Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

|

Received: 5 March 2020    Accepted: 10 March 2020

DOI: 10.1111/imr.12858

INTRODUCTION

Immunometabolism: From basic mechanisms to translation

Liza Makowski1  | Mehdi Chaib2 | Jeffrey C. Rathmell3

1
Division of Hematology and Oncology,
Department of Medicine, University of Abstract
Tennessee Health Science Center, Memphis, Immunometabolism has emerged as a major mechanism central to adaptive and in-
Tennessee
2 nate immune regulation. From early observations that inflammatory cytokines were
Department of Pharmaceutical Sciences,
College of Pharmacy, University of induced in obese adipose tissue and that these cytokines contributed to metabolic
Tennessee Health Science Center, Memphis,
disease, it was clear that metabolism and the immunological state are inextricably
Tennessee
3
Department of Pathology, Microbiology,
linked. With a second research wave arising from studies in cancer metabolism to also
and Immunology, Vanderbilt Center for study the intrinsic metabolic pathways of immune cells themselves and how those
Immunobiology, Vanderbilt University
Medical Center, Nashville, Tennessee
pathways influence cell fate and function, immunometabolism is a rapidly maturing
area of research. Several key themes and goals drive the field. There is abundant
Correspondence
Jeffrey C. Rathmell, Department of
evidence that metabolic pathways are closely tied to cell signaling and differentiation
Pathology, Microbiology, and Immunology, which leads different subsets of immune cells to adopt unique metabolic programs
Vanderbilt Center for Immunobiology,
Vanderbilt University Medical Center,
specific to their state and environment. In this way, metabolic signaling drives cell
Nashville, Tennessee TN 37232, USA. fate. It is also apparent that microenvironment greatly influences cell metabolism.
Email: jeff.rathmell@vanderbilt.edu
Immune cells adopt programs specific for the tissues where they infiltrate and reside.
Ultimately, a central goal of the field is to apply immunometabolism findings to the
discovery of novel therapeutic strategies in a wide range of diseases, including can-
cer, autoimmunity, and metabolic syndrome. This review summarizes these facets of
immunometabolism and highlights opportunities for clinical translation.

KEYWORDS

autoimmunity, immune-mediated diseases, infectious diseases, met

1 |  I NTRO D U C TI O N immune suppressive,1 Hotamisligil et al showed in a landmark


paper in 1993 that adipose tissue produced TNFα in murine mod-
Two of the most fundamental features of multicellular life are the els of obesity and diabetes2 to demonstrate that overnutrition can
need to effectively distribute nutrients across cells and tissues and promote inflammation. Inflammation drives insulin resistance and
to protect from infection and injury. It is not surprising, therefore, features of diabetes and metabolic syndrome. 3 In addition to met-
that links between metabolism and immunity have become apparent abolic tissues regulating immune cells, the metabolism of immune
in the past twenty years. Indeed, inflammation is now known to me- cells themselves is highly regulated. Warburg described metabolic
diate many of the pathologies of metabolic syndrome. Conversely, changes and aerobic glycolysis in cancer cells early in the 20th cen-
the metabolic programs of individual cells of the immune system tury4 that are now known to be driven by activated oncogenic sig-
are under tight control and strongly influence cell function and naling pathways. 5 Those same signaling pathways are activated to
differentiation. While malnutrition has long been known as widely promote aerobic glycolysis in stimulated immune cells6-8 and play
key roles to reprogram metabolism from catabolic oxidative path-
This article introduces a series of reviews covering Immunometabolism: From Basic ways to anabolic pathways.
Mechanisms to Translation appearing in Volume 295 of Immunological Reviews.

© 2020 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Immunological Reviews. 2020;295:5–14.  |


wileyonlinelibrary.com/journal/imr     5
|
6       MAKOWSKI et al.

Each hematopoietic cell type has clearly defined morpho- pathways to be targeted or modified in disease are reviewed here
logical, expression, and functional characteristics with defined and this volume (Figure 1).
subtypes that are continuing to be defined as technological ap-
proaches enabling increased sensitivity arise. However, these var-
ied lineages and subsets of each cell type also reside in or travel 2 | BA S I C M EC H A N I S M S TH AT R EG U L ATE
to distinct tissue depots which impact function. In this light, it is I M M U N E M E TA B O LI S M
not surprising that each population of immune cells has a distinct
metabolism and nutrient usage. First demonstrated by Vats et al9 The metabolism of T cells has been an area of intense interest and
that macrophage metabolism can influence production of inflam- has many potential biological and therapeutic implications that illus-
matory cytokines as alternatively activated macrophages rely on trate the potential of immunometabolism. The majority of research
a different metabolic program than classically activated macro- has focused on uptake and metabolism of glucose, amino acids pri-
phages, this principle has since been shown in T cells,10 myeloid marily glutamine, and certain fatty acids, although other metabolites
derived suppressor cells (MDSCs),11 and dendritic cells (DCs).12 impact metabolism. When T cells are stimulated through the anti-
The cytokines and signaling pathways that guide distinct immune gen receptor, CD28 costimulation can increase glucose transporter
responses also promote these specific programs that support bio- GLUT1 expression, glucose uptake and subsequent glycolysis, and
energetics and biosynthesis. The intersection of metabolism and mitochondrial capacity.6,13,14 Conversely, CD28 family inhibitory re-
immunity at the systemic and cellular levels now forms the rap- ceptors cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and
idly evolving field of immunometabolism. The most up-to-date programmed cell death-1 (PD-1) can suppress this metabolic tran-
concepts on the control of cell metabolism through cell intrinsic sition.15,16 T cells that fail to receive CD28 costimulation become
signaling mechanisms, the effect of the tissue microenvironment anergic and are metabolically suppressed (ie, have low uptake of
and nutrient availability on metabolism, and the potential for these substrates and little mitochondrial metabolism).17 The metabolic

