Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell Rep Med ; 4(5): 101051, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37196633

ABSTRACT

Alterations in the microbiome correlate with improved metabolism in patients following bariatric surgery. While fecal microbiota transplantation (FMT) from obese patients into germ-free (GF) mice has suggested a significant role of the gut microbiome in metabolic improvements following bariatric surgery, causality remains to be confirmed. Here, we perform paired FMT from the same obese patients (BMI > 40; four patients), pre- and 1 or 6 months post-Roux-en-Y gastric bypass (RYGB) surgery, into Western diet-fed GF mice. Mice colonized by FMT from patients' post-surgery stool exhibit significant changes in microbiota composition and metabolomic profiles and, most importantly, improved insulin sensitivity compared with pre-RYGB FMT mice. Mechanistically, mice harboring the post-RYGB microbiome show increased brown fat mass and activity and exhibit increased energy expenditure. Moreover, improvements in immune homeostasis within the white adipose tissue are also observed. Altogether, these findings point to a direct role for the gut microbiome in mediating improved metabolic health post-RYGB surgery.


Subject(s)
Bariatric Surgery , Gastrointestinal Microbiome , Insulin Resistance , Mice , Animals , Adipose Tissue, Brown , Obesity/surgery , Energy Metabolism
2.
Cell Rep ; 34(2): 108609, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33440149

ABSTRACT

Stiffness in the tissue microenvironment changes in most diseases and immunological conditions, but its direct influence on the immune system is poorly understood. Here, we show that static tension impacts immune cell function, maturation, and metabolism. Bone-marrow-derived and/or splenic dendritic cells (DCs) grown in vitro at physiological resting stiffness have reduced proliferation, activation, and cytokine production compared with cells grown under higher stiffness, mimicking fibro-inflammatory disease. Consistently, DCs grown under higher stiffness show increased activation and flux of major glucose metabolic pathways. In DC models of autoimmune diabetes and tumor immunotherapy, tension primes DCs to elicit an adaptive immune response. Mechanistic workup identifies the Hippo-signaling molecule, TAZ, as well as Ca2+-related ion channels, including potentially PIEZO1, as important effectors impacting DC metabolism and function under tension. Tension also directs the phenotypes of monocyte-derived DCs in humans. Thus, mechanical stiffness is a critical environmental cue of DCs and innate immunity.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate/immunology , Immunotherapy/methods , Vascular Stiffness/immunology , Humans , Signal Transduction
3.
Nat Commun ; 10(1): 3650, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409776

ABSTRACT

The intestinal immune system is emerging as an important contributor to obesity-related insulin resistance, but the role of intestinal B cells in this context is unclear. Here, we show that high fat diet (HFD) feeding alters intestinal IgA+ immune cells and that IgA is a critical immune regulator of glucose homeostasis. Obese mice have fewer IgA+ immune cells and less secretory IgA and IgA-promoting immune mediators. HFD-fed IgA-deficient mice have dysfunctional glucose metabolism, a phenotype that can be recapitulated by adoptive transfer of intestinal-associated pan-B cells. Mechanistically, IgA is a crucial link that controls intestinal and adipose tissue inflammation, intestinal permeability, microbial encroachment and the composition of the intestinal microbiome during HFD. Current glucose-lowering therapies, including metformin, affect intestinal-related IgA+ B cell populations in mice, while bariatric surgery regimen alters the level of fecal secretory IgA in humans. These findings identify intestinal IgA+ immune cells as mucosal mediators of whole-body glucose regulation in diet-induced metabolic disease.


