness behaviour promises new therapeutic options. There is also a temporal dimension to this problem, in that different stages of infection (i.e. early acute phases and later septic phases) are likely different with respect to metabolism of immune cells and pathogens, as they adapt to each other and to changing nutrient levels in their environment. This means that different strategies might be needed at different stages of disease. Regardless of these challenges, we and many others believe that manipulating metabolism holds great promise for combatting microbial pathogenesis. It is also a strategy that might be broadly applicable to infections with diverse pathogens, as demonstrated by the discussed examples of protection in bacterial and fungal infections. In this context, it is worth noting that polymicrobial infections occur, including mixed bacterial–fungal infections that present distinct challenges 128. Immunometabolic interventions could potentially be very useful in the situation of multiple infecting pathogens.
Box 1. In need of answers.
-
What nutrients are available to immune cells and microbes locally in infection microenvironments? How do the levels of nutrients change during infection?
During infection, the host–pathogen interaction “wars” occur in localised environments. These include intracellular locations in immune cells (phagosome, cytoplasm), as well as various tissues and organs. Tissue culture media and ex vivo infections might not faithfully reproduce the situation in these infection microenvironments. To fully understand the metabolism of both immune cells and microbes during their interaction, we need to define the local availability of nutrients in infection microenvironments, as well as understand how the levels of nutrients might dynamically change during infection.
-
What are the metabolic profiles of immune cells and microbes in infection microenvironments?
Metabolic profiles of host immune cells and microbial pathogens have mostly been analysed in ex vivo challenge scenarios, with defined nutrient composition of the media and the presence of only two cell types (the immune cell and the microbe under study). While these reductionist approaches are critical for mechanistic insight, infection microenvironments are complex, not only nutritionally as described above in Question 1, but also in relation to the presence of other cell types. To fully understand how we could manipulate metabolism to help the host fight infection, it will be important to profile the metabolic status of both host and pathogen from infection microenvironments.
-
How can metabolic interventions modulate inflammation to promote pathogen clearance, but prevent pathology driven by over inflammation?
The different stages of infection in patients might require distinct metabolic approaches to promote antimicrobial functions of immune cells, restart the immune system in immune paralysis or dampen inflammation if it has become pathological. Which metabolic interventions will be most useful in these diverse scenarios and how the timing of the intervention during diseases might impact on the outcome are open questions.
-
What is the nature of metabolic host–pathogen interactions in polymicrobial infections?
The majority of studies in the field of immunometabolism have been done using infections with a single species of microbe. However, polymicrobial infections are common. Microbes can influence immune cells in diverse ways 45. Moreover, supplementation of nutrients can have opposing effects (i.e. be beneficial or make things worse) depending on the nature of infection, as has been shown for glucose in viral versus bacterial infection 125. Therefore, we should study immunometabolic shifts and the interactions of host and pathogen metabolism not only in single‐microbe infections, but also in polymicrobial challenge of immune cells and in disease.
Conflict of interest
The authors declare that they have no conflict of interest.
Acknowledgements
Our work on metabolism in infection has been supported by project grants from the Australian National Health and Medical Research Council (APP1081072 to TN and AT, and APP1101562 and APP1158678 to AT). TN is a Future Fellow of the Australian Research Council (FT170100313).
EMBO Reports (2019) 20: e47995
See the Glossary for abbreviations used in this article.
Contributor Information
Ana Traven, Email: ana.traven@monash.edu.
Thomas Naderer, Email: thomas.naderer@monash.edu.