F I G U R E 1   Immunometabolism: From Basic Mechanisms to Translation—Graphical summary of reviews in this collection. This figure
summarizes the series of reviews provided by experts in the field demonstrating the mechanisms of immunometabolism, impacts on certain
tissues such as adipose tissue, and disease states such as inflammation, cancer, and lupus. ATM: adipose tissue macrophage; VAT: visceral
adipose tissue; OXPHOS: oxidative phosphorylation
MAKOWSKI et al. |
      7

program of T cells as they differentiate into different subsets and in immunometabolism and cell fate. mTOR acts as part of two pro-
fates, however, is not shared. While effector T cells induce aerobic tein complexes, with the predominant immunometabolic regulation
glycolysis, it was shown that memory T cells instead rely on mito- occurring through the actions of mTOR complex I (mTORC1) that is
chondrial metabolism and lipid oxidation18,19 but can rapidly revert activated downstream of receptor stimulation of phosphatidylinosi-
to glycolysis upon restimulation through endoplasmic reticulum- tol 3-kinase (PI3K) and Akt. As described by Huang et al34 the PI3K/
20
mitochondrial direct interactions that serve as metabolic hubs. Akt/mTORC1 pathway serves as a central mechanism to sense and
Unlike effector T cell subsets, regulatory T cells (Tregs) do not re- integrate nutrient availability and signaling to promote T cell metab-
quire GLUT1 or high levels of glutamine uptake through the amino olism and function. mTORC1 is activated at lysosomes through the
acid transporter ASCT221,22 and instead predominantly rely on mi- combined actions of T cell activation and signaling through PI3K and
10,23-25
tochondrial lipid, pyruvate, and lactate oxidation. Tregs can be Akt that lead to activation of the Rheb GTPase and the coordinated
highly glycolytic, but the primary Treg transcription factor, FoxP3, sensing of essential amino acids through the sestrin/GATOR com-
has also been shown to repress glycolysis and high rates of glucose plex. The lysosomal localization plays a key role in this process as
metabolism can impair Treg suppressive capacity. 24,26,27 Individual the amino acid transporter SLC38a9 transports arginine from the
effector T cell populations also differ, as glutamine metabolism is lysosome allow the ability of the sestrin/GATOR complex to sense
essential for CD4 Th17 cells, but effector differentiation of CD4 leucine and isoleucine and activate mTORC1.35,36 When activated,
Th1 and CD8 cytotoxic T cells as well as that of classically activated mTORC1 regulates a host of cellular functions to promote the transi-
M1-like macrophages is restrained by this pathway. 28-30 These dif- tion from quiescence to anabolic metabolism and proliferation while
ferences likely reflect varied modes of regulation, roles, and require- suppressing catabolic metabolism. A key target for mTORC1 is the
ments of specific tissue sites, and offer distinct opportunities to suppression of the AMP-activated protein kinase (AMPK) by phos-
selectively modulate immunity (Figure 2). phorylation of ULK1. AMPK is activated when cells experience en-
Multiple signaling mechanisms coordinate after immune cells ergy stress to maximize oxidative pathways and promote autophagy,
are stimulated to mediate metabolic reprogramming and the spe- but also can have a role in normal physiology to balance mTORC1 ac-
cific differences of each cell type and subset. In addition to tran- tivity.37,38 The generally shared aerobic glycolysis processes across
scriptional regulation through Myc and hypoxia-inducible factor effector T cells may suggest similar dependence on mTORC1. Yet,
(HIF1α) that drive induction of anabolic genes for glycolysis and it is clear that the mTORC1 pathway has distinct roles in each ef-
8,31,32 33
mitochondrial metabolism in T cells and macrophages, the fector T cell subset and that a balanced and dynamically regulated
mechanistic target of rapamycin (mTOR) pathway plays a critical role metabolism may be the key to these differential roles. For instance,

F I G U R E 2   Metabolism regulates immune cell activation. Glycolytic metabolism in immune cells typically leads to activation of an effector
phenotype (eg, effector T, M1-like macrophages, DC, NK, and B cells), whereas oxidative metabolism of substrates such as fatty acids (lipid)
and amino acids including glutamine leads to a regulatory or memory phenotype. Immunometabolism is necessary for not only cellular
energetics, but also contributes to biosynthetic intermediates and signaling
|
8       MAKOWSKI et al.

while CD4 Th1 cells rely on the mTORC1 activator protein Rheb, Th2 acid (TCA) cycle after pro-inflammatory stimuli. A main function of in-
has been shown capable of Rheb-independent activation. This may, hibition of the TCA cycle is that this allows excess citrate to feed into
however, reflect graded dependencies on the levels of mTORC1 ac- other anabolic pathways and succinate to promote inflammation.49,50
tivity for each effector T cell subtype. Indeed, Tregs are dependent Also, production of the bioactive metabolite itaconate is stimulated
on a graded level of mTORC1 activity. In the absence of mTORC1 by pro-inflammatory stimuli. Itaconate plays an anti-microbial role but
function, Tregs fail to suppress effector T cells and severe autoim- more critically it blunts succinate dehydrogenase (SDH) enzymatic
munity can ensue.39,40 However, AMPK can promote Treg accumula- function,51 leading to a build-up of succinate. The block at SDH leads to
10
tion in vivo and may do so in part by moderating mTORC1 activity, the second break in the TCA cycle which promotes mitochondrial ROS
as the mTORC1 inhibitor rapamycin can promote development of production, HIF1α stabilization, and a continued commitment to gly-
41
Treg. These findings about regulator and effector T cells demon- colysis in the activated cell. In addition to inhibition of SDH, itaconate
strate that factors that balance and fine-tune mTORC1 signaling leads to activation of a potent anti-inflammatory and anti-oxidant tran-
function to integrate microenvironmental nutrients and signals and scription factor nuclear factor erythroid 2—related factor 2 (Nrf2).52
are fundamental elements of immunometabolic signaling. There is great interest in itaconate or its derivatives in therapeutic
B cell metabolism is also highly regulated and undergoes repro- approaches due to its ability to alkylate cysteines on metabolically
gramming as cells progress through development and differentiation. sensitive enzymes. In contrast, alternative M2-like activation of macro-
B cell activation and cytokine stimulation promote glycolysis42,43 much phages is reliant upon TCA cycle—mediated metabolism of glutamine,
like T cells, but other pathways can differ. Indeed, flux through the pen- glucose, and fatty acids.9,53 Indeed, transgenic expression of Glut1 in-
44
tose phosphate pathway may be limiting in B cells and mitochondrial creased classically activated macrophage inflammatory function, while
activity is tightly regulated that may influence susceptibility to apop- genetic deletion of Glut1 specifically repressed those functions.54,55
45
tosis. Germinal centers also provide a dynamic microenvironment Lastly, Bakker and Pearce touch upon the role of mitochondrial
which may be hypoxic to drive glycolysis and alter B cell proliferation metabolism downstream of a little studied pathway called hypu-
and fate,31 although increased fatty acid metabolism in germinal center sination through the polyamine spermidine derived from arginine,
localized B cells has also been reported.46 How these changes relate an amino acid well studied in macrophage metabolism. 56 Inhibition
to the particular activation and differentiation states of B cells as they of hypusination of eukaryotic initiation factor 5 (eIF5A) led to the
transition through activation, germinal centers, and plasmablast differ- reduction of alternatively activated macrophage markers, TCA
47
entiation is discussed in detail by Jellusova. Because germinal center cycle flux, fewer electron transport chain (ETC) components, and
B cells undergo class switch DNA recombination and somatic hyper- an overall reduced OXPHOS metabolism. Surprisingly, inhibition of
mutation, errors leading to mutations can occur. Hence, many of the this pathway had no effect on pro-inflammatory macrophage activa-
factors that drive B cell activation and metabolism can also lead to ma- tion. These disparate findings emphasize the fact that mitochondrial
lignant transformation. The mismatch between the oncogene-driven metabolism in pro-inflammatory macrophages is not to fuel energy
metabolic demands and the metabolic pathways and flux meant to production but to generate metabolic intermediates, whereas in al-
support those demands, however, can lead to B cell apoptosis. Thus, ternatively activated macrophages, OXPHOS is necessary for the
Jellusova et al emphasize that understanding mechanisms that regu- M2 phenotype.
late B cell metabolism, therefore, informs normal B cell biology as well Another innate mechanism of immunometabolic regulation is
as B cell transformation to leukemia and lymphoma. Ultimately, anti- through the complement system. The complement system is a pat-
body-producing B cells have clear metabolic demands to support high tern recognition receptor (PRR) system that has newly identified
rates of protein synthesis; however, the metabolic requirements of roles to regulate basic T cell metabolism and physiology.57 In addi-
plasmablasts and long-lived plasma cells remain uncertain. tion to secreted complement that arises from the liver, West et al re-
Innate immune cells also undergo metabolic programming and use view recent findings showing that T cells produce small amounts of
metabolic pathways to control cell fate. Bakker and Pearce provide a intracellular forms of complement.58 C3 and C5 in this setting are
detailed review on the past decade of progress in understanding im- each cleaved to C3a and C3b and C5a and C5b by intracellular pro-
48
munometabolism with a focus on DC and macrophage metabolism. teases. A subsequent set of intracellular receptors then recognize
Mononuclear phagocytes are important in sampling the environment these ligands that promote glycolysis and lipid oxidation. Indeed,
and crosstalk with neighboring cells, effectively bridging innate and West et al speculate that complement arose in single-celled organ-
adaptive immunity. Appropriate control over cell metabolism is nec- isms as a metabolic and stress-sensing mechanism that subsequently
essary for these interactions and acts to fine-tune inflammatory re- evolved to recognize and help eliminate pathogens. This intracellular
sponses. After lipopolysaccharide (LPS) stimulation, DCs exhibit a brief system may be essential for cell viability, as individuals with comple-
burst of oxidative phosphorylation (OXPHOS) that is downregulated ment deficiency syndromes nevertheless have been shown to retain
after several hours to allow cells to engage aerobic glycolysis to fuel expression of intracellular complement components. In addition to
the need for metabolic intermediates. Macrophages have similar met- direct actions of complement, the complement regulatory cofactor
abolic reprogramming after danger signal activation such as LPS. Much CD46 is a human-specific complement receptor which can promote
of the foundational work for this myeloid immunometabolism arose T cell glycolysis in response to engagement with extracellular com-
from work in macrophages showing two breaks in the tricarboxylic plement.57 While the CD46 proximal signals are not certain, the
MAKOWSKI et al. |
      9