Subject(s)
Immunoglobulin A/immunology , Insulin Resistance , Obesity/immunology , Adipose Tissue/immunology , Animals , B-Lymphocytes/immunology , Cohort Studies , Feces/microbiology , Gastrointestinal Microbiome , Glucose/metabolism , Humans , Intestines/immunology , Male , Mice , Obesity/metabolism , Obesity/microbiology
4.
Int J Obes (Lond) ; 43(12): 2407-2421, 2019 12.
Article in English | MEDLINE | ID: mdl-30944419

ABSTRACT

BACKGROUND/OBJECTIVES: Low-grade chronic inflammation in visceral adipose tissue and the intestines are important drivers of obesity associated insulin resistance. Bioactive compounds derived from plants are an important source of potential novel therapies for the treatment of chronic diseases. In search for new immune based treatments of obesity associated insulin resistance, we screened for tissue relevant anti-inflammatory properties in 20 plant-based extracts. METHODS: We screened 20 plant-based extracts to assess for preferential production of IL-10 compared to TNFα, specifically targetting metabolic tissues, including the visceral adipose tissue. We assessed the therapeutic potential of the strongest anti-inflammatory compound, indigo, in the C57BL/6J diet-induced obesity mouse model with supplementation for up to 16 weeks by measuring changes in body weight, glucose and insulin tolerance, and gut barrier function. We also utilized flow cytometry, quantitative PCR, enzyme-linked immunosorbent assay (ELISA), and histology to measure changes to immune cells populations and cytokine profiles in the intestine, visceral adipose tissue (VAT), and liver. 16SrRNA sequencing was performed to examine gut microbial differences induced by indigo supplementation. RESULTS: We identifed indigo, an aryl hydrocarbon receptor (AhR) ligand agonist, as a potent inducer of IL-10 and IL-22, which protects against high-fat diet (HFD)-induced insulin resistance and fatty liver disease in the diet-induced obesity model. Therapeutic actions were mechanistically linked to decreased inflammatory immune cell tone in the intestine, VAT and liver. Specifically, indigo increased Lactobacillus bacteria and elicited IL-22 production in the gut, which improved intestinal barrier permeability and reduced endotoxemia. These changes were associated with increased IL-10 production by immune cells residing in liver and VAT. CONCLUSIONS: Indigo is a naturally occurring AhR ligand with anti-inflammatory properties that effectively protects against HFD-induced glucose dysregulation. Compounds derived from indigo or those with similar properties could represent novel therapies for diseases associated with obesity-related metabolic tissue inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Indigo Carmine/pharmacology , Insulin Resistance/physiology , Obesity/metabolism , Receptors, Aryl Hydrocarbon/agonists , Animals , Cytokines/metabolism , Diet, High-Fat , Gastrointestinal Microbiome , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Plant Extracts/chemistry
5.
Cell Metab ; 28(6): 922-934.e4, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30174303

ABSTRACT

T cells represent a critical effector of cell-mediated immunity. Activated T cells engage in metabolic reprogramming during effector differentiation to accommodate dynamic changes in energy demands. Here, we show that the hormone, insulin, and downstream signaling through its insulin receptor shape adaptive immune function through modulating T cell metabolism. T cells lacking insulin receptor expression (LckCre+ Insrfl/fl) show reduced antigen-specific proliferation and compromised production of pro-inflammatory cytokines. In vivo, T cell-specific insulin receptor deficiency reduces T cell-driven colonic inflammation. In a model of severe influenza infection with A/PR8 (H1N1), lack of insulin receptor on T cells curtails antigen-specific immunity to influenza viral antigens. Mechanistically, insulin receptor signaling reinforces a metabolic program that supports T cell nutrient uptake and associated glycolytic and respiratory capacities. These data highlight insulin receptor signaling as an important node integrating immunometabolic pathways to drive optimal T cell effector function in health and disease.


