Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
J Clin Invest ; 132(2)2022 01 18.
Article in English | MEDLINE | ID: mdl-35040439

ABSTRACT

Macrophages exposed to inflammatory stimuli including LPS undergo metabolic reprogramming to facilitate macrophage effector function. This metabolic reprogramming supports phagocytic function, cytokine release, and ROS production that are critical to protective inflammatory responses. The Krebs cycle is a central metabolic pathway within all mammalian cell types. In activated macrophages, distinct breaks in the Krebs cycle regulate macrophage effector function through the accumulation of several metabolites that were recently shown to have signaling roles in immunity. One metabolite that accumulates in macrophages because of the disturbance in the Krebs cycle is itaconate, which is derived from cis-aconitate by the enzyme cis-aconitate decarboxylase (ACOD1), encoded by immunoresponsive gene 1 (Irg1). This Review focuses on itaconate's emergence as a key immunometabolite with diverse roles in immunity and inflammation. These roles include inhibition of succinate dehydrogenase (which controls levels of succinate, a metabolite with multiple roles in inflammation), inhibition of glycolysis at multiple levels (which will limit inflammation), activation of the antiinflammatory transcription factors Nrf2 and ATF3, and inhibition of the NLRP3 inflammasome. Itaconate and its derivatives have antiinflammatory effects in preclinical models of sepsis, viral infections, psoriasis, gout, ischemia/reperfusion injury, and pulmonary fibrosis, pointing to possible itaconate-based therapeutics for a range of inflammatory diseases. This intriguing metabolite continues to yield fascinating insights into the role of metabolic reprogramming in host defense and inflammation.


Subject(s)
Citric Acid Cycle/immunology , Macrophage Activation , Macrophages/immunology , Succinates/immunology , Animals , Humans , Inflammation/immunology
2.
Front Immunol ; 12: 739591, 2021.
Article in English | MEDLINE | ID: mdl-34950133

ABSTRACT

Vaccines are safe and efficient in controlling bacterial diseases in the aquaculture industry and are in line with green farming. The present study develops a previously unreported approach to prepare a live-attenuated V. alginolyticus vaccine by culturing bacteria in a high concentration of magnesium to attenuate bacterial virulence. Furthermore, metabolomes of zebrafish immunized with the live-attenuated vaccines were compared with those of survival and dying zebrafish infected by V. alginolyticus. The enhanced TCA cycle and increased fumarate were identified as the most key metabolic pathways and the crucial biomarker of vaccine-mediated and survival fish, respectively. Exogenous fumarate promoted expression of il1ß, il8, il21, nf-κb, and lysozyme in a dose-dependent manner. Among the five innate immune genes, the elevated il1ß, il8, and lysozyme are overlapped in the vaccine-immunized zebrafish and the survival from the infection. These findings highlight a way in development of vaccines and exploration of the underlying mechanisms.


Subject(s)
Bacterial Vaccines/immunology , Citric Acid Cycle/immunology , Magnesium/immunology , Vibrio alginolyticus/immunology , Animals , Vaccines, Attenuated/immunology , Zebrafish/immunology
3.
JCI Insight ; 6(22)2021 11 22.
Article in English | MEDLINE | ID: mdl-34609963

ABSTRACT

Mounting evidence points to alterations in mitochondrial metabolism in renal cell carcinoma (RCC). However, the mechanisms that regulate the TCA cycle in RCC remain uncharacterized. Here, we demonstrate that loss of TCA cycle enzyme expression is retained in RCC metastatic tissues. Moreover, proteomic analysis demonstrates that reduced TCA cycle enzyme expression is far more pronounced in RCC relative to other tumor types. Loss of TCA cycle enzyme expression is correlated with reduced expression of the transcription factor PGC-1α, which is also lost in RCC tissues. PGC-1α reexpression in RCC cells restores the expression of TCA cycle enzymes in vitro and in vivo and leads to enhanced glucose carbon incorporation into TCA cycle intermediates. Mechanistically, TGF-ß signaling, in concert with histone deacetylase 7 (HDAC7), suppresses TCA cycle enzyme expression. Our studies show that pharmacologic inhibition of TGF-ß restores the expression of TCA cycle enzymes and suppresses tumor growth in an orthotopic model of RCC. Taken together, this investigation reveals a potentially novel role for the TGF-ß/HDAC7 axis in global suppression of TCA cycle enzymes in RCC and provides insight into the molecular basis of altered mitochondrial metabolism in this malignancy.