References
- 1. Davies J, Davies D (2010) Origins and evolution of antibiotic resistance. Microbiol Mol Biol Rev 74: 417–433 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Selmecki A, Forche A, Berman J (2006) Aneuploidy and isochromosome formation in drug‐resistant Candida albicans . Science 313: 367–370 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Taddei F, Radman M, Maynard‐Smith J, Toupance B, Gouyon PH, Godelle B (1997) Role of mutator alleles in adaptive evolution. Nature 387: 700–702 [DOI] [PubMed] [Google Scholar]
- 4. Woodford N, Ellington MJ (2007) The emergence of antibiotic resistance by mutation. Clin Microbiol Infect 13: 5–18 [DOI] [PubMed] [Google Scholar]
- 5. Ford CB, Funt JM, Abbey D, Issi L, Guiducci C, Martinez DA, Delorey T, Li BY, White TC, Cuomo C et al (2015) The evolution of drug resistance in clinical isolates of Candida albicans . Elife 4: e00662 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Haldar K, Bhattacharjee S, Safeukui I (2018) Drug resistance in plasmodium. Nat Rev Microbiol 16: 156–170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Berman J (2016) Ploidy plasticity: a rapid and reversible strategy for adaptation to stress. FEMS Yeast Res 16: pii: fow020 [DOI] [PubMed] [Google Scholar]
- 8. O'Neill J (2014) Review on Antimicrobial Resistance Antimicrobial Resistance: Tackling a crisis for the health and wealth of nations. London: Review on Antimicrobial Resistance. https://amr-revieworg/sites/default/files/AMR Review Paper ‐ Tackling a crisis for the health and wealth of nations_1pdf
- 9. de Kraker ME, Stewardson AJ, Harbarth S (2016) Will 10 million people die a year due to antimicrobial resistance by 2050? PLoS Med 13: e1002184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Pearce EL, Pearce EJ (2013) Metabolic pathways in immune cell activation and quiescence. Immunity 38: 633–643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, Mehler MF, Conway SJ, Ng LG, Stanley ER et al (2010) Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330: 841–845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F et al (2015) Tissue‐resident macrophages originate from yolk‐sac‐derived erythro‐myeloid progenitors. Nature 518: 547–551 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Schulz C, Gomez Perdiguero E, Chorro L, Szabo‐Rogers H, Cagnard N, Kierdorf K, Prinz M, Wu B, Jacobsen SE, Pollard JW et al (2012) A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336: 86–90 [DOI] [PubMed] [Google Scholar]
- 14. Hoeffel G, Chen J, Lavin Y, Low D, Almeida FF, See P, Beaudin AE, Lum J, Low I, Forsberg EC et al (2015) C‐Myb(+) erythro‐myeloid progenitor‐derived fetal monocytes give rise to adult tissue‐resident macrophages. Immunity 42: 665–678 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. van de Laar L, Saelens W, De Prijck S, Martens L, Scott CL, Van Isterdael G, Hoffmann E, Beyaert R, Saeys Y, Lambrecht BN et al (2016) Yolk sac macrophages, fetal liver, and adult monocytes can colonize an empty niche and develop into functional tissue‐resident macrophages. Immunity 44: 755–768 [DOI] [PubMed] [Google Scholar]
- 16. Naderer T, McConville MJ (2008) The Leishmania‐macrophage interaction: a metabolic perspective. Cell Microbiol 10: 301–308 [DOI] [PubMed] [Google Scholar]
- 17. Gordon S (2003) Alternative activation of macrophages. Nat Rev Immunol 3: 23–35 [DOI] [PubMed] [Google Scholar]
- 18. Tannahill GM, Curtis AM, Adamik J, Palsson‐McDermott EM, McGettrick AF, Goel G, Frezza C, Bernard NJ, Kelly B, Foley NH et al (2013) Succinate is an inflammatory signal that induces IL‐1beta through HIF‐1alpha. Nature 496: 238–242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Van den Bossche J, O'Neill LA, Menon D (2017) Macrophage immunometabolism: where are we (going)? Trends Immunol 38: 395–406 [DOI] [PubMed] [Google Scholar]
- 20. O'Neill LA, Pearce EJ (2016) Immunometabolism governs dendritic cell and macrophage function. J Exp Med 213: 15–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Jha AK, Huang SC, Sergushichev A, Lampropoulou V, Ivanova Y, Loginicheva E, Chmielewski K, Stewart KM, Ashall J, Everts B et al (2015) Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity 42: 419–430 [DOI] [PubMed] [Google Scholar]
- 22. Rodriguez‐Prados JC, Traves PG, Cuenca J, Rico D, Aragones J, Martin‐Sanz P, Cascante M, Bosca L (2010) Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation. J Immunol 185: 605–614 [DOI] [PubMed] [Google Scholar]