Subject(s)
Antigens, CD/immunology , Immunity, Cellular/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Lymphocyte Activation/immunology , Receptor, Insulin/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/genetics , Cytokines/immunology , Cytokines/metabolism , Glycolysis/immunology , Humans , Inflammation/immunology , Inflammation/virology , Insulin/metabolism , Lymph Nodes , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections , Receptor, Insulin/genetics , Signal Transduction , Spleen , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
6.
Obesity (Silver Spring) ; 26(3): 547-558, 2018 03.
Article in English | MEDLINE | ID: mdl-29405636

ABSTRACT

OBJECTIVE: This study aimed to determine whether dietary curcumin intervention targets both white adipose tissue (WAT) inflammation and brown adipose tissue (BAT)-mediated energy expenditure. METHODS: C57BL/6J mice were fed with a low-fat diet, high-fat diet (HFD), or HFD plus curcumin. In addition to assessing the effect of curcumin intervention on metabolic profiles, this study assessed WAT macrophage infiltration and composition and inflammatory cytokine production. Metabolic cages were applied for determining energy expenditure. Raw264.7 (ATCC, Manassas, Virginia) and other cell models were utilized to test the in vitro effect of curcumin treatment. RESULTS: Curcumin intervention reduced WAT macrophage infiltration and altered macrophage functional polarity, as the ratio of M2-like versus M1-like macrophages increased after curcumin intervention. Curcumin treatment reduced M1-like macrophage markers or proinflammation cytokine expression in both macrophages and adipocytes. Curcumin intervention also increased energy expenditure and body temperature in response to a cold challenge. Finally, the in vivo and in vitro investigations suggested that curcumin increased expression of uncoupling protein 1 (UCP1), possibly involving PPAR-dependent and -independent mechanisms. CONCLUSIONS: Curcumin intervention targets both WAT inflammation and BAT UCP1 expression. These observations advanced our knowledge on the metabolic beneficial effects of the curry compound curcumin, bringing us a novel perspective on dietary polyphenol research.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Curcumin/therapeutic use , Uncoupling Protein 1/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Male , Mice , Mice, Inbred C57BL
7.
Sci Immunol ; 2(10)2017 Apr 21.
Article in English | MEDLINE | ID: mdl-28567448

ABSTRACT

Obesity-related insulin resistance is driven by low-grade chronic inflammation of metabolic tissues. In the liver, non-alcoholic fatty liver disease (NAFLD) is associated with hepatic insulin resistance and systemic glucose dysregulation. However, the immunological factors supporting these processes are poorly understood. We found that the liver accumulates pathogenic CD8+ T cell subsets which control hepatic insulin sensitivity and gluconeogenesis during diet-induced obesity in mice. In a cohort of human patients, CD8+ T cells represent a dominant intrahepatic immune cell population which links to glucose dysregulation. Accumulation and activation of these cells are largely supported by type I interferon (IFN-I) responses in the liver. Livers from obese mice upregulate critical interferon regulatory factors (IRFs), interferon stimulatory genes (ISGs), and IFNα protein, while IFNαR1-/- mice, or CD8-specific IFNαR1-/- chimeric mice are protected from disease. IFNαR1 inhibitors improve metabolic parameters in mice, while CD8+ T cells and IFN-I responses correlate with NAFLD activity in human patients. Thus, IFN-I responses represent a central immunological axis that governs intrahepatic T cell pathogenicity during metabolic disease.

8.
Cell Rep ; 16(3): 717-30, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27373163

ABSTRACT

Obesity-related inflammation of metabolic tissues, including visceral adipose tissue (VAT) and liver, are key factors in the development of insulin resistance (IR), though many of the contributing mechanisms remain unclear. We show that nucleic-acid-targeting pathways downstream of extracellular trap (ET) formation, unmethylated CpG DNA, or ribonucleic acids drive inflammation in IR. High-fat diet (HFD)-fed mice show increased release of ETs in VAT, decreased systemic clearance of ETs, and increased autoantibodies against conserved nuclear antigens. In HFD-fed mice, this excess of nucleic acids and related protein antigens worsens metabolic parameters through a number of mechanisms, including activation of VAT macrophages and expansion of plasmacytoid dendritic cells (pDCs) in the liver. Consistently, HFD-fed mice lacking critical responders of nucleic acid pathways, Toll-like receptors (TLR)7 and TLR9, show reduced metabolic inflammation and improved glucose homeostasis. Treatment of HFD-fed mice with inhibitors of ET formation or a TLR7/9 antagonist improves metabolic disease. These findings reveal a pathogenic role for nucleic acid targeting as a driver of metabolic inflammation in IR.