Notch and PI3K pathways are likely candidates to then promote T (ATM) where extrinsic effects on local and systemic immunometab-
cell glycolysis and inflammation. Importantly, individuals deficient olism, as well as intrinsic immunometabolism and effects on phe-
in CD46 have T cell immunodeficiencies. Overall, complement notype, are discussed. From the first report of differences in ATM
has been previously recognized to promote defense from invading characteristics between lean and obese,67 the evolution of the ATM
pathogens, but current data support an additional role to maintain phenotype has varied. In short, in vitro differentiation studies to
cell survival and tissue homeostasis. generate pro-inflammatory M1-like macrophages vs. anti-inflamma-
One approach to identify novel immunometabolic regulators is to tory immunomodulatory M2-like macrophages failed to hold true in
take a human genetics strategy. In particular, Mendelian genetic met- vivo. Advances in flow cytometry, proteomics, metabolomics, and
abolic disorders provide insight into genes and pathways essential for single cell sequencing of macrophages have shown varied pheno-
normal metabolic function and influence on immunity, as reviewed in types that blend both classical M1-like and alternative M2-like mark-
detail by McGuire.59 While the majority of these diverse disorders ers depending on the age of mice, duration on diet, and extent of
have not presented apparent immunological phenotypes, immune obesity or weight loss. The interaction of ATM with oxidized lipids
cells have not yet been extensively explored in these settings. With and other metabolites secreted from adipocytes or varied stromal
clear examples, such as glycogen storage disease-1b that leads to cells in adipose impacts inflammation and insulin resistance.68 Thus,
lymphopenia and diminished ability of T cells to upregulate glycol- it is increasingly important to assess the metabolic and cellular inter-
ysis, it is apparent that these human genetic diseases can provide actions in the adipose tissue.
a wide-ranging opportunity for immunometabolic discovery. Indeed, In addition to macrophages, a unique visceral fat—specific Treg
screening patients with immune disorders for metabolic phenotypes population was recently defined that functions to keep this vast
recently identified gain-of-function mutations in SDH,60 whereas depot of adipose tissue quiescent in terms of inflammation to max-
polymorphisms in ETC proteins were found to lead to increased in- imally sensitize insulin signaling.69,70 A review by Li and Spallanzani
cidences of infection.61 In sum, taking this genetic approach based et al details a critical role of visceral adipose tissue (VAT) mesenchy-
on human disease coupled with biochemical analyses of T cells may mal stromal cells (VmSCs) in regulating VAT Tregs, local, and systemic
reveal new immunometabolic regulators and pathways. inflammation in obesity.71 VAT Tregs were first identified in 2009
and comprise 40%-80% of CD4+ T cells. VAT Tregs are distinct from
lymphoid tissue—associated Tregs which comprise only 5%-15% of
3 | I M M U N O M E TA B O LI S M I N TI S S U E S CD4T cells in that they are derived from the thymus, become more
“VAT like” in the spleen (a poorly understood step), and then seed
Much of the work on immunometabolism has been performed by VAT early in life. VAT Tregs proliferate and survive in VAT, rather
necessity in vitro and with immune cells from secondary lymphoid than result from conversion or recruitment. VAT Tregs are typically
organs, such as spleen and lymph nodes. This is due to technical more clonal in a MHCII VAT antigen presenting cell-dependent man-
challenges caused by the need for large numbers of cells for many ner and less heterogeneous than lymphoid-resident Tregs. VAT Tregs
biochemical assays and of the rarity and heterogeneity of specific function to improve insulin sensitivity, reduce pro-inflammatory
immune cell populations in other tissues. These barriers are rapidly macrophages, and increase anti-inflammatory macrophages and re-
being overcome due to technical advances, and it is clear that while duce inflammation in adipocytes, among other functions. VAT Tregs
metabolism based on in vitro assays can teach us a great deal about are highly dependent upon the nuclear hormone receptor PPARγ
immunometabolism, pathways in vivo can be regulated differently. (distal to TCR signaling) and have a unique transcriptome with many
13
For example, it was recently shown using C-glucose tracing that lipid metabolism genes upregulated compared to non-VAT Tregs.
effector T cells use glucose differently in vivo, with a greater pro- Tregs in human studies have been shown to be both higher and lower
portion directed to biosynthetic pathways derived from glycolysis.62 in obesity, but the nature of these comparative studies is difficult to
Stable isotope metabolomics and other in vivo and tissue-based as- interpret without controlled interventions.
says are now increasingly accessible and allow for innovative direc- VAT Tregs are highly dependent on the IL-33-ST2 axis, which is
tions in our approaches to understand immunometabolism. typically associated with allergic Th2 response, but is increasingly
One tissue that has been a focus of immunological research is appreciated for potent wound healing capacity, promotion of tumor
adipose tissue. A wide range of changes in the adipose tissue—resi- immunosuppression, and activation of increased metabolism of white
dent immune cell populations occur with obesity, which can vary by and brown fat, potentially through VmSC-dependent IL-33 produc-
depot. Macrophages, in particular, change in content and phenotype tion.72,73 They describe a fibroblast-like population of mSC with
from lean to obese to weight-cycled states and influence the devel- several phenotypic subsets, three of which produce IL-33 (ie, immu-
opment of insulin resistance, diabetes, cardiovascular disease, and nocyte promoting) compared to IL-33-negative adipogenic cells with
cancer. Since back-to-back reports of adipose tissue infiltration by markers of pre-adipocytes. High-fat diet feeding transiently reduces
63-65
macrophages in 2003, the field has shown the importance of IL-33-producing VmSC cells followed by a reduction in VAT Tregs.
essentially every immune cell in adipose tissue with unique depot Interestingly, sex hormones appear to regulate subsets of VmSC pop-
specific differences. Caslin et al66 review the contribution of the pre- ulations and production of IL-33, which could account for differences
dominant immune cell in fat tissue and adipose tissue macrophages in Tregs in basal or obese state between males and females. Like
|
10       MAKOWSKI et al.