- 23. Infantino V, Convertini P, Cucci L, Panaro MA, Di Noia MA, Calvello R, Palmieri F, Iacobazzi V (2011) The mitochondrial citrate carrier: a new player in inflammation. Biochem J 438: 433–436 [DOI] [PubMed] [Google Scholar]
- 24. O'Neill LA (2011) A critical role for citrate metabolism in LPS signalling. Biochem J 438: e5–e6 [DOI] [PubMed] [Google Scholar]
- 25. Meiser J, Kramer L, Sapcariu SC, Battello N, Ghelfi J, D'Herouel AF, Skupin A, Hiller K (2016) Pro‐inflammatory macrophages sustain pyruvate oxidation through pyruvate dehydrogenase for the synthesis of itaconate and to enable cytokine expression. J Biol Chem 291: 3932–3946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Wei X, Song H, Yin L, Rizzo MG, Sidhu R, Covey DF, Ory DS, Semenkovich CF (2016) Fatty acid synthesis configures the plasma membrane for inflammation in diabetes. Nature 539: 294–298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Michelucci A, Cordes T, Ghelfi J, Pailot A, Reiling N, Goldmann O, Binz T, Wegner A, Tallam A, Rausell A et al (2013) Immune‐responsive gene 1 protein links metabolism to immunity by catalyzing itaconic acid production. Proc Natl Acad Sci USA 110: 7820–7825 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Cordes T, Wallace M, Michelucci A, Divakaruni AS, Sapcariu SC, Sousa C, Koseki H, Cabrales P, Murphy AN, Hiller K et al (2016) Immunoresponsive gene 1 and itaconate inhibit succinate dehydrogenase to modulate intracellular succinate levels. J Biol Chem 291: 14274–14284 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Mills EL, Ryan DG, Prag HA, Dikovskaya D, Menon D, Zaslona Z, Jedrychowski MP, Costa ASH, Higgins M, Hams E et al (2018) Itaconate is an anti‐inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 556: 113–117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Bambouskova M, Gorvel L, Lampropoulou V, Sergushichev A, Loginicheva E, Johnson K, Korenfeld D, Mathyer ME, Kim H, Huang LH et al (2018) Electrophilic properties of itaconate and derivatives regulate the IkappaBzeta‐ATF3 inflammatory axis. Nature 556: 501–504 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Dominguez‐Andres J, Novakovic B, Li Y, Scicluna BP, Gresnigt MS, Arts RJW, Oosting M, Moorlag S, Groh LA, Zwaag J et al (2018) The itaconate pathway is a central regulatory node linking innate immune tolerance and trained immunity. Cell Metab 29: 211–220.e5 [DOI] [PubMed] [Google Scholar]
- 32. Lampropoulou V, Sergushichev A, Bambouskova M, Nair S, Vincent EE, Loginicheva E, Cervantes‐Barragan L, Ma X, Huang SC, Griss T et al (2016) Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab 24: 158–166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Mills EL, Kelly B, Logan A, Costa AS, Varma M, Bryant CE, Tourlomousis P, Dabritz JH, Gottlieb E, Latorre I et al (2016) Succinate dehydrogenase supports metabolic repurposing of mitochondria to drive inflammatory macrophages. Cell 167: 457–470 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Palsson‐McDermott EM, Curtis AM, Goel G, Lauterbach MA, Sheedy FJ, Gleeson LE, van den Bosch MW, Quinn SR, Domingo‐Fernandez R, Johnston DG et al (2015) Pyruvate kinase M2 regulates Hif‐1alpha activity and IL‐1beta induction and is a critical determinant of the warburg effect in LPS‐activated macrophages. Cell Metab 21: 65–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. West AP, Brodsky IE, Rahner C, Woo DK, Erdjument‐Bromage H, Tempst P, Walsh MC, Choi Y, Shadel GS, Ghosh S (2011) TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature 472: 476–480 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Abuaita BH, Schultz TL, O'Riordan MX (2018) Mitochondria‐derived vesicles deliver antimicrobial reactive oxygen species to control phagosome‐localized Staphylococcus aureus . Cell Host Microbe 24: 625–636 e5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Menu P, Vince JE (2011) The NLRP3 inflammasome in health and disease: the good, the bad and the ugly. Clin Exp Immunol 166: 1–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Lood C, Blanco LP, Purmalek MM, Carmona‐Rivera C, De Ravin SS, Smith CK, Malech HL, Ledbetter JA, Elkon KB, Kaplan MJ (2016) Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus‐like disease. Nat Med 22: 146–153 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, Ramanujan VK, Wolf AJ, Vergnes L, Ojcius DM et al (2012) Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 36: 401–414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. White MJ, McArthur K, Metcalf D, Lane RM, Cambier JC, Herold MJ, van Delft MF, Bedoui S, Lessene G, Ritchie ME et al (2014) Apoptotic caspases