Subject(s)
Citric Acid Cycle/immunology , Gene Expression Profiling/methods , Histone Deacetylases/metabolism , Kidney Neoplasms/immunology , Transforming Growth Factor beta/metabolism , Animals , Humans , Mice , Transfection
4.
PLoS One ; 16(7): e0253618, 2021.
Article in English | MEDLINE | ID: mdl-34214099

ABSTRACT

The proportion of Staphylococcus aureus in the skin microbiome is associated with the severity of inflammation in the skin disease atopic dermatitis. Staphylococcus epidermidis, a commensal skin bacterium, inhibits the growth of S. aureus in the skin. Therefore, the balance between S. epidermidis and S. aureus in the skin microbiome is important for maintaining healthy skin. In the present study, we demonstrated that the heat-treated culture supernatant of Delftia acidovorans, a member of the skin microbiome, inhibits the growth of S. epidermidis, but not that of S. aureus. Comprehensive gene expression analysis by RNA sequencing revealed that culture supernatant of D. acidovorans increased the expression of genes related to glycolysis and the tricarboxylic acid cycle (TCA) cycle in S. epidermidis. Malonate, an inhibitor of succinate dehydrogenase in the TCA cycle, suppressed the inhibitory effect of the heat-treated culture supernatant of D. acidovorans on the growth of S. epidermidis. Reactive oxygen species production in S. epidermidis was induced by the heat-treated culture supernatant of D. acidovorans and suppressed by malonate. Further, the inhibitory effect of the heat-treated culture supernatant of D. acidovorans on the growth of S. epidermidis was suppressed by N-acetyl-L-cysteine, a free radical scavenger. These findings suggest that heat-resistant substances secreted by D. acidovorans inhibit the growth of S. epidermidis by inducing the production of reactive oxygen species via the TCA cycle.


Subject(s)
Delftia acidovorans/immunology , Dermatitis, Atopic/immunology , Skin/microbiology , Staphylococcal Infections/immunology , Staphylococcus epidermidis/isolation & purification , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Citric Acid Cycle/immunology , Delftia acidovorans/genetics , Delftia acidovorans/metabolism , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/pathology , Gene Expression Regulation, Bacterial/immunology , Humans , Microbiota/immunology , RNA-Seq , Reactive Oxygen Species/metabolism , Skin/immunology , Skin/pathology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/immunology , Staphylococcus aureus/isolation & purification , Staphylococcus epidermidis/immunology
5.
Signal Transduct Target Ther ; 6(1): 2, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33384409

ABSTRACT

NAD+ was discovered during yeast fermentation, and since its discovery, its important roles in redox metabolism, aging, and longevity, the immune system and DNA repair have been highlighted. A deregulation of the NAD+ levels has been associated with metabolic diseases and aging-related diseases, including neurodegeneration, defective immune responses, and cancer. NAD+ acts as a cofactor through its interplay with NADH, playing an essential role in many enzymatic reactions of energy metabolism, such as glycolysis, oxidative phosphorylation, fatty acid oxidation, and the TCA cycle. NAD+ also plays a role in deacetylation by sirtuins and ADP ribosylation during DNA damage/repair by PARP proteins. Finally, different NAD hydrolase proteins also consume NAD+ while converting it into ADP-ribose or its cyclic counterpart. Some of these proteins, such as CD38, seem to be extensively involved in the immune response. Since NAD cannot be taken directly from food, NAD metabolism is essential, and NAMPT is the key enzyme recovering NAD from nicotinamide and generating most of the NAD cellular pools. Because of the complex network of pathways in which NAD+ is essential, the important role of NAD+ and its key generating enzyme, NAMPT, in cancer is understandable. In the present work, we review the role of NAD+ and NAMPT in the ways that they may influence cancer metabolism, the immune system, stemness, aging, and cancer. Finally, we review some ongoing research on therapeutic approaches.