other immune cells in diet-induced obese models, short-term high- because they shape the immune response. Like tumors, influenza
fat diet—induced obesity versus long-term and severe obesity affect infection may induce immune suppression and infected epithelial
this axis differently. While short-term high-fat diet VmSC-IL33 + cells cells become glucose- and glutamine-dependent. Therefore, a
are associated with Tregs, this positive association is lost in severe delicate balance is needed to fuel the immune response without
obesity. Morbidly obese patient adipose tissue contains VmSCs (ie, exacerbating damage to the host.
are CD9hi) that associate with diabetes, fibrosis, and inflammation. An ultimate goal of immunometabolism is to exploit the specific
On the other hand, the CD9lo population that is enriched for PPARγ metabolic programs of distinct immune cell populations to treat dis-
and is reduced with diabetes. However, the role of VmSCs in IL-33- ease. Systemic lupus erythematosus (SLE) has been studied exten-
dependent Treg homeostasis in obesity is unclear. Controlled time sively in this regard and is reviewed by Teng et al77. The metabolism
course single cell sequencing studies in the future are necessary to of T cells in SLE has been recognized to be altered for nearly twenty
inform on the specifics of cell kinetics that are lacking in single end- years.78 The wide range of T cell subsets, B cells, and DCs that contrib-
point obesity studies. More research is necessary to understand the ute to SLE have been shown to have differential metabolic require-
bi-directional crosstalk between stromal cells and Tregs, commonal- ments. These are impacted by altered systemic nutrition, diet, and cell
ities between human and mouse, and the kinetics and phenotype of intrinsic changes to metabolism. Obesity and metabolic syndrome are
these cells in obesity and other diseases. Tregs also play key roles to associated with SLE and may contribute to a variety of SLE-associated
maintain homeostasis in other peripheral tissues including liver, skin, pathologies.79 This may occur due to increased nutrient acquisition by
and tumors and are reviewed by Wang et al74. inflammatory cells in settings of hyperglycemia and hyperlipidemia,
but may also be regulated through changes in hormones or adipokines,
such as leptin. A variety of metabolic inhibitors have been tested in
4 |  I M M U N O M E TA B O LI S M I N D I S E A S E animal models of SLE.80-82 Inhibitors of mTORC1 have shown some
A N D TR A N S L ATI O N promise, and direct inhibition of glycolysis may also protect from
disease. This effect was strengthened, however, by addition of the
Like activation and differentiation of lymphocytes and myeloid AMPK activator, metformin. A concern in these metabolic inhibitor
cells that lead to metabolic reprogramming, viral infections have treatments has been the potential for widespread toxicity. This con-
been shown to reshape cell metabolism in ways that are central cern, however, has not materialized, and treatments have been found
to the antiviral immune response. Viral infection can alter choles- to be remarkably well tolerated. Even blockade of the fundamental
terol synthesis,75 and Bahadoran et al focus on influenza-induced process of glucose uptake was not found to be widely cytotoxic and
metabolomic reprogramming of immune cells in the lung airway.76 provided protection in animal models of SLE.81 Combinations of met-
The lung microenvironment comprises barrier cells and mecha- abolic inhibitors for glycolysis and alternate metabolic pathways now
nisms as well as immune regulatory mechanisms that maintain offer new directions with successful preclinical proof of principle
homeostasis and defend the body against pathogens. Influenza studies, and it is now important to move to human clinical trials to
infection induces metabolic reprogramming of both immune and combat this auto-immune disease.
epithelial cell populations to co-opt the host's cells. In epithelial Tumor microenvironments can also alter the metabolism of T cells
cells, one example of influenza-induced metabolic reprogramming and macrophages to impair anti-tumor immunity.83 Critically, treat-
is by the induction of an aberrant activation of the PI3K/mTOR/ ments such as checkpoint blockade therapies act in part through reliev-
Akt pathway. Additionally, influenza induces immune cell activa- ing this metabolic inhibition.15-16,84 Siska et al focus on the “Warburg
tion and differentiation, although the impact of influenza A viral effect” which is a key feature of tumor cells and leads to acidification
infection on immune cells is largely uncharacterized. The authors and the accumulation of lactate in the tumor microenvironment.85 As
discussed metabolism in both steady state and upon influenza a consequence of localized lactic acidosis, immune cell populations
viral infection relevant to immune cell populations such as DCs, within the tumor microenvironment exert an immunosuppressive
macrophages, natural killer (NK) cells, and T cells. For example, effect and mediate an effective immune escape by tumor cells. The
influenza viral infection causes DCs to enter a state of hypergly- authors suggested a metabolic-tumor-stroma score (MeTS) as a pre-
colysis with relatively low oxygen utilization. Macrophages in the dictor of the efficacy of anti-tumor immune responses. MeTS are bro-
respiratory tract largely play a role in phagocytosing the virus and ken down into four sub-categories: MeTS 1 which consists of respiring
infected cells. Yet, M1 macrophages activate a strong antiviral tumor cells (OXPHOS) with high T cell infiltration (“Hot” tumors);
response including macrophage-derived reactive oxygen species MeTS 2 with a respiring tumor and glycolytic (Warburg-like) stromal
(ROS) that is deleterious to the host, thus representing potential cells; MeTS 3 with a glycolytic tumor and respiring stromal cells; and
targets to reduce disease severity. Influenza infection of T cells MeTS 4 with a glycolytic tumor and low T cell infiltration (“Cold” tu-
induces a switch from OXPHOS to T effector utilization of glucose mors). First, the authors provided a historical timeline of the Warburg
and glutamine. In sum, effector functions of immune cells such as effect, and then discussed the Warburg effect in immune cells and
DCs, NK cells, macrophages, and effector T cells rely upon glycoly- its underlying mechanisms in thorough detail. For example, HIF acti-
sis, while T regulatory cells use OXPHOS. Also, systemic glucose or vation is not solely dependent on hypoxic conditions, but can also be
other microenvironmental foci of influenza infection are discussed achieved under normoxia conditions and the presence of lactate or
MAKOWSKI et al. |
      11