suppress mtDNA‐induced STING‐mediated type I IFN production. Cell 159: 1549–1562 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Zhong Z, Liang S, Sanchez‐Lopez E, He F, Shalapour S, Lin XJ, Wong J, Ding S, Seki E, Schnabl B et al (2018) New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature 560: 198–203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Minhas PS, Liu L, Moon PK, Joshi AU, Dove C, Mhatre S, Contrepois K, Wang Q, Lee BA, Coronado M et al (2019) Macrophage de novo NAD(+) synthesis specifies immune function in aging and inflammation. Nat Immunol 20: 50–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Cheng SC, Scicluna BP, Arts RJ, Gresnigt MS, Lachmandas E, Giamarellos‐Bourboulis EJ, Kox M, Manjeri GR, Wagenaars JA, Cremer OL et al (2016) Broad defects in the energy metabolism of leukocytes underlie immunoparalysis in sepsis. Nat Immunol 17: 406–413 [DOI] [PubMed] [Google Scholar]
- 44. Tucey TM, Verma J, Harrison PF, Snelgrove SL, Lo TL, Scherer AK, Barugahare AA, Powell DR, Wheeler RT, Hickey MJ et al (2018) Glucose homeostasis is important for immune cell viability during Candida challenge and host survival of systemic fungal infection. Cell Metab 27: 988–1006 e7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Lachmandas E, Boutens L, Ratter JM, Hijmans A, Hooiveld GJ, Joosten LA, Rodenburg RJ, Fransen JA, Houtkooper RH, van Crevel R et al (2016) Microbial stimulation of different Toll‐like receptor signalling pathways induces diverse metabolic programmes in human monocytes. Nat Microbiol 2: 16246 [DOI] [PubMed] [Google Scholar]
- 46. Vats D, Mukundan L, Odegaard JI, Zhang L, Smith KL, Morel CR, Wagner RA, Greaves DR, Murray PJ, Chawla A (2006) Oxidative metabolism and PGC‐1beta attenuate macrophage‐mediated inflammation. Cell Metab 4: 13–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Odegaard JI, Ricardo‐Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Eagle AR, Vats D, Brombacher F, Ferrante AW et al (2007) Macrophage‐specific PPARgamma controls alternative activation and improves insulin resistance. Nature 447: 1116–1120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Huang SC, Smith AM, Everts B, Colonna M, Pearce EL, Schilling JD, Pearce EJ (2016) Metabolic reprogramming mediated by the mTORC2‐IRF4 signaling axis is essential for macrophage alternative activation. Immunity 45: 817–830 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Van den Bossche J, Baardman J, Otto NA, van der Velden S, Neele AE, van den Berg SM, Luque‐Martin R, Chen HJ, Boshuizen MC, Ahmed M et al (2016) Mitochondrial dysfunction prevents repolarization of inflammatory macrophages. Cell Rep 17: 684–696 [DOI] [PubMed] [Google Scholar]
- 50. Covarrubias AJ, Aksoylar HI, Yu J, Snyder NW, Worth AJ, Iyer SS, Wang J, Ben‐Sahra I, Byles V, Polynne‐Stapornkul T et al (2016) Akt‐mTORC1 signaling regulates Acly to integrate metabolic input to control of macrophage activation. Elife 5: e11612 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Divakaruni AS, Hsieh WY, Minarrieta L, Duong TN, Kim KKO, Desousa BR, Andreyev AY, Bowman CE, Caradonna K, Dranka BP et al (2018) Etomoxir inhibits macrophage polarization by disrupting CoA homeostasis. Cell Metab 28: 490–503 e7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Arts RJ, Gresnigt MS, Joosten LA, Netea MG (2017) Cellular metabolism of myeloid cells in sepsis. J Leukoc Biol 101: 151–164 [DOI] [PubMed] [Google Scholar]
- 53. Hagar JA, Edin ML, Lih FB, Thurlow LR, Koller BH, Cairns BA, Zeldin DC, Miao EA (2017) Lipopolysaccharide potentiates insulin‐driven hypoglycemic shock. J Immunol 199: 3634–3643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Thaiss CA, Levy M, Grosheva I, Zheng D, Soffer E, Blacher E, Braverman S, Tengeler AC, Barak O, Elazar M et al (2018) Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science 359: 1376–1383 [DOI] [PubMed] [Google Scholar]
- 55. Su L, Li H, Xie A, Liu D, Rao W, Lan L, Li X, Li F, Xiao K, Wang H et al (2015) Dynamic changes in amino acid concentration profiles in patients with sepsis. PLoS ONE 10: e0121933 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Langley RJ, Tsalik EL, van Velkinburgh JC, Glickman SW, Rice BJ, Wang C, Chen B, Carin L, Suarez A, Mohney RP et al (2013) An integrated clinico‐metabolomic model improves prediction of death in sepsis. Sci Transl Med 5: 195ra95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Standage SW, Caldwell CC, Zingarelli B, Wong HR (2012) Reduced peroxisome proliferator‐activated receptor alpha expression is associated with decreased survival and increased tissue bacterial load in sepsis. Shock 37: 164–169 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Kapoor A, Shintani Y, Collino M, Osuchowski MF, Busch D, Patel NS, Sepodes B, Castiglia S, Fantozzi R, Bishop‐Bailey D et al (2010) Protective role of peroxisome proliferator‐activated receptor‐beta/delta in septic shock. Am J Respir Crit Care Med 182: 1506–1515 [DOI] [PubMed] [Google Scholar]