Subject(s)
NAD/immunology , Neoplasms/immunology , Neoplastic Stem Cells/immunology , Citric Acid Cycle/immunology , Cytokines/immunology , DNA Damage/immunology , Humans , Neoplasm Proteins/immunology , Neoplasms/therapy , Nicotinamide Phosphoribosyltransferase/immunology
6.
FEBS J ; 288(12): 3694-3714, 2021 06.
Article in English | MEDLINE | ID: mdl-33460504

ABSTRACT

Macrophages represent the first line of defence in innate immune responses and additionally serve important functions for the regulation of host inflammation and tissue homeostasis. The M1/M2 model describes the two extremes of macrophage polarization states, which can be induced by multiple stimuli, most notably by LPS/IFN-γ and IL-4/IL-13. Historically, the expression of two genes encoding for enzymes, which use the same amino acid as their substrate, iNOS and ARG1, has been used to define classically activated M1 (iNOS) and alternatively activated M2 (ARG1) macrophages. This 'arginine dichotomy' has recently become a matter of debate; however, in parallel with the emerging field of immunometabolism there is accumulating evidence that these two enzymes and their related metabolites are fundamentally involved in the intrinsic regulation of macrophage polarization and function. The aim of this review is to highlight recent advances in macrophage biology and immunometabolism with a specific focus on amino acid metabolism and their related metabolic pathways: iNOS/ARG1 (arginine), TCA cycle and OXPHOS (glutamine) as well as the one-carbon metabolism (serine, glycine).


Subject(s)
Arginase/metabolism , Arginine/metabolism , Glutamine/metabolism , Glycine/immunology , Macrophages/metabolism , Nitric Oxide Synthase Type II/metabolism , Serine/metabolism , Arginase/genetics , Arginase/immunology , Arginine/immunology , Cell Differentiation/drug effects , Citric Acid Cycle/genetics , Citric Acid Cycle/immunology , Gene Expression Regulation , Glutamine/immunology , Glycine/metabolism , Humans , Immunity, Innate , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/classification , Macrophages/drug effects , Macrophages/immunology , Mitochondria/drug effects , Mitochondria/immunology , Mitochondria/metabolism , Nitric Oxide/immunology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Oxidative Phosphorylation , Serine/immunology
7.
Immunology ; 161(3): 165-174, 2020 11.
Article in English | MEDLINE | ID: mdl-32418209

ABSTRACT

Metabolites are the essential substrates for epigenetic modification enzymes to write or erase the epigenetic blueprint in cells. Hence, the availability of nutrients and activity of metabolic pathways strongly influence the enzymatic function. Recent studies have shed light on the choreography between metabolome and epigenome in the control of immune cell differentiation and function, with a major focus on histone modifications. Yet, despite its importance in gene regulation, DNA methylation and its relationship with metabolism is relatively unclear. In this review, we will describe how the metabolic flux can influence epigenetic networks in innate and adaptive immune cells, with a focus on the DNA methylation cycle and the metabolites S-adenosylmethionine and α-ketoglutarate. Future directions will be discussed for this rapidly emerging field.


Subject(s)
Citric Acid Cycle/immunology , Epigenome/immunology , Metabolome/immunology , Adaptive Immunity , Animals , DNA Methylation , Epigenesis, Genetic , Humans , Immunity, Innate , Ketoglutaric Acids/metabolism , S-Adenosylmethionine/metabolism
9.
FEBS J ; 287(16): 3350-3369, 2020 08.
Article in English | MEDLINE | ID: mdl-32255251