pyruvate. The authors also discussed how tumor-derived lactate has cells. Current literature suggests that bile acids or agonism of bile
a broad spectrum of effects on tumor growth and metastasis such as acid receptors such as FXR or TGR5 in innate and adaptive immune
the promotion of cell motility, modulation of tumor-associated fibro- cells leads to a general downregulation of inflammation in most cell
blasts, impairment of anti-tumor immune cells (NK cells, CD8 T cells, types. However, microbial modification of bile acids and signaling
M1 macrophages, and DCs), and promotion of immunosuppressive through epithelial cells led to activation of bile acid targeted re-
immune cells (MDSC, M2 macrophages, Tregs). Finally, the authors cruitment of NKT cells and improved anti-tumor immunity.90 Thus,
focused on strategies targeting the Warburg effect for tumor therapy BA composition may influence diseases such as cancer through re-
and immune activation. For example, preclinical and clinical strategies programming of the immune microenvironment. Interestingly, mi-
aimed at targeting LDH-A, LDH-B, MCT1, and MCT4 blocked lactate crobial dysbiosis and specific microbes including bacteria and fungi
secretion which improved anti-tumor immune responses. correlate with several cancers, notably tumor subtypes, stage, or
Both systemic and local influences can also affect metabolism race in breast cancer.91 Furthermore, certain microbes have been
and anti-tumor immunity. shown to impact response to immune checkpoint blockade in an-
Turbitt et al focus on the effect of obesity on T cell metabolism ti-tumor therapy in mice and correlate with therapeutic response in
86
and function in the presence and absence of solid tumors. Obesity humans.92-94 Based on these and other related studies, it is possible
alters not just Treg, but also CD4 and CD8 T cells that play roles in that immune perturbations resulting from microbiome composition
anti-tumor immunity.87,88 Adipokines, such as leptin, are elevated in and microbially modified metabolites can be targeted for thera-
obese individuals and can promote T cell glycolysis and polarization peutic benefit. There is increasing interest in “bugs as drugs.” The
89
toward an anti-tumor Th1 phenotype. Thus, the authors provide discovery of novel pathways driven by microbial metabolism that
an explanation of why immune checkpoint blockade provides su- could be exploited in the treatment of cancer including the clinical
perior anti-tumor immune responses in both obese preclinical and efficacy of immune checkpoint blockade, obesity, and metabolic
clinical models compared to lean models. Leptin reverses metabolic syndrome, as well as auto-immune diseases, would be impactful to
defects in T cells and increases the expression of PD-1 on their sur- understand risk and impact of personalized therapy and improve
face. Therefore, immunotherapy such as anti-PD-1 is more efficient therapeutic outcomes.
in high-PD-1-expressing T cells that are present in obese individuals.
Moreover, the authors discuss the effects of glycolytic tumors on
the impairment of T cell functions. Highly glycolytic tumor cells in- 5 | CO N C LU S I O N A N D PE R S PEC TI V E
duce diminished glucose uptake by T cells and promote immunosup-
pressive myeloid cells such as MDSCs, therefore rendering T cells The field of immunometabolism has emerged as a major and at-
less effective. Additionally, the authors discussed obesity-driven tractive area of research in immunology and metabolic disease. The
intracellular lipid accumulation in T cells and DCs as a mechanism by focus on systemic and cellular metabolism and the diversity of meta-
which tumors inhibit anti-tumor immune cell functions. Finally, the bolic programs of specific cell populations have enhanced our un-
authors highlight novel therapeutic approaches focused on improv- derstanding of both basic biology and metabolic dysfunction, as well
ing T cell metabolic fitness. Few examples include mTOR inhibition as identified novel targets for anti-inflammatory and anti-tumor im-
and the use of anti-diabetic drugs such as metformin in combination mune-based therapies. This issue aims to highlight key areas of cur-
with current immunotherapeutic approaches. These strategies aim rent research, but many important questions and challenges remain
at improving T cell OXPHOS and cytokine production to increase the especially with regard to commonalities or differences between hu-
efficacy of anti-PD-1 blockade. mans and preclinical models, sex, or race. It is clear that the tissue lo-
Sipe and Chaib et al tackle the role of host-microbial interac- cation is critical to immune cell intrinsic metabolism through changes
tions and the impact of microbially derived metabolites on immu- such as cell-cell crosstalk, viral foci, competition for nutrients, and
nometabolism in anti-tumor immunity and other chronic diseases. extent of hypoxia. Future studies must incorporate the effect of
Microbes are altered by many interventions and lifestyle factors, the microenvironment on immune cell metabolism, and vice versa.
and the metabolic sequelae are just beginning to be uncovered. Finding new approaches to understand the diversity of metabolic
While most work on the microbiome has focused on the gut, ex- programs in specific tissue sites will open new avenues for discovery
tra-intestinal microbes are detected in various tissues, including and immune modulation. Ultimately, identifying therapeutic targets
tumors, and are susceptible to modification by dietary intake or is a key goal for the field. While standard-of-care treatments, such
changes in body composition such as weight gain or loss. How gut or as methotrexate, demonstrate the potential of targeting metabolic
extra-intestinal microbes alter their own and the host's metabolism pathways, better understanding of cell and tissue biology will cer-
is actively being researched because the magnitude of our micro- tainly reveal new targets with increased specificity and reduced tox-
biome's non-mammalian genetic capacity to generate or modulate icity. As a rapidly maturing field, immunometabolism is now poised
metabolites is vast. The impact of host bile acids and modification to move forward to address these needs.
of bile acids by microbes is discussed in detail for cell types includ-
ing tumor-associated macrophages, DCs, MDSC, T cells including ORCID
Tregs, Th17, mucosal-associated invariant T (MAIT), NKT, and B Liza Makowski  https://orcid.org/0000-0002-5337-8037
|
12       MAKOWSKI et al.