- 59. Mukherjee S, Chen LY, Papadimos TJ, Huang S, Zuraw BL, Pan ZK (2009) Lipopolysaccharide‐driven Th2 cytokine production in macrophages is regulated by both MyD88 and TRAM. J Biol Chem 284: 29391–29398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T et al (2014) Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41: 14–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Liu TF, Vachharajani VT, Yoza BK, McCall CE (2012) NAD + ‐dependent sirtuin 1 and 6 proteins coordinate a switch from glucose to fatty acid oxidation during the acute inflammatory response. J Biol Chem 287: 25758–25769 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Zhong L, D'Urso A, Toiber D, Sebastian C, Henry RE, Vadysirisack DD, Guimaraes A, Marinelli B, Wikstrom JD, Nir T et al (2010) The histone deacetylase Sirt6 regulates glucose homeostasis via Hif1alpha. Cell 140: 280–293 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Ip WKE, Hoshi N, Shouval DS, Snapper S, Medzhitov R (2017) Anti‐inflammatory effect of IL‐10 mediated by metabolic reprogramming of macrophages. Science 356: 513–519 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Dominguez‐Andres J, Netea MG (2018) Long‐term reprogramming of the innate immune system. J Leukoc Biol 105: 329–338 [DOI] [PubMed] [Google Scholar]
- 65. Arts RJW, Moorlag S, Novakovic B, Li Y, Wang SY, Oosting M, Kumar V, Xavier RJ, Wijmenga C, Joosten LAB et al (2018) BCG vaccination protects against experimental viral infection in humans through the induction of cytokines associated with trained immunity. Cell Host Microbe 23: 89–100 e5 [DOI] [PubMed] [Google Scholar]
- 66. Cheng SC, Quintin J, Cramer RA, Shepardson KM, Saeed S, Kumar V, Giamarellos‐Bourboulis EJ, Martens JH, Rao NA, Aghajanirefah A et al (2014) mTOR‐ and HIF‐1alpha‐mediated aerobic glycolysis as metabolic basis for trained immunity. Science 345: 1250684 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Novakovic B, Habibi E, Wang SY, Arts RJW, Davar R, Megchelenbrink W, Kim B, Kuznetsova T, Kox M, Zwaag J et al (2016) beta‐glucan reverses the epigenetic state of LPS‐induced immunological tolerance. Cell 167: 1354–1368 e14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Saeed S, Quintin J, Kerstens HH, Rao NA, Aghajanirefah A, Matarese F, Cheng SC, Ratter J, Berentsen K, van der Ent MA et al (2014) Epigenetic programming of monocyte‐to‐macrophage differentiation and trained innate immunity. Science 345: 1251086 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Arts RJ, Novakovic B, Ter Horst R, Carvalho A, Bekkering S, Lachmandas E, Rodrigues F, Silvestre R, Cheng SC, Wang SY et al (2016) Glutaminolysis and fumarate accumulation integrate immunometabolic and epigenetic programs in trained immunity. Cell Metab 24: 807–819 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Christ A, Gunther P, Lauterbach MAR, Duewell P, Biswas D, Pelka K, Scholz CJ, Oosting M, Haendler K, Bassler K et al (2018) Western diet triggers NLRP3‐dependent innate immune reprogramming. Cell 172: 162–175 e14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Kaufmann E, Sanz J, Dunn JL, Khan N, Mendonca LE, Pacis A, Tzelepis F, Pernet E, Dumaine A, Grenier JC et al (2018) BCG educates hematopoietic stem cells to generate protective innate immunity against tuberculosis. Cell 172: 176–190 e19 [DOI] [PubMed] [Google Scholar]
- 72. Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, Becker CD, See P, Price J, Lucas D et al (2013) Tissue‐resident macrophages self‐maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38: 792–804 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Yao Y, Jeyanathan M, Haddadi S, Barra NG, Vaseghi‐Shanjani M, Damjanovic D, Lai R, Afkhami S, Chen Y, Dvorkin‐Gheva A et al (2018) Induction of autonomous memory alveolar macrophages requires t cell help and is critical to trained immunity. Cell 175: 1634–1650 e17 [DOI] [PubMed] [Google Scholar]