Subject(s)
Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/physiology , Nucleic Acids/metabolism , Obesity/metabolism , Obesity/pathology , Adult , Animals , Dendritic Cells/metabolism , Dendritic Cells/pathology , Diet, High-Fat/methods , Glucose/metabolism , Homeostasis/physiology , Humans , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Toll-Like Receptors/metabolism
9.
Cell Metab ; 21(4): 527-42, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25863246

ABSTRACT

Obesity has reached epidemic proportions, but little is known about its influence on the intestinal immune system. Here we show that the gut immune system is altered during high-fat diet (HFD) feeding and is a functional regulator of obesity-related insulin resistance (IR) that can be exploited therapeutically. Obesity induces a chronic phenotypic pro-inflammatory shift in bowel lamina propria immune cell populations. Reduction of the gut immune system, using beta7 integrin-deficient mice (Beta7(null)), decreases HFD-induced IR. Treatment of wild-type HFD C57BL/6 mice with the local gut anti-inflammatory, 5-aminosalicyclic acid (5-ASA), reverses bowel inflammation and improves metabolic parameters. These beneficial effects are dependent on adaptive and gut immunity and are associated with reduced gut permeability and endotoxemia, decreased visceral adipose tissue inflammation, and improved antigen-specific tolerance to luminal antigens. Thus, the mucosal immune system affects multiple pathways associated with systemic IR and represents a novel therapeutic target in this disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gastrointestinal Tract/immunology , Immunity, Mucosal/immunology , Insulin Resistance/immunology , Obesity/immunology , Animals , Blotting, Western , Cytokines/blood , Diet, High-Fat/adverse effects , Flow Cytometry , Gastrointestinal Tract/drug effects , Histological Techniques , Immunohistochemistry , Integrin beta Chains/metabolism , Mesalamine/pharmacology , Mice , Mice, Inbred C57BL , Mucous Membrane/cytology , Mucous Membrane/immunology
10.
Diabetes ; 64(1): 90-103, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25048196

ABSTRACT

Obesity-related insulin resistance is associated with an influx of pathogenic T cells into visceral adipose tissue (VAT), but the mechanisms regulating lymphocyte balance in such tissues are unknown. Here we describe an important role for the immune cytotoxic effector molecule perforin in regulating this process. Perforin-deficient mice (Prf1(null)) show early increased body weight and adiposity, glucose intolerance, and insulin resistance when placed on high-fat diet (HFD). Regulatory effects of perforin on glucose tolerance are mechanistically linked to the control of T-cell proliferation and cytokine production in inflamed VAT. HFD-fed Prf1(null) mice have increased accumulation of proinflammatory IFN-γ-producing CD4(+) and CD8(+) T cells and M1-polarized macrophages in VAT. CD8(+) T cells from the VAT of Prf1(null) mice have increased proliferation and impaired early apoptosis, suggesting a role for perforin in the regulation of T-cell turnover during HFD feeding. Transfer of CD8(+) T cells from Prf1(null) mice into CD8-deficient mice (CD8(null)) resulted in worsening of metabolic parameters compared with wild-type donors. Improved metabolic parameters in HFD natural killer (NK) cell-deficient mice (NK(null)) ruled out a role for NK cells as a single source of perforin in regulating glucose homeostasis. The findings support the importance of T-cell function in insulin resistance and suggest that modulation of lymphocyte homeostasis in inflamed VAT is one possible avenue for therapeutic intervention.


Subject(s)
Glucose Intolerance/immunology , Insulin Resistance/immunology , Intra-Abdominal Fat/immunology , Obesity/immunology , Panniculitis/immunology , Perforin/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Diet, High-Fat , Embryonic Stem Cells/cytology , Female , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Insulin/metabolism , Intra-Abdominal Fat/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Panniculitis/genetics , Panniculitis/metabolism , Perforin/genetics , Perforin/metabolism , Spleen/cytology , Spleen/immunology
11.
Dis Model Mech ; 6(2): 467-78, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23118341

ABSTRACT

Multiple sclerosis (MS) is the most common CNS-demyelinating disease of humans, showing clinical and pathological heterogeneity and a general resistance to therapy. We first discovered that abnormal myelin hypercitrullination, even in normal-appearing white matter, by peptidylarginine deiminases (PADs) correlates strongly with disease severity and might have an important role in MS progression. Hypercitrullination is known to promote focal demyelination through reduced myelin compaction. Here we report that 2-chloroacetamidine (2CA), a small-molecule, PAD active-site inhibitor, dramatically attenuates disease at any stage in independent neurodegenerative as well as autoimmune MS mouse models. 2CA reduced PAD activity and protein citrullination to pre-disease status. In the autoimmune models, disease induction uniformly induced spontaneous hypercitrullination with citrulline+ epitopes targeted frequently. 2CA rapidly suppressed T cell autoreactivity, clearing brain and spinal cord infiltrates, through selective removal of newly activated T cells. 2CA essentially prevented disease when administered before disease onset or before autoimmune induction, making hypercitrullination, and specifically PAD enzymes, a therapeutic target in MS models and thus possibly in MS.


Subject(s)
Citrulline/metabolism , Hydrolases/antagonists & inhibitors , Multiple Sclerosis/enzymology , Multiple Sclerosis/pathology , Amidines/chemistry , Amidines/pharmacology , Amidines/therapeutic use , Animals , Brain/enzymology , Brain/pathology , CD3 Complex/metabolism , Demyelinating Diseases/enzymology , Demyelinating Diseases/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Hydrolases/metabolism , Lymphocytes/drug effects , Lymphocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/drug therapy , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve/ultrastructure , Protein-Arginine Deiminases , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , Survival Analysis
12.
Dis Model Mech ; 1(4-5): 229-40, 2008.
Article in English | MEDLINE | ID: mdl-19093029

ABSTRACT

Demyelination in the central nervous system is the hallmark feature in multiple sclerosis (MS). The mechanism resulting in destabilization of myelin is a complex multi-faceted process, part of which involves deimination of myelin basic protein (MBP). Deimination, the conversion of protein-bound arginine to citrulline, is mediated by the peptidylarginine deiminase (PAD) family of enzymes, of which the PAD2 and PAD4 isoforms are present in myelin. To test the hypothesis that PAD contributes to destabilization of myelin in MS, we developed a transgenic mouse line (PD2) containing multiple copies of the cDNA encoding PAD2, under the control of the MBP promoter. Using previously established criteria, clinical signs were more severe in PD2 mice than in their normal littermates. The increase in PAD2 expression and activity in white matter was demonstrated by immunohistochemistry, reverse transcriptase-PCR, enzyme activity assays, and increased deimination of MBP. Light and electron microscopy revealed more severe focal demyelination and thinner myelin in the PD2 homozygous mice compared with heterozygous PD2 mice. Quantitation of the disease-associated molecules GFAP and CD68, as measured by immunoslot blots, were indicative of astrocytosis and macrophage activation. Concurrently, elevated levels of the pro-inflammatory cytokine TNF-alpha and nuclear histone deimination support initiation of demyelination by increased PAD activity. These data support the hypothesis that elevated PAD levels in white matter represents an early change that precedes demyelination.


Subject(s)
Central Nervous System/pathology , Hydrolases/metabolism , Myelin Sheath/pathology , Animals , Hydrolases/genetics , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Phenotype , Protein-Arginine Deiminases , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...