ABSTRACT

The inflammatory response involves the activation of several cell types to fight insults caused by a plethora of agents, and to maintain the tissue homoeostasis. On the one hand, cells involved in the pro-inflammatory response, such as inflammatory M1 macrophages, Th1 and Th17 lymphocytes or activated microglia, must rapidly provide energy to fuel inflammation, which is essentially accomplished by glycolysis and high lactate production. On the other hand, regulatory T cells or M2 macrophages, which are involved in immune regulation and resolution of inflammation, preferentially use fatty acid oxidation through the TCA cycle as a main source for energy production. Here, we discuss the impact of glycolytic metabolism at the different steps of the inflammatory response. Finally, we review a wide variety of molecular mechanisms which could explain the relationship between glycolytic metabolites and the pro-inflammatory phenotype, including signalling events, epigenetic remodelling, post-transcriptional regulation and post-translational modifications. Inflammatory processes are a common feature of many age-associated diseases, such as cardiovascular and neurodegenerative disorders. The finding that immunometabolism could be a master regulator of inflammation broadens the avenue for treating inflammation-related pathologies through the manipulation of the vascular and immune cell metabolism.


Subject(s)
Citric Acid Cycle/immunology , Glycolysis/immunology , Inflammation/immunology , Macrophage Activation/immunology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Inflammation/metabolism , Macrophages/classification , Macrophages/metabolism , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism
10.
Antioxid Redox Signal ; 32(12): 834-852, 2020 04 20.
Article in English | MEDLINE | ID: mdl-31847530

ABSTRACT

Significance: The tricarboxylic acid (TCA) cycle is a housekeeping metabolic pathway essential for generation of energy and biosynthetic intermediates. Alterations of the TCA cycle play a pivotal role in oncogenesis and inflammation. As such, some metabolic vulnerabilities, imposed by TCA cycle dysfunction in cancer, have been identified. Similarly, the TCA cycle appeared as an actionable pathway in immunopathologies. Recent Advances: Metabolic changes accompanying cell transformation have been usually considered as adaptive mechanisms to malignant transformation. The identification of oncogenic mutations in some TCA cycle enzymes changed this view, indicating altered mitochondrial metabolism as an instrumental mechanism for cancer initiation. Similarly, the observation that TCA cycle-derived metabolites have multiple signaling roles in immune cells supports the idea of this pathway as a metabolic rheostat of immune responses. Critical Issues: This review summarizes the crucial role of the TCA cycle in pathophysiology describing the post-translational and epigenetic impact of oncometabolites accumulation in cancer and immune cells. Future Directions: Additional studies will be necessary to further explore the role of oncometabolites in paracrine signaling and to identify genuine metabolic and nutritional liabilities imposed by TCA cycle dysfunction in cancer, hardly to be escaped by resistance mechanisms.


Subject(s)
Citric Acid Cycle , Neoplasms/immunology , Neoplasms/metabolism , Animals , Citric Acid Cycle/immunology , Humans , Neoplasms/pathology
11.
Front Immunol ; 10: 1462, 2019.
Article in English | MEDLINE | ID: mdl-31333642

ABSTRACT

Macrophages are a heterogeneous population of immune cells playing several and diverse functions in homeostatic and immune responses. The broad spectrum of macrophage functions depends on both heterogeneity and plasticity of these cells, which are highly specialized in sensing the microenvironment and modify their properties accordingly. Although it is clear that macrophage phenotypes are difficult to categorize and should be seen as plastic and adaptable, they can be simplified into two extremes: a pro-inflammatory (M1) and an anti-inflammatory/pro-resolving (M2) profile. Based on this definition, M1 macrophages are able to start and sustain inflammatory responses, secreting pro-inflammatory cytokines, activating endothelial cells, and inducing the recruitment of other immune cells into the inflamed tissue; on the other hand, M2 macrophages promote the resolution of inflammation, phagocytose apoptotic cells, drive collagen deposition, coordinate tissue integrity, and release anti-inflammatory mediators. Dramatic switches in cell metabolism accompany these phenotypic and functional changes of macrophages. In particular, M1 macrophages rely mainly on glycolysis and present two breaks on the TCA cycle that result in accumulation of itaconate (a microbicide compound) and succinate. Excess of succinate leads to Hypoxia Inducible Factor 1α (HIF1α) stabilization that, in turn, activates the transcription of glycolytic genes, thus sustaining the glycolytic metabolism of M1 macrophages. On the contrary, M2 cells are more dependent on oxidative phosphorylation (OXPHOS), their TCA cycle is intact and provides the substrates for the complexes of the electron transport chain (ETC). Moreover, pro- and anti-inflammatory macrophages are characterized by specific pathways that regulate the metabolism of lipids and amino acids and affect their responses. All these metabolic adaptations are functional to support macrophage activities as well as to sustain their polarization in specific contexts. The aim of this review is to discuss recent findings linking macrophage functions and metabolism.