REFERENCES 24. Angelin A, Gil-de-Gómez L, Dahiya S, et al. Foxp3 reprograms T cell


metabolism to function in low-glucose, high-lactate environments.
1. Ibrahim MK, Zambruni M, Melby CL, Melby PC. Impact of child-
Cell Metab. 2017;25(6):1282-1293.e7.
hood malnutrition on host defense and infection. Clin Microbiol Rev.
25. Beier UH, Angelin A, Akimova T, et al. Essential role of mitochon-
2017;30(4):919-971.
drial energy metabolism in Foxp3(+) T-regulatory cell function and
2. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression
allograft survival. FASEB J. 2015;29(6):2315-2326.
of tumor necrosis factor-alpha: direct role in obesity-linked insulin
26. Gerriets VA, Kishton RJ, Johnson MO, et al. Foxp3 and Toll-like re-
resistance. Science. 1993;259(5091):87-91.
ceptor signaling balance Treg cell anabolic metabolism for suppres-
3. Hotamisligil GS. Foundations of immunometabolism and implications
sion. Nat Immunol. 2016;17(12):1459-1466.
for metabolic health and disease. Immunity. 2017;47(3):406-420.
27. Layman AAK, Deng G, O’Leary CE, et al. Ndfip1 restricts mTORC1
4. Warburg O. On the origin of cancer cells. Science.
signalling and glycolysis in regulatory T cells to prevent autoinflam-
1956;123(3191):309-314.
matory disease. Nat Commun. 2017;8:15677.
5. Kim J, DeBerardinis RJ. Mechanisms and implications of metabolic
28. Johnson MO, Wolf MM, Madden MZ, et al. Distinct regulation of
heterogeneity in cancer. Cell Metab. 2019;30(3):434-446.
Th17 and Th1 cell differentiation by glutaminase-dependent me-
6. Frauwirth KA, Riley JL, Harris MH, et al. The CD28 signaling path-
tabolism. Cell. 2018;175(7):1780-1795.e19.
way regulates glucose metabolism. Immunity. 2002;16(6):769-777.
29. Leone RD, Zhao L, Englert JM, et al. Glutamine blockade induces
7. Rathmell JC, Vander Heiden MG, Harris MH, Frauwirth KA,
divergent metabolic programs to overcome tumor immune evasion.
Thompson CB. In the absence of extrinsic signals, nutrient utiliza-
Science. 2019;366(6468):1013-1021.
tion by lymphocytes is insufficient to maintain either cell size or
3 0. Liu PS, Wang H, Li X, et al. alpha-ketoglutarate orchestrates macro-
viability. Mol Cell. 2000;6(3):683-692.
phage activation through metabolic and epigenetic reprogramming.
8. Wang R, Dillon CP, Shi LZ, et al. The transcription factor Myc con-
Nat Immunol. 2017;18(9):985-994.
trols metabolic reprogramming upon T lymphocyte activation.
31. Cho SH, Raybuck AL, Blagih J, et al. Hypoxia-inducible factors
Immunity. 2011;35(6):871-882.
in CD4(+) T cells promote metabolism, switch cytokine secre-
9. Vats D, Mukundan L, Odegaard JI, et al. Oxidative metabolism and
tion, and T cell help in humoral immunity. Proc Natl Acad Sci USA.
PGC-1beta attenuate macrophage-mediated inflammation. Cell
2019;116(18):8975-8984.
Metab. 2006;4(1):13-24.
32. Shi LZ, Wang R, Huang G, et al. HIF1alpha-dependent glycolytic
10. Michalek RD, Gerriets VA, Jacobs SR, et al. Cutting edge: dis-
pathway orchestrates a metabolic checkpoint for the differentia-
tinct glycolytic and lipid oxidative metabolic programs are essen-
tion of TH17 and Treg cells. J Exp Med. 2011;208(7):1367-1376.
tial for effector and regulatory CD4+ T cell subsets. J Immunol.
33. Liu L, Lu Y, Martinez J, et al. Proinflammatory signal suppresses
2011;186(6):3299-3303.
proliferation and shifts macrophage metabolism from Myc-
11. Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol
dependent to HIF1alpha-dependent. Proc Natl Acad Sci USA.
Res. 2017;5(1):3-8.
2016;113(6):1564-1569.
12. Krawczyk CM, Holowka T, Sun J, et al. Toll-like receptor-induced
3 4. Huang H, Long L, Zhou P, Chapman NM, Chi H. mTORsignaling at
changes in glycolytic metabolism regulate dendritic cell activation.
the crossroads of environmental signals and T-cell fate decisions.
Blood. 2010;115(23):4742-4749.
Immunol Rev. 2020;295:15–38.
13. Klein Geltink RI, O’Sullivan D, Corrado M, et al. Mitochondrial prim-
35. Shen K, Sabatini DM. Ragulator and SLC38A9 activate the Rag
ing by CD28. Cell. 2017;171(2):385-397.e11.
GTPases through noncanonical GEF mechanisms. Proc Natl Acad Sci
14. Buck MD, O’Sullivan D, Klein Geltink RI, et al. Mitochondrial dy-
USA. 2018;115(38):9545-9550.
namics controls T cell fate through metabolic programming. Cell.
36. Wyant GA, Abu-Remaileh M, Wolfson RL, et al. mTORC1 Activator
2016;166(1):63-76.
SLC38A9 is required to efflux essential amino acids from lyso-
15. Parry RV, Chemnitz JM, Frauwirth KA, et al. CTLA-4 and PD-1 re-
somes and use protein as a nutrient. Cell. 2017;171(3):642-654.
ceptors inhibit T-cell activation by distinct mechanisms. Mol Cell
e12.
Biol. 2005;25(21):9543-9553.
37. MacIver NJ, Blagih J, Saucillo DC, et al. The liver kinase B1 is a cen-
16. Patsoukis N, Bardhan K, Chatterjee P, et al. PD-1 alters T-cell meta-
tral regulator of T cell development, activation, and metabolism. J
bolic reprogramming by inhibiting glycolysis and promoting lipolysis
Immunol. 2011;187(8):4187-4198.
and fatty acid oxidation. Nat Commun. 2015;6:6692.
38. Blagih J, Coulombe F, Vincent EE, et al. The energy sensor AMPK
17. Zheng Y, Delgoffe GM, Meyer CF, Chan W, Powell JD. Anergic T
regulates T cell metabolic adaptation and effector responses in
cells are metabolically anergic. J Immunol. 2009;183(10):6095-6101.
vivo. Immunity. 2015;42(1):41-54.
18. Araki K, Turner AP, Shaffer VO, et al. mTOR regulates memory CD8
39. Zeng H, Yang K, Cloer C, Neale G, Vogel P, Chi H. mTORC1 couples
T-cell differentiation. Nature. 2009;460(7251):108-112.
immune signals and metabolic programming to establish T(reg)-cell
19. Pearce EL, Walsh MC, Cejas PJ, et al. Enhancing CD8 T-cell
function. Nature. 2013;499(7459):485-490.
memory by modulating fatty acid metabolism. Nature.
4 0. Shi H, Chapman NM, Wen J, et al. Amino acids license kinase
2009;460(7251):103-107.
mTORC1 activity and treg cell function via small G proteins Rag and
20. Bantug GR, Fischer M, Grählert J, et al. Mitochondria-endoplasmic
Rheb. Immunity. 2019;51(6):pp. 1012–1027 e7.
reticulum contact sites function as immunometabolic hubs that
41. Sun IH, Oh MH, Zhao L, et al. mTOR complex 1 signaling regulates
orchestrate the rapid recall response of memory CD8(+) T cells.
the generation and function of central and effector Foxp3(+) regu-
Immunity. 2018;48(3):542-555.e6.
latory T cells. J Immunol. 2018;201(2):481-492.
21. Macintyre AN, Gerriets VA, Nichols AG, et al. The glucose trans-
42. Dufort FJ, Bleiman BF, Gumina MR, et al. Cutting edge: IL-4-
porter Glut1 is selectively essential for CD4 T cell activation and
mediated protection of primary B lymphocytes from apoptosis via
effector function. Cell Metab. 2014;20(1):61-72.
Stat6-dependent regulation of glycolytic metabolism. J Immunol.
22. Nakaya M, Xiao Y, Zhou X, et al. Inflammatory T cell responses rely
2007;179(8):4953-4957.
on amino acid transporter ASCT2 facilitation of glutamine uptake
43. Caro-Maldonado A, Wang R, Nichols AG, et al. Metabolic repro-
and mTORC1 kinase activation. Immunity. 2014;40(5):692-705.
gramming is required for antibody production that is suppressed
23. Gerriets VA, Kishton RJ, Nichols AG, et al. Metabolic programming
in anergic but exaggerated in chronically BAFF-exposed B cells. J
and PDHK1 control CD4+ T cell subsets and inflammation. J Clin
Immunol. 2014;192(8):3626-3636.
Invest. 2015;125(1):194-207.
MAKOWSKI et al. |
      13