- 74. Billig S, Schneefeld M, Huber C, Grassl GA, Eisenreich W, Bange FC (2017) Lactate oxidation facilitates growth of Mycobacterium tuberculosis in human macrophages. Sci Rep 7: 6484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Escoll P, Song OR, Viana F, Steiner B, Lagache T, Olivo‐Marin JC, Impens F, Brodin P, Hilbi H, Buchrieser C (2017) Legionella pneumophila modulates mitochondrial dynamics to trigger metabolic repurposing of infected macrophages. Cell Host Microbe 22: 302–316 e7 [DOI] [PubMed] [Google Scholar]
- 76. Czyz DM, Willett JW, Crosson S (2017) Brucella abortus induces a Warburg shift in host metabolism that is linked to enhanced intracellular survival of the pathogen. J Bacteriol 199: e00227–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Gillmaier N, Gotz A, Schulz A, Eisenreich W, Goebel W (2012) Metabolic responses of primary and transformed cells to intracellular Listeria monocytogenes. PLoS ONE 7: e52378 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Kloehn J, Blume M, Cobbold S, Saunders E, Dagley M, McConville M (2016) Using metabolomics to dissect host–parasite interactions. Curr Opin Microbiol 32: 59–65 [DOI] [PubMed] [Google Scholar]
- 79. Creek DJ, Mazet M, Achcar F, Anderson J, Kim D‐H, Kamour R, Morand P, Millerioux Y, Biran M, Kerkhoven EJ et al (2015) Probing the metabolic network in bloodstream‐form Trypanosoma brucei using untargeted metabolomics with stable isotope labelled glucose. PLoS Pathog 11: e1004689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Oppenheim RD, Creek DJ, Macrae JI, Modrzynska KK, Pino P, Limenitakis J, Polonais V, Seeber F, Barrett MP, Billker O et al (2014) BCKDH: the missing link in apicomplexan mitochondrial metabolism is required for full virulence of Toxoplasma gondii and Plasmodium berghei . PLoS Pathog 10: e1004263 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Cumming BM, Addicott KW, Adamson JH, Steyn AJ (2018) Mycobacterium tuberculosis induces decelerated bioenergetic metabolism in human macrophages. Elife 7: e39169 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Marrero J, Trujillo C, Rhee KY, Ehrt S (2013) Glucose phosphorylation is required for Mycobacterium tuberculosis persistence in mice. PLoS Pathog 9: e1003116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Puckett S, Trujillo C, Wang Z, Eoh H, Ioerger TR, Krieger I, Sacchettini J, Schnappinger D, Rhee KY, Ehrt S (2017) Glyoxylate detoxification is an essential function of malate synthase required for carbon assimilation in Mycobacterium tuberculosis . Proc Natl Acad Sci USA 114: E2225–E2232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. McKinney JD, Honer zu Bentrup K, Munoz‐Elias EJ, Miczak A, Chen B, Chan WT, Swenson D, Sacchettini JC, Jacobs WR Jr, Russell DG (2000) Persistence of Mycobacterium tuberculosis in macrophages and mice requires the glyoxylate shunt enzyme isocitrate lyase. Nature 406: 735–738 [DOI] [PubMed] [Google Scholar]
- 85. Murima P, Zimmermann M, Chopra T, Pojer F, Fonti G, Dal Peraro M, Alonso S, Sauer U, Pethe K, McKinney JD (2016) A rheostat mechanism governs the bifurcation of carbon flux in mycobacteria. Nat Commun 7: 12527 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Russell DG, Vanderven BC, Lee W, Abramovitch RB, Kim M‐J, Homolka S, Niemann S, Rohde KH (2010) Mycobacterium tuberculosis wears what it eats. Cell Host Microbe 8: 68–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Uwamahoro N, Verma‐Gaur J, Shen HH, Qu Y, Lewis R, Lu J, Bambery K, Masters SL, Vince JE, Naderer T et al (2014) The pathogen Candida albicans hijacks pyroptosis for escape from macrophages. MBio 5: e00003–e00014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Wellington M, Koselny K, Sutterwala FS, Krysan DJ (2014) Candida albicans triggers NLRP3‐mediated pyroptosis in macrophages. Eukaryot Cell 13: 329–340 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Lorenz MC, Fink GR (2001) The glyoxylate cycle is required for fungal virulence. Nature 412: 83–86 [DOI] [PubMed] [Google Scholar]
- 90. Barelle CJ, Priest CL, Maccallum DM, Gow NA, Odds FC, Brown AJ (2006) Niche‐specific regulation of central metabolic pathways in a fungal pathogen. Cell Microbiol 8: 961–971 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Ramirez MA, Lorenz MC (2007) Mutations in alternative carbon utilization pathways in Candida albicans attenuate virulence and confer pleiotropic phenotypes. Eukaryot Cell 6: 280–290 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Piekarska K, Mol E, van den Berg M, Hardy G, van den Burg J, van Roermund C, MacCallum D, Odds F, Distel B (2006) Peroxisomal fatty acid beta‐oxidation is not essential for virulence of Candida albicans . Eukaryot Cell 5: 1847–1856 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Lorenz MC, Bender JA, Fink GR (2004) Transcriptional response of Candida albicans upon internalization by macrophages. Eukaryot Cell 3: 1076–1087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Fradin C, Kretschmar M, Nichterlein T, Gaillardin C, d'Enfert C, Hube B (2003) Stage‐specific gene expression of Candida albicans in human blood. Mol Microbiol 47: 1523–1543 [DOI] [PubMed] [Google Scholar]