Subject(s)
Citric Acid Cycle/immunology , Glycolysis/immunology , Macrophage Activation , Macrophages , Oxidative Phosphorylation , Humans , Inflammation/immunology , Inflammation/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Oxidation-Reduction
12.
Article in English | MEDLINE | ID: mdl-31355153

ABSTRACT

Detrimental effects of malnutrition on immune responses to pathogens have long been recognized and it is considered a main risk factor for various infectious diseases, including visceral leishmaniasis (VL). Thymus is a target of both malnutrition and infection, but its role in the immune response to Leishmania infantum in malnourished individuals is barely studied. Because we previously observed thymic atrophy and significant reduction in cellularity and chemokine levels in malnourished mice infected with L. infantum, we postulated that the thymic microenvironment is severely compromised in those animals. To test this, we analyzed the microarchitecture of the organ and measured the protein abundance in its interstitial space in malnourished BALB/c mice infected or not with L. infantum. Malnourished-infected animals exhibited a significant reduction of the thymic cortex:medulla ratio and altered abundance of proteins secreted in the thymic interstitial fluid. Eighty-one percent of identified proteins are secreted by exosomes and malnourished-infected mice showed significant decrease in exosomal proteins, suggesting that exosomal carrier system, and therefore intrathymic communication, is dysregulated in those animals. Malnourished-infected mice also exhibited a significant increase in the abundance of proteins involved in lipid metabolism and tricarboxylic acid cycle, suggestive of a non-proliferative microenvironment. Accordingly, flow cytometry analysis revealed decreased proliferation of single positive and double positive T cells in those animals. Together, the reduced cortical area, decreased proliferation, and altered protein abundance suggest a dysfunctional thymic microenvironment where T cell migration, proliferation, and maturation are compromised, contributing for the thymic atrophy observed in malnourished animals. All these alterations could affect the control of the local and systemic infection, resulting in an impaired response to L. infantum infection.


Subject(s)
Host-Pathogen Interactions/immunology , Leishmania infantum/immunology , Leishmaniasis, Visceral/immunology , Malnutrition/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Biological Transport , Cell Movement , Cell Proliferation , Citric Acid Cycle/genetics , Citric Acid Cycle/immunology , Exosomes/immunology , Exosomes/metabolism , Exosomes/parasitology , Extracellular Fluid/immunology , Extracellular Fluid/metabolism , Extracellular Fluid/parasitology , Galectin 1/genetics , Galectin 1/immunology , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Immunity, Innate , Leishmania infantum/growth & development , Leishmaniasis, Visceral/genetics , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Lipid Metabolism , Male , Malnutrition/genetics , Malnutrition/metabolism , Malnutrition/parasitology , Mice , Mice, Inbred BALB C , Plasminogen/genetics , Plasminogen/immunology , Proteome/genetics , Proteome/immunology , T-Lymphocytes/parasitology , Thymus Gland/metabolism , Thymus Gland/parasitology
13.
Nat Commun ; 10(1): 2042, 2019 05 03.
Article in English | MEDLINE | ID: mdl-31053703