4 4. Xiao G, Chan LN, Klemm L, et al. B-cell-specific diversion of glucose 66. Caslin HL, Bhanot M, Bolus WR, Hasty A. Adipose tissue macro-
carbon utilization reveals a unique vulnerability in B cell malignan- phages: Unique polarization and bioenergetics in obesity. Immunol
cies. Cell. 2018;173(2):470-484.e18. Rev. 2020;295:101–113.
45. Akkaya M, Traba J, Roesler AS, et al. Second signals rescue B cells 67. Lumeng CN, DeYoung SM, Bodzin JL, Saltiel AR. Increased inflam-
from activation-induced mitochondrial dysfunction and death. Nat matory properties of adipose tissue macrophages recruited during
Immunol. 2018;19(8):871-884. diet-induced obesity. Diabetes. 2007;56(1):16-23.
46. Weisel FJ, Mullett SJ, Elsner RA, et al. Germinal center B cells se- 68. Flaherty SE, Grijalva A, Xu X, Ables E, Nomani A, Ferrante AW. A li-
lectively oxidize fatty acids for energy while conducting minimal pase-independent pathway of lipid release and immune modulation
glycolysis. Nat Immunol. 2020;21(3):331-342. by adipocytes. Science. 2019;363(6430):989-993.
47. Jellusova J. The role of metabolic checkpoint regulators in B cell 69. Cipolletta D, Feuerer M, Li A, et al. PPAR-gamma is a major driver
survival and transformation. Immunol Rev. 2020;295:39–53. of the accumulation and phenotype of adipose tissue Treg cells.
48. van Teijlingen BN, Pearce EJ. Cell-intrinsic metabolic regulation of Nature. 2012;486(7404):549-553.
mononuclear phagocyte activation: Findings from the tip of the ice- 70. Wu D, Han JM, Yu X, et al. Characterization of regulatory T
berg. Immunol Rev. 2020;295:54–67. cells in obese omental adipose tissue in humans. Eur J Immunol.
49. Tannahill GM, Curtis AM, Adamik J, et al. Succinate is an inflam- 2019;49(2):336-347.
matory signal that induces IL-1beta through HIF-1alpha. Nature. 71. Li C, Spallanzani RG, Mathis D. Visceral adipose tissue Tregs and the
2013;496(7444):238-242. cells that nurture them. Immunol Rev. 2020;295:114–125.
50. Mills EL, Kelly B, Logan A, et al. Succinate dehydrogenase supports 72. Vasanthakumar A, Moro K, Xin A, et al. The transcriptional regu-
metabolic repurposing of mitochondria to drive inflammatory mac- lators IRF4, BATF and IL-33 orchestrate development and mainte-
rophages. Cell. 2016;167(2):457-470.e13. nance of adipose tissue-resident regulatory T cells. Nat Immunol.
51. Lampropoulou V, Sergushichev A, Bambouskova M, et al. Itaconate 2015;16(3):276-285.
links inhibition of succinate dehydrogenase with macrophage 73. Li C, DiSpirito JR, Zemmour D, et al. TCR transgenic mice reveal
metabolic remodeling and regulation of inflammation. Cell Metab. stepwise, multi-site acquisition of the distinctive Fat-Treg pheno-
2016;24(1):158-166. type. Cell. 2018;174(2):285-299.e12.
52. Mills EL, Ryan DG, Prag HA, et al. Itaconate is an anti-inflamma- 74. Wang H, Lu C-H, Ho P-C. Metabolic adaptation orchestrates tis-
tory metabolite that activates Nrf2 via alkylation of KEAP1. Nature. sue context-dependent behavior in regulatory T cells. Immunol Rev.
2018;556(7699):113-117. 2020;295:126–139.
53. Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, et al. Macrophage- 75. York AG, Williams KJ, Argus JP, et al. Limiting cholesterol bio-
specific PPARgamma controls alternative activation and improves synthetic flux spontaneously engages type I IFN signaling. Cell.
insulin resistance. Nature. 2007;447(7148):1116-1120. 2015;163(7):1716-1729.
54. Freemerman AJ, Johnson AR, Sacks GN, et al. Metabolic reprogram- 76. Bahadoran A, Bezavada L, Smallwood HS. Fueling influenza and
ming of macrophages: glucose transporter 1 (GLUT1)-mediated glu- the immune response: Implications for metabolic reprogramming
cose metabolism drives a proinflammatory phenotype. J Biol Chem. during influenza infection and immunometabolism. Immunol Rev.
2014;289(11):7884-7896. 2020;295:140–166.
55. Freemerman AJ, Zhao L, Pingili AK, et al. Myeloid Slc2a1-deficient 77. Teng X, Brown J, Choi S-C, Li W, Morel L. Metabolic determinants
murine model revealed macrophage activation and metabolic phe- of lupus pathogenesis. Immunol Rev. 2020;295:167–186.
notype are fueled by GLUT1. J Immunol. 2019;202(4):1265-1286. 78. Gergely P, Grossman C, Niland B, et al. Mitochondrial hyperpolar-
56. Puleston DJ, Buck MD, Klein Geltink RI, et al. Polyamines and eIF5A ization and ATP depletion in patients with systemic lupus erythe-
hypusination modulate mitochondrial respiration and macrophage matosus. Arthritis Rheum. 2002;46(1):175-190.
activation. Cell Metab. 2019;30(2):352-363.e8. 79. Giannelou M, Mavragani CP. Cardiovascular disease in systemic lupus
57. Kolev M, Dimeloe S, Le Friec G, et al. Complement regulates nutri- erythematosus: a comprehensive update. J Autoimmun. 2017;82:1-12.
ent influx and metabolic reprogramming during Th1 cell responses. 8 0. Abboud G, Choi S-C, Kanda N, Zeumer-Spataro L, Roopenian DC,
Immunity. 2015;42(6):1033-1047. Morel L. Inhibition of glycolysis reduces disease severity in an
58. West EE, Kunz N, Kemper C. Complement and human T cell metab- autoimmune model of rheumatoid arthritis. Front Immunol.
olism: Location, location, location. Immunol Rev. 2020;295:68–81. 2018;9:1973.
59. McGuire PJ. Chemical individuality in T cells: a Garrodian view of 81. Li W, Qu G, Choi S-C, et al. Targeting T cell activation and lupus
immunometabolism. Immunol Rev. 2020;295:82–100. autoimmune phenotypes by inhibiting glucose transporters. Front
60. Burgener A-V, Bantug GR, Meyer BJ, et al. SDHA gain-of-func- Immunol. 2019;10:833.
tion engages inflammatory mitochondrial retrograde signaling via 82. Yin Y, Choi SC, Xu Z, et al. Normalization of CD4+ T cell metabolism
KEAP1-Nrf2. Nat Immunol. 2019;20(10):1311-1321. reverses lupus. Sci Transl Med. 2015;7(274):274ra18.
61. Tarasenko TN, Pacheco SE, Koenig MK, et al. Cytochrome c oxi- 83. Siska PJ, Beckermann KE, Mason FM, et al. Mitochondrial dysregula-
dase activity is a metabolic checkpoint that regulates cell fate tion and glycolytic insufficiency functionally impair CD8 T cells infil-
decisions during T cell activation and differentiation. Cell Metab. trating human renal cell carcinoma. JCI Insight. 2017;2(12):93411.
2017;25(6):1254-1268.e7. 8 4. Ogando J, Sáez ME, Santos J, et al. PD-1 signaling affects cristae
62. Ma EH, Verway MJ, Johnson RM, et al. Metabolic profiling using stable morphology and leads to mitochondrial dysfunction in human
isotope tracing reveals distinct patterns of glucose utilization by phys- CD8(+) T lymphocytes. J Immunother Cancer. 2019;7(1):151.
iologically activated CD8(+) T cells. Immunity. 2019;51(5):856-870.e5. 85. Siska PJ, Singer K, Evert K, Renner K, Kreutz M. The immunological
63. Wellen KE, Hotamisligil GS. Obesity-induced inflammatory changes Warburg effect: Can a metabolic-tumor-stroma score (MeTS) guide
in adipose tissue. J Clin Invest. 2003;112(12):1785-1788. cancer immunotherapy? Immunol Rev. 2020;295:187–202.
6 4. Xu H, Barnes GT, Yang Q, et al. Chronic inflammation in fat plays a 86. Turbitt WJ, Rosean CB, Weber KS, Norian LA. Obesity and CD8 T
crucial role in the development of obesity-related insulin resistance. cell metabolism: Implications for anti-tumor immunity and cancer
J Clin Invest. 2003;112(12):1821-1830. immunotherapy outcomes. Immunol Rev. 2020;295:203–219.
65. Weisberg SP, McCann D, Desai M, et al. Obesity is associated 87. Nishimura S, Manabe I, Nagasaki M, et al. CD8+ effector T cells
with macrophage accumulation in adipose tissue. J Clin Invest. contribute to macrophage recruitment and adipose tissue inflam-
2003;112(12):1796-1808. mation in obesity. Nat Med. 2009;15(8):914-920.
|
14       MAKOWSKI et al.

