Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 4331, 2024 02 21.
Article in English | MEDLINE | ID: mdl-38383709

ABSTRACT

Insulin and insulin-like growth factor 1 (IGF-1) are metabolic hormones with known effects on CD4+ T cells through insulin receptor (IR) and IGF-1 receptor (IGF-1R) signaling. Here, we describe specific and distinct roles for these hormones and receptors. We have found that IGF-1R, but not IR, expression is increased following CD4+ T cell activation or following differentiation toward Th17 cells. Although both insulin and IGF-1 increase the metabolism of CD4+ T cells, insulin has a more potent effect. However, IGF-1 has a unique role and acts specifically on Th17 cells to increase IL-17 production and Th17 cell metabolism. Furthermore, IGF-1 decreases mitochondrial membrane potential and mitochondrial reactive oxygen species (mROS) in Th17 cells, providing a cytoprotective effect. Interestingly, both IR and IGF-1R are required for this effect of IGF-1 on mitochondria, which suggests that the hybrid IR/IGF-1R may be required for mediating the effect of IGF-1 on mitochondrial membrane potential and mROS production.


Subject(s)
Insulin-Like Growth Factor I , Insulin , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Signal Transduction , Mitochondria/metabolism , CD4-Positive T-Lymphocytes/metabolism
2.
Nat Rev Endocrinol ; 20(3): 136-148, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38129700

ABSTRACT

Obesity is associated with a wide range of complications, including type 2 diabetes mellitus, cardiovascular disease, hypertension and nonalcoholic fatty liver disease. Obesity also increases the incidence and progression of cancers, autoimmunity and infections, as well as lowering vaccine responsiveness. A unifying concept across these differing diseases is dysregulated immunity, particularly inflammation, in response to metabolic overload. Herein, we review emerging mechanisms by which obesity drives inflammation and autoimmunity, as well as impairing tumour immunosurveillance and the response to infections. Among these mechanisms are obesity-associated changes in the hormones that regulate immune cell metabolism and function and drive inflammation. The cargo of extracellular vesicles derived from adipose tissue, which controls cytokine secretion from immune cells, is also dysregulated in obesity, in addition to impairments in fatty acid metabolism related to inflammation. Furthermore, an imbalance exists in obesity in the biosynthesis and levels of polyunsaturated fatty acid-derived oxylipins, which control a range of outcomes related to inflammation, such as immune cell chemotaxis and cytokine production. Finally, there is a need to investigate how obesity influences immunity using innovative model systems that account for the heterogeneous nature of obesity in the human population.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Inflammation/metabolism , Cytokines
3.
PLoS One ; 18(6): e0286470, 2023.
Article in English | MEDLINE | ID: mdl-37276236

ABSTRACT

BACKGROUND/OBJECTIVES: Leptin is an adipokine secreted in proportion to adipocyte mass and is therefore increased in obesity. Leptin signaling has been shown to directly promote inflammatory T helper 1 (Th1) and T helper 17 (Th17) cell number and function. Since T cells have a critical role in driving inflammation and systemic glucose intolerance in obesity, we sought to determine the role of leptin signaling in this context. METHODS: Male and female T cell-specific leptin receptor knockout mice and littermate controls were placed on low-fat diet or high-fat diet to induce obesity for 18 weeks. Weight gain, serum glucose levels, systemic glucose tolerance, T cell metabolism, and T cell differentiation and cytokine production were examined. RESULTS: In both male and female mice, T cell-specific leptin receptor deficiency did not reverse impaired glucose tolerance in obesity, although it did prevent impaired fasting glucose levels in obese mice compared to littermate controls, in a sex dependent manner. Despite these minimal effects on systemic metabolism, T cell-specific leptin signaling was required for changes in T cell metabolism, differentiation, and cytokine production observed in mice fed high-fat diet compared to low-fat diet. Specifically, we observed increased T cell oxidative metabolism, increased CD4+ T cell IFN-γ expression, and increased proportion of T regulatory (Treg) cells in control mice fed high-fat diet compared to low-fat diet, which were not observed in the leptin receptor conditional knockout mice, suggesting that leptin receptor signaling is required for some of the inflammatory changes observed in T cells in obesity. CONCLUSIONS: T cell-specific deficiency of leptin signaling alters T cell metabolism and function in obesity but has minimal effects on obesity-associated systemic metabolism. These results suggest a redundancy in cytokine receptor signaling pathways in response to inflammatory signals in obesity.