ABSTRACT

Metabolic pathways that regulate T-cell function show promise as therapeutic targets in diverse diseases. Here, we show that at rest cultured human effector memory and central memory CD4+ T-cells have elevated levels of glycolysis and oxidative phosphorylation (OXPHOS), in comparison to naïve T-cells. Despite having low resting metabolic rates, naive T-cells respond to TCR stimulation with robust and rapid increases in glycolysis and OXPHOS. This early metabolic switch requires Akt activity to support increased rates of glycolysis and STAT5 activity for amino acid biosynthesis and TCA cycle anaplerosis. Importantly, both STAT5 inhibition and disruption of TCA cycle anaplerosis are associated with reduced IL-2 production, demonstrating the functional importance of this early metabolic program. Our results define STAT5 as a key node in modulating the early metabolic program following activation in naive CD4+ T-cells and in turn provide greater understanding of how cellular metabolism shapes T-cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/metabolism , STAT5 Transcription Factor/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Citric Acid Cycle/immunology , Glycolysis/immunology , Healthy Volunteers , Humans , Immunologic Memory , Lymphocyte Activation , Oxidative Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins c-akt/immunology , Receptors, Antigen, T-Cell/immunology , STAT5 Transcription Factor/immunology
14.
Cell ; 177(5): 1201-1216.e19, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31031005

ABSTRACT

Innate immune responses are intricately linked with intracellular metabolism of myeloid cells. Toll-like receptor (TLR) stimulation shifts intracellular metabolism toward glycolysis, while anti-inflammatory signals depend on enhanced mitochondrial respiration. How exogenous metabolic signals affect the immune response is unknown. We demonstrate that TLR-dependent responses of dendritic cells (DCs) are exacerbated by a high-fatty-acid (FA) metabolic environment. FAs suppress the TLR-induced hexokinase activity and perturb tricarboxylic acid cycle metabolism. These metabolic changes enhance mitochondrial reactive oxygen species (mtROS) production and, in turn, the unfolded protein response (UPR), leading to a distinct transcriptomic signature with IL-23 as hallmark. Interestingly, chemical or genetic suppression of glycolysis was sufficient to induce this specific immune response. Conversely, reducing mtROS production or DC-specific deficiency in XBP1 attenuated IL-23 expression and skin inflammation in an IL-23-dependent model of psoriasis. Thus, fine-tuning of innate immunity depends on optimization of metabolic demands and minimization of mtROS-induced UPR.


Subject(s)
Cellular Microenvironment/immunology , Dendritic Cells/immunology , Immunity, Innate , Mitochondria/immunology , Reactive Oxygen Species/immunology , Unfolded Protein Response/immunology , Animals , Cellular Microenvironment/genetics , Citric Acid Cycle/genetics , Citric Acid Cycle/immunology , Dendritic Cells/pathology , Hexokinase/genetics , Hexokinase/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Unfolded Protein Response/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology
15.
J Proteome Res ; 17(11): 3780-3790, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30229649

ABSTRACT

Despite osteoarthritis (OA) and rheumatoid arthritis (RA) being typically age-related, their underlying etiologies are markedly different. We used 1H nuclear magnetic resonance (NMR) spectroscopy to identify differences in metabolite profiles in low volumes of OA and RA synovial fluid (SF). SF was aspirated from knee joints of 10 OA and 14 RA patients. 100 µL SF was analyzed using a 700 MHz Avance IIIHD Bruker NMR spectrometer with a TCI cryoprobe. Spectra were analyzed by Chenomx, Bruker TopSpin and AMIX software. Statistical analysis was undertaken using Metaboanalyst. 50 metabolites were annotated, including amino acids, saccharides, nucleotides and soluble lipids. Discriminant analysis identified group separation between OA and RA cohorts, with 32 metabolites significantly different between OA and RA SF (false discovery rate (FDR) < 0.05). Metabolites of glycolysis and the tricarboxylic acid cycle were lower in RA compared to OA; these results concur with higher levels of inflammation, synovial proliferation and hypoxia found in RA compared to OA. Elevated taurine in OA may indicate increased subchondral bone sclerosis. We demonstrate that quantifiable differences in metabolite abundance can be measured in low volumes of SF by 1H NMR spectroscopy, which may be clinically useful to aid diagnosis and improve understanding of disease pathogenesis.