88. Michelet X, Dyck L, Hogan A, et al. Metabolic reprogramming 93. Matson V, Fessler J, Bao R, et al. The commensal microbiome is as-
of natural killer cells in obesity limits antitumor responses. Nat sociated with anti-PD-1 efficacy in metastatic melanoma patients.
Immunol. 2018;19(12):1330-1340. Science. 2018;359(6371):104-108.
89. Saucillo DC, Gerriets VA, Sheng J, Rathmell JC, MacIver NJ. Leptin 94. Goedert JJ, Jones G, Hua X, et al. Investigation of the association
metabolically licenses T cells for activation to link nutrition and im- between the fecal microbiota and breast cancer in postmenopausal
munity. J Immunol. 2014;192(1):136-144. women: a population-based case-control pilot study. J Natl Cancer
90. Ma C, Han M, Heinrich B, et al. Gut microbiome-mediated bile Inst. 2015;107(8):djv147.
acid metabolism regulates liver cancer via NKT cells. Science.
2018;360(6391):eaan5931.
91. Smith A, Pierre JF, Makowski L, et al. Distinct microbial commu-
How to cite this article: Makowski L, Chaib M, Rathmell JC.
nities that differ by race, stage, or breast-tumor subtype in breast
Immunometabolism: From basic mechanisms to translation.
tissues of non-Hispanic Black and non-Hispanic White women. Sci
Rep. 2019;9(1):11940. Immunol Rev. 2020;295:5–14. https://doi.org/10.1111/
92. Santoni M, Piva F, Conti A, et al. Gut microbiome influences efficacy imr.12858
of PD-1-based immunotherapy against epithelial tumors. Science.
2018;359(6371):91-97.

You might also like