Subject(s)
Glucose Intolerance , Leptin , Animals , Female , Male , Mice , Cytokines , Diet, High-Fat/adverse effects , Glucose/metabolism , Leptin/metabolism , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Receptors, Leptin/genetics
4.
J Infect Dis ; 227(1): 92-102, 2022 12 28.
Article in English | MEDLINE | ID: mdl-35975968

ABSTRACT

BACKGROUND: Obesity dysregulates immunity to influenza infection. Therefore, there is a critical need to investigate how obesity impairs immunity and to establish therapeutic approaches that mitigate the impact of increased adiposity. One mechanism by which obesity may alter immune responses is through changes in cellular metabolism. METHODS: We studied inflammation and cellular metabolism of peripheral blood mononuclear cells (PBMCs) isolated from individuals with obesity relative to lean controls. We also investigated if impairments to PBMC metabolism were reversible upon short-term weight loss following bariatric surgery. RESULTS: Obesity was associated with systemic inflammation and poor inflammation resolution. Unstimulated PBMCs from participants with obesity had lower oxidative metabolism and adenosine triphosphate (ATP) production compared to PBMCs from lean controls. PBMC secretome analyses showed that ex vivo stimulation with A/Cal/7/2009 H1N1 influenza led to a notable increase in IL-6 with obesity. Short-term weight loss via bariatric surgery improved biomarkers of systemic metabolism but did not improve markers of inflammation resolution, PBMC metabolism, or the PBMC secretome. CONCLUSIONS: These results show that obesity drives a signature of impaired PBMC metabolism, which may be due to persistent inflammation. PBMC metabolism was not reversed after short-term weight loss despite improvements in measures of systemic metabolism.


Subject(s)
Bariatric Surgery , Influenza A Virus, H1N1 Subtype , Influenza, Human , Humans , Adult , Leukocytes, Mononuclear , Influenza, Human/metabolism , Obesity/surgery , Obesity/metabolism , Inflammation/metabolism , Weight Loss
5.
Sci Rep ; 12(1): 850, 2022 01 17.
Article in English | MEDLINE | ID: mdl-35039539

ABSTRACT

Immunity Related GTPases (IRG) are a family of proteins produced during infection that regulate membrane remodeling events in cells, particularly autophagy and mitophagy. The human IRGM gene has been strongly associated with Crohn's disease and other inflammatory diseases through Genome-Wide Association studies. Absence of Irgm1 in mice prompts intestinal inflammation, autoimmunity, and impaired immune control of pathogenic bacteria and protozoa. Although prior work has focused on a prominent role for IRGM/Irgm1 in regulating macrophage function, the work described here addresses a potential role of Irgm1 in regulating the function of mature T cells. Irgm1 was found to be highly expressed in T cells in a manner that varied with the particular T cell subset and increased with activation. Mice with a complete lack of Irgm1, or a conditional lack of Irgm1 specifically in T cells, displayed numerous changes in T cell numbers and function in all subsets examined, including CD4+ (Th1 and Treg) and CD8+ T cells. Related to changes in T cell number, apoptosis was found to be increased in Irgm1-deficient CD4+ and CD8+ T cells. Altered T cell metabolism appeared to be a key driver of the phenotypes: Glucose metabolism and glycolysis were increased in Irgm1-deficient CD4+ and CD8+ T cells, and muting these effects with glycolytic inhibitors partially restored T cell function and viability.


Subject(s)
GTP-Binding Proteins/physiology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/physiology , Animals , Apoptosis/genetics , Autophagy/genetics , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cells, Cultured , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression/genetics , Glucose/metabolism , Glycolysis , Lymphocyte Activation/genetics , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocyte Subsets/immunology
6.
Int J Obes (Lond) ; 44(12): 2419-2429, 2020 12.
Article in English | MEDLINE | ID: mdl-33037327

ABSTRACT

BACKGROUND: Obesity is associated with impaired primary and secondary immune responses to influenza infection, with T cells playing a critical role. T-cell function is highly influenced by the cellular metabolic state; however, it remains unknown how altered systemic metabolism in obesity alters T-cell metabolism and function to influence immune response. Our objective was to identify the altered cellular metabolic state of T cells from obese mice so that we may target T-cell metabolism to improve immune response to infection. METHODS: Mice were fed normal chow or high-fat diet for 18-19 weeks. Changes in T-cell populations were analyzed in both adipose tissue and spleens using flow cytometry. Splenic T cells were further analyzed for nutrient uptake and extracellular metabolic flux. As changes in T-cell mitochondrial oxidation were observed in obesity, obese mice were treated with metformin for 6 weeks and compared to lean control mice or obese mice undergoing weight loss through diet switch; immunity was measured by survival to influenza infection. RESULTS: We found changes in T-cell populations in adipose tissue of high-fat diet-induced obese mice, characterized by decreased proportions of Treg cells and increased proportions of CD8+ T cells. Activated CD4+ T cells from obese mice had increased glucose uptake and oxygen consumption rate (OCR), compared to T cells from lean controls, indicating increased mitochondrial oxidation of glucose. Treatment of isolated CD4+ T cells with metformin was found to inhibit OCR in vitro and alter the expression of several activation markers. Last, treatment of obese mice with metformin, but not weight loss, was able to improve survival to influenza in obesity. CONCLUSIONS: T cells from obese mice have an altered metabolic profile characterized by increased glucose oxidation, which can be targeted to improve survival against influenza infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Obesity/immunology , Orthomyxoviridae Infections/immunology , Oxidative Stress , Adipose Tissue/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Diet, High-Fat , Influenza A Virus, H1N1 Subtype , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Obesity/metabolism , Spleen/immunology
7.
J Infect Dis ; 219(10): 1652-1661, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30535161

ABSTRACT

BACKGROUND: Obesity is an independent risk factor for increased influenza mortality and is associated with impaired memory T-cell response, resulting in increased risk of infection. In this study, we investigated if weight loss would restore memory T-cell response to influenza. METHODS: Male C57BL/6J mice were fed either low-fat or high-fat diet to induce obesity. Once obesity was established, all mice received primary infection with influenza X-31. Following a recovery period, we switched half of the obese group to a low-fat diet to induce weight loss. Fifteen weeks after diet switch, all mice were given a secondary infection with influenza PR8, and memory T-cell function and T-cell metabolism were measured. RESULTS: Following secondary influenza infection, memory T-cell subsets in the lungs of obese mice were decreased compared to lean mice. At the same time, T cells from obese mice were found to have altered cellular metabolism, largely characterized by an increase in oxygen consumption. Neither impaired memory T-cell response nor altered T-cell metabolism was reversed with weight loss. CONCLUSION: Obesity-associated changes in T-cell metabolism are associated with impaired T-cell response to influenza, and are not reversed with weight loss.


Subject(s)
Immunologic Memory/physiology , Obesity/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Diet, High-Fat , Influenza A virus , Male , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Oxygen Consumption , Weight Loss/physiology
8.
Front Immunol ; 9: 1055, 2018.
Article in English | MEDLINE | ID: mdl-29868016

ABSTRACT

Immune cell function and metabolism are closely linked. Many studies have now clearly demonstrated that alterations in cellular metabolism influence immune cell function and that, conversely, immune cell function determines the cellular metabolic state. Less well understood, however, are the effects of systemic metabolism or whole organism nutritional status on immune cell function and metabolism. Several studies have demonstrated that undernutrition is associated with immunosuppression, which leads to both increased susceptibility to infection and protection against several types of autoimmune disease, whereas overnutrition is associated with low-grade, chronic inflammation that increases the risk of metabolic and cardiovascular disease, promotes autoreactivity, and disrupts protective immunity. Here, we review the effects of nutritional status on immunity and highlight the effects of nutrition on circulating cytokines and immune cell populations in both human studies and mouse models. As T cells are critical members of the immune system, which direct overall immune response, we will focus this review on the influence of systemic nutritional status on T cell metabolism and function. Several cytokines and hormones have been identified which mediate the effects of nutrition on T cell metabolism and function through the expression and action of key regulatory signaling proteins. Understanding how T cells are sensitive to both inadequate and overabundant nutrients may enhance our ability to target immune cell metabolism and alter immunity in both malnutrition and obesity.


Subject(s)
Energy Metabolism , Immune System/cytology , Immune System/physiology , Nutritional Status , Animals , Biomarkers , Disease Susceptibility , Humans , Immune System/drug effects , Immunity/drug effects , Molecular Targeted Therapy , Signal Transduction
9.
Retrovirology ; 14(1): 45, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28962653

ABSTRACT

BACKGROUND: Like all viruses, HIV-1 relies on host systems to replicate. The human purinome consists of approximately two thousand proteins that bind and use purines such as ATP, NADH, and NADPH. By virtue of their purine binding pockets, purinome proteins are highly druggable, and many existing drugs target purine-using enzymes. Leveraging a protein affinity media that uses the purine-binding pocket to capture the entire purinome, we sought to define purine-binding proteins regulated by HIV-1 infection. RESULTS: Using purinome capture media, we observed that HIV-1 infection increases intracellular levels of fatty acid synthase (FASN), a NADPH-using enzyme critical to the synthesis of de novo fatty acids. siRNA mediated knockdown of FASN reduced HIV-1 particle production by 80%, and treatment of tissue culture cells or primary PBMCs with Fasnall, a newly described selective FASN inhibitor, reduced HIV-1 virion production by 90% (EC50 = 213 nM). Despite the requirement of FASN for nascent virion production, FASN activity was not required for intracellular Gag protein production, indicating that FASN dependent de novo fatty acid biosynthesis contributes to a late step of HIV-1 replication. CONCLUSIONS: Here we show that HIV-1 replication both increases FASN levels and requires host FASN activity. We also report that Fasnall, a novel FASN inhibitor that demonstrates anti-tumor activity in vivo, is a potent and efficacious antiviral, blocking HIV-1 replication in both tissue culture and primary cell models of HIV-1 replication. In adults, most fatty acids are obtained exogenously from the diet, thus making FASN a plausible candidate for pharmacological intervention. In conclusion, we hypothesize that FASN is a novel host dependency factor and that inhibition of FASN activity has the potential to be exploited as an antiretroviral strategy.


Subject(s)
Fatty Acid Synthase, Type I/metabolism , HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , Virus Replication/physiology , Antiviral Agents/pharmacology , Cell Line, Tumor , Chromatography, Affinity , Fatty Acid Synthase, Type I/antagonists & inhibitors , Fatty Acid Synthase, Type I/genetics , Gene Expression Regulation, Enzymologic , HIV Core Protein p24/metabolism , HIV Infections/immunology , HIV-1/drug effects , HIV-1/enzymology , Humans , Proteomics , Pyrimidines/pharmacology , RNA Interference , RNA Processing, Post-Transcriptional , Sepharose/chemistry , Thiophenes/pharmacology , Virion/physiology , Virus Replication/drug effects , gag Gene Products, Human Immunodeficiency Virus/metabolism
10.
Cell Chem Biol ; 23(6): 678-88, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27265747

ABSTRACT

Many tumors are dependent on de novo fatty acid synthesis to maintain cell growth. Fatty acid synthase (FASN) catalyzes the final synthetic step of this pathway, and its upregulation is correlated with tumor aggressiveness. The consequences and adaptive responses of acute or chronic inhibition of essential enzymes such as FASN are not fully understood. Herein we identify Fasnall, a thiophenopyrimidine selectively targeting FASN through its co-factor binding sites. Global lipidomics studies with Fasnall showed profound changes in cellular lipid profiles, sharply increasing ceramides, diacylglycerols, and unsaturated fatty acids as well as increasing exogenous palmitate uptake that is deviated more into neutral lipid formation rather than phospholipids. We also showed that the increase in ceramide levels contributes to some extent in the mediation of apoptosis. Consistent with this mechanism of action, Fasnall showed potent anti-tumor activity in the MMTV-Neu model of HER2(+) breast cancer, particularly when combined with carboplatin.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Fatty Acid Synthase, Type I/antagonists & inhibitors , Pyrimidines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Thiophenes/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Fatty Acid Synthase, Type I/metabolism , Female , Humans , Injections, Intraperitoneal , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Receptor, ErbB-2/metabolism , Swine , Thiophenes/administration & dosage , Thiophenes/chemistry
11.
Chem Biol ; 21(12): 1648-59, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25500222

ABSTRACT

Inducible Hsp70 (Hsp70i) is overexpressed in a wide spectrum of human tumors, and its expression correlates with metastasis, poor outcomes, and resistance to chemotherapy in patients. Identification of small-molecule inhibitors selective for Hsp70i could provide new therapeutic tools for cancer treatment. In this work, we used fluorescence-linked enzyme chemoproteomic strategy (FLECS) to identify HS-72, an allosteric inhibitor selective for Hsp70i. HS-72 displays the hallmarks of Hsp70 inhibition in cells, promoting substrate protein degradation and growth inhibition. Importantly, HS-72 is selective for Hsp70i over the closely related constitutively active Hsc70. Studies with purified protein show HS-72 acts as an allosteric inhibitor, reducing ATP affinity. In vivo HS-72 is well-tolerated, showing bioavailability and efficacy, inhibiting tumor growth and promoting survival in a HER2+ model of breast cancer. The HS-72 scaffold is amenable to resynthesis and iteration, suggesting an ideal starting point for a new generation of anticancer therapeutics targeting Hsp70i.


Subject(s)
Benzimidazoles/chemistry , Benzimidazoles/pharmacology , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/metabolism , Nipecotic Acids/chemistry , Nipecotic Acids/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Allosteric Regulation/drug effects , Animals , Benzimidazoles/metabolism , Benzimidazoles/pharmacokinetics , Biological Availability , Caspases/metabolism , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , HEK293 Cells , HSP70 Heat-Shock Proteins/chemistry , Humans , Mice , Models, Molecular , Nipecotic Acids/metabolism , Nipecotic Acids/pharmacokinetics , Permeability , Piperidines/metabolism , Piperidines/pharmacokinetics , Protein Aggregates/drug effects , Protein Structure, Tertiary , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...