Subject(s)
Arthritis, Rheumatoid/metabolism , Magnetic Resonance Spectroscopy/methods , Metabolome , Metabolomics/methods , Osteoarthritis/metabolism , Synovial Fluid/chemistry , Aged , Amino Acids/chemistry , Amino Acids/classification , Amino Acids/isolation & purification , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Citric Acid Cycle/immunology , Cohort Studies , Female , Glycolysis/immunology , Humans , Knee Joint/immunology , Knee Joint/metabolism , Knee Joint/pathology , Lipids/chemistry , Lipids/classification , Lipids/isolation & purification , Male , Metabolomics/instrumentation , Middle Aged , Nucleotides/chemistry , Nucleotides/classification , Nucleotides/isolation & purification , Oligosaccharides/chemistry , Oligosaccharides/classification , Oligosaccharides/isolation & purification , Osteoarthritis/immunology , Osteoarthritis/pathology , Synovial Fluid/metabolism
16.
Nat Cell Biol ; 20(1): 21-27, 2018 01.
Article in English | MEDLINE | ID: mdl-29230018

ABSTRACT

CD8+ memory T (Tm) cells are fundamental for protective immunity against infections and cancers 1-5 . Metabolic activities are crucial in controlling memory T-cell homeostasis, but mechanisms linking metabolic signals to memory formation and survival remain elusive. Here we show that CD8+ Tm cells markedly upregulate cytosolic phosphoenolpyruvate carboxykinase (Pck1), the hub molecule regulating glycolysis, tricarboxylic acid cycle and gluconeogenesis, to increase glycogenesis via gluconeogenesis. The resultant glycogen is then channelled to glycogenolysis to generate glucose-6-phosphate and the subsequent pentose phosphate pathway (PPP) that generates abundant NADPH, ensuring high levels of reduced glutathione in Tm cells. Abrogation of Pck1-glycogen-PPP decreases GSH/GSSG ratios and increases levels of reactive oxygen species (ROS), leading to impairment of CD8+ Tm formation and maintenance. Importantly, this metabolic regulatory mechanism could be readily translated into more efficient T-cell immunotherapy in mouse tumour models.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glycogen/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Melanoma, Experimental/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Skin Neoplasms/genetics , 3-Mercaptopropionic Acid/pharmacology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Citric Acid Cycle/immunology , Enzyme Inhibitors/pharmacology , Female , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Gluconeogenesis/immunology , Glucose/immunology , Glycogen/immunology , Glycolysis/drug effects , Glycolysis/genetics , Glycolysis/immunology , Homeostasis/immunology , Immunologic Memory , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADP/immunology , NADP/metabolism , Pentose Phosphate Pathway/drug effects , Pentose Phosphate Pathway/genetics , Pentose Phosphate Pathway/immunology , Phosphoenolpyruvate Carboxykinase (GTP)/antagonists & inhibitors , Phosphoenolpyruvate Carboxykinase (GTP)/immunology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/metabolism
17.
Nat Immunol ; 18(9): 985-994, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714978

ABSTRACT

Glutamine metabolism provides synergistic support for macrophage activation and elicitation of desirable immune responses; however, the underlying mechanisms regulated by glutamine metabolism to orchestrate macrophage activation remain unclear. Here we show that the production of α-ketoglutarate (αKG) via glutaminolysis is important for alternative (M2) activation of macrophages, including engagement of fatty acid oxidation (FAO) and Jmjd3-dependent epigenetic reprogramming of M2 genes. This M2-promoting mechanism is further modulated by a high αKG/succinate ratio, whereas a low ratio strengthens the proinflammatory phenotype in classically activated (M1) macrophages. As such, αKG contributes to endotoxin tolerance after M1 activation. This study reveals new mechanistic regulations by which glutamine metabolism tailors the immune responses of macrophages through metabolic and epigenetic reprogramming.


Subject(s)
Cellular Reprogramming/immunology , Epigenesis, Genetic , Ketoglutaric Acids/immunology , Macrophage Activation/immunology , Macrophages/immunology , Animals , Chromatin Immunoprecipitation , Citric Acid Cycle/immunology , Fatty Acids/metabolism , Gene Expression Profiling , Glutamine/metabolism , Glycolysis/immunology , Ketoglutaric Acids/metabolism , Lipopolysaccharides , Macrophages/metabolism , Metabolomics , Mice , NF-kappa B/immunology , Oxidation-Reduction , Oxidative Phosphorylation , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Succinic Acid/metabolism
18.
Nat Immunol ; 18(5): 488-498, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28418387