- 95. Fradin C, De Groot P, MacCallum D, Schaller M, Klis F, Odds FC, Hube B (2005) Granulocytes govern the transcriptional response, morphology and proliferation of Candida albicans in human blood. Mol Microbiol 56: 397–415 [DOI] [PubMed] [Google Scholar]
- 96. Zakikhany K, Naglik JR, Schmidt‐Westhausen A, Holland G, Schaller M, Hube B (2007) In vivo transcript profiling of Candida albicans identifies a gene essential for interepithelial dissemination. Cell Microbiol 9: 2938–2954 [DOI] [PubMed] [Google Scholar]
- 97. Thewes S, Kretschmar M, Park H, Schaller M, Filler SG, Hube B (2007) In vivo and ex vivo comparative transcriptional profiling of invasive and non‐invasive Candida albicans isolates identifies genes associated with tissue invasion. Mol Microbiol 63: 1606–1628 [DOI] [PubMed] [Google Scholar]
- 98. Amorim‐Vaz S, Tran Vdu T, Pradervand S, Pagni M, Coste AT, Sanglard D (2015) RNA enrichment method for quantitative transcriptional analysis of pathogens in vivo applied to the fungus Candida albicans . MBio 6: e00942 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Hebecker B, Vlaic S, Conrad T, Bauer M, Brunke S, Kapitan M, Linde J, Hube B, Jacobsen ID (2016) Dual‐species transcriptional profiling during systemic candidiasis reveals organ‐specific host‐pathogen interactions. Sci Rep 6: 36055 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Perez JC, Kumamoto CA, Johnson AD (2013) Candida albicans commensalism and pathogenicity are intertwined traits directed by a tightly knit transcriptional regulatory circuit. PLoS Biol 11: e1001510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Askew C, Sellam A, Epp E, Hogues H, Mullick A, Nantel A, Whiteway M (2009) Transcriptional regulation of carbohydrate metabolism in the human pathogen Candida albicans . PLoS Pathog 5: e1000612 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Andes D, Lepak A, Pitula A, Marchillo K, Clark J (2005) A simple approach for estimating gene expression in Candida albicans directly from a systemic infection site. J Infect Dis 192: 893–900 [DOI] [PubMed] [Google Scholar]
- 103. Sandai D, Yin Z, Selway L, Stead D, Walker J, Leach MD, Bohovych I, Ene IV, Kastora S, Budge S et al (2012) The evolutionary rewiring of ubiquitination targets has reprogrammed the regulation of carbon assimilation in the pathogenic yeast Candida albicans . MBio 3: e00495–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Ballou ER, Avelar GM, Childers DS, Mackie J, Bain JM, Wagener J, Kastora SL, Panea MD, Hardison SE, Walker LA et al (2016) Lactate signalling regulates fungal beta‐glucan masking and immune evasion. Nat Microbiol 2: 16238 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Saunders EC, Ng WW, Kloehn J, Chambers JM, Ng M, McConville MJ (2014) Induction of a stringent metabolic response in intracellular stages of Leishmania mexicana leads to increased dependence on mitochondrial metabolism. PLoS Pathog 10: e1003888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Naderer T, Ellis MA, Sernee MF, De Souza DP, Curtis J, Handman E, McConville MJ (2006) Virulence of Leishmania major in macrophages and mice requires the gluconeogenic enzyme fructose‐1,6‐bisphosphatase. Proc Natl Acad Sci USA 103: 5502–5507 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Saunders EC, Naderer T, Chambers J, Landfear SM, McConville MJ (2018) Leishmania mexicana can utilize amino acids as major carbon sources in macrophages but not in animal models. Mol Microbiol 108: 143–158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Lemaitre B, Girardin SE (2013) Translation inhibition and metabolic stress pathways in the host response to bacterial pathogens. Nat Rev Microbiol 11: 365–369 [DOI] [PubMed] [Google Scholar]
- 109. Naderer T, Wee E, McConville MJ (2008) Role of hexosamine biosynthesis in Leishmania growth and virulence. Mol Microbiol 69: 858–869 [DOI] [PubMed] [Google Scholar]
- 110. Naderer T, Heng J, Saunders EC, Kloehn J, Rupasinghe TW, Brown TJ, McConville MJ (2015) Intracellular survival of Leishmania major depends on uptake and degradation of extracellular matrix glycosaminoglycans by macrophages. PLoS Pathog 11: e1005136 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Abu Kwaik Y, Bumann D (2015) Host delivery of favorite meals for intracellular pathogens. PLoS Pathog 11: e1004866 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Sanman LE, Qian Y, Eisele NA, Ng TM, van der Linden WA, Monack DM, Weerapana E, Bogyo M (2016) Disruption of glycolytic flux is a signal for inflammasome signaling and pyroptotic cell death. Elife 5: e13663 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Wickersham M, Wachtel S, Wong Fok Lung T, Soong G, Jacquet R, Richardson A, Parker D, Prince A (2017) Metabolic stress drives keratinocyte defenses against Staphylococcus aureus Infection. Cell Rep 18: 2742–2751 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Stoudt K, Chawla S (2018) Don't sugar coat it: glycemic control in the intensive care unit. J Intensive Care Med 10.1177/0885066618801748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Weis S, Carlos AR, Moita MR, Singh S, Blankenhaus B, Cardoso S, Larsen R, Rebelo S, Schauble S, Del Barrio L et al (2017) Metabolic adaptation establishes disease tolerance to sepsis. Cell 169: 1263–1275 e14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Walker SD, Kalviainen R (2011) Non‐vision adverse events with vigabatrin therapy. Acta Neurol Scand Suppl 192: 72–82 [DOI] [PubMed] [Google Scholar]
- 117. Rosler B, Wang X, Keating ST, Joosten LAB, Netea MG, van de Veerdonk FL (2018) HDAC inhibitors modulate innate immune responses to micro‐organisms relevant to chronic mucocutaneous candidiasis. Clin Exp Immunol 194: 205–219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Shih CJ, Wu YL, Chao PW, Kuo SC, Yang CY, Li SY, Ou SM, Chen YT (2015) Association between use of oral anti‐diabetic drugs and the risk of sepsis: a nested case‐control study. Sci Rep 5: 15260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Fensterheim BA, Young JD, Luan L, Kleinbard RR, Stothers CL, Patil NK, McAtee‐Pereira AG, Guo Y, Trenary I, Hernandez A et al (2018) The TLR4 agonist monophosphoryl lipid A drives broad resistance to infection via dynamic reprogramming of macrophage metabolism. J Immunol 200: 3777–3789 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Rice PJ, Adams EL, Ozment‐Skelton T, Gonzalez AJ, Goldman MP, Lockhart BE, Barker LA, Breuel KF, Deponti WK, Kalbfleisch JH et al (2005) Oral delivery and gastrointestinal absorption of soluble glucans stimulate increased resistance to infectious challenge. J Pharmacol Exp Ther 314: 1079–1086 [DOI] [PubMed] [Google Scholar]
- 121. Mourits VP, Wijkmans JC, Joosten LA, Netea MG (2018) Trained immunity as a novel therapeutic strategy. Curr Opin Pharmacol 41: 52–58 [DOI] [PubMed] [Google Scholar]
- 122. Arts RJW, Carvalho A, La Rocca C, Palma C, Rodrigues F, Silvestre R, Kleinnijenhuis J, Lachmandas E, Goncalves LG, Belinha A et al (2016) Immunometabolic pathways in BCG‐induced trained immunity. Cell Rep 17: 2562–2571 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Vachharajani VT, Liu T, Brown CM, Wang X, Buechler NL, Wells JD, Yoza BK, McCall CE (2014) SIRT1 inhibition during the hypoinflammatory phenotype of sepsis enhances immunity and improves outcome. J Leukoc Biol 96: 785–796 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Delsing CE, Gresnigt MS, Leentjens J, Preijers F, Frager FA, Kox M, Monneret G, Venet F, Bleeker‐Rovers CP, van de Veerdonk FL et al (2014) Interferon‐gamma as adjunctive immunotherapy for invasive fungal infections: a case series. BMC Infect Dis 14: 166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Wang A, Huen SC, Luan HH, Yu S, Zhang C, Gallezot JD, Booth CJ, Medzhitov R (2016) Opposing effects of fasting metabolism on tissue tolerance in bacterial and viral inflammation. Cell 166: 1512–1525 e12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Rao S, Schieber AM, O'Connor CP, Leblanc M, Michel D, Ayres JS (2017) Pathogen‐mediated inhibition of anorexia promotes host survival and transmission. Cell 168: 503–516e12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Sanchez KK, Chen GY, Schieber AMP, Redford SE, Shokhirev MN, Leblanc M, Lee YM, Ayres JS (2018) Cooperative metabolic adaptations in the host can favor asymptomatic infection and select for attenuated virulence in an enteric pathogen. Cell 175: 146–158.e15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Peleg AY, Hogan DA, Mylonakis E (2010) Medically important bacterial‐fungal interactions. Nat Rev Microbiol 8: 340–349 [DOI] [PubMed] [Google Scholar]