ABSTRACT

Recent evidence indicates that mitochondria lie at the heart of immunity. Mitochondrial DNA acts as a danger-associated molecular pattern (DAMP), and the mitochondrial outer membrane is a platform for signaling molecules such as MAVS in RIG-I signaling, and for the NLRP3 inflammasome. Mitochondrial biogenesis, fusion and fission have roles in aspects of immune-cell activation. Most important, Krebs cycle intermediates such as succinate, fumarate and citrate engage in processes related to immunity and inflammation, in both innate and adaptive immune cells. These discoveries are revealing mitochondrial targets that could potentially be exploited for therapeutic gain in inflammation and cancer.


Subject(s)
Adaptive Immunity , Citric Acid Cycle/immunology , Immunity, Innate , Mitochondria/immunology , Mitochondrial Membranes/immunology , Animals , DEAD Box Protein 58/metabolism , Energy Metabolism , Humans , Immunomodulation , Inflammasomes/metabolism , Lymphocyte Activation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Immunologic , Receptors, Pattern Recognition/metabolism , Signal Transduction
19.
Cancer Lett ; 390: 45-47, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28109906

ABSTRACT

Succinate is an important intermediate of the tricarboxylic acid (TCA) cycle. In mitochondria, it plays a crucial role in generating adenosine triphosphate. Succinate metabolism is also intertwined with the metabolism of other metabolites and with the "GABA shunt" of the glutamine pathway. Recently, it has become increasingly apparent that the roles of succinate extend into the realms of immunity and cancer. Succinate is a key modulator of the hypoxic response, an important player in tumorigenesis; succinate is also involved in protein succinylation, a novel posttranslational modification pathway. This expanding repertoire of succinate functions suggests that it has broad roles in cellular contexts. Mutations in enzymes such as succinate dehydrogenase (SDH) that participate in succinate-related pathways lead to various pathologies, including tumor formation and innate inflammatory processes. Succinate can have both pro- or anti-tumor effectiveness. Therefore, investigation of succinate as an inflammatory signal may increase our understanding of the cancer-immunity cycle involved in both inflammatory diseases and cancer. Here, we briefly review the emerging roles of succinate, extending beyond metabolism, into anti-cancer immunity. This expansion of succinate roles suggests that it may represent a novel class of regulators in inflammation, which act as key signals in human cancers.


Subject(s)
Neoplasms/immunology , Succinic Acid/metabolism , Citric Acid Cycle/immunology , Humans , Inflammation/immunology , Neoplasms/physiopathology
20.
Trends Biochem Sci ; 41(5): 460-471, 2016 05.
Article in English | MEDLINE | ID: mdl-26935843

ABSTRACT

The integration of biochemistry into immune cell biology has contributed immensely to our understanding of immune cell function and the associated pathologies. So far, most studies have focused on the regulation of metabolic pathways during an immune response and their contribution to its success. More recently, novel signalling functions of metabolic intermediates are being discovered that might play important roles in the regulation of immunity. Here we describe the three long-known small metabolites lactate, acetyl-CoA, and succinate in the context of immunometabolic signalling. Functions of these ubiquitous molecules are largely dependent on their intra- and extracellular concentrations as well as their subcompartmental localisation. Importantly, the signalling functions of these metabolic intermediates extend beyond self-regulatory roles and include cell-to-cell communication and sensing of microenvironmental conditions to elicit stress responses and cellular adaptation.


Subject(s)
Citric Acid Cycle/immunology , Glycolysis/immunology , Immunity, Innate , Macrophages/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism , Acetyl Coenzyme A/immunology , Acetyl Coenzyme A/metabolism , Cell Communication/immunology , Cytokines/biosynthesis , Cytokines/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Fatty Acids/immunology , Fatty Acids/metabolism , Humans , Lactic Acid/immunology , Lactic Acid/metabolism , Macrophages/cytology , Macrophages/immunology , Neurons/cytology , Neurons/immunology , Neurons/metabolism , Succinic Acid/immunology , Succinic Acid/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL