Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.795
1.
Microbiome ; 12(1): 103, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38845049

BACKGROUND: The metabolic disturbances of obesity can be mitigated by strategies modulating the gut microbiota. In this study, we sought to identify whether innate or adaptive immunity mediates the beneficial metabolic effects of the human intestinal bacterium Bacteroides uniformis CECT 7771 in obesity. METHODS: We evaluated the effects of orally administered B. uniformis on energy homeostasis, intestinal immunity, hormone levels, and gut microbiota in wild-type and Rag1-deficient mice with diet-induced obesity. We also assessed whether B. uniformis needed to be viable to exert its beneficial effects in obesity and to directly induce immunoregulatory effects. RESULTS: The administration of B. uniformis to obese mice improved glucose tolerance and insulin secretion, restored the caloric intake suppression after an oral glucose challenge, and reduced hyperglycemia. The pre- and post-prandial glucose-related benefits were associated with restoration of the anti-inflammatory tone mediated by type 2 macrophages and regulatory T cells (Tregs) in the lamina propria of the small intestine. Contrastingly, B. uniformis administration failed to improve glucose tolerance in obese Rag1-/- mice, but prevented the increased body weight gain and adiposity. Overall, the beneficial effects seemed to be independent of enteroendocrine effects and of major changes in gut microbiota composition. B. uniformis directly induced Tregs generation from naïve CD4+ T cells in vitro and was not required to be viable to improve glucose homeostasis but its viability was necessary to prevent body weight gain in diet-induced obese wild-type mice. CONCLUSIONS: Here we demonstrate that B. uniformis modulates the energy homeostasis in diet-induced obese mice through different mechanisms. The bacterium improves oral glucose tolerance by adaptive immunity-dependent mechanisms that do not require cell viability and prevents body weight gain by adaptive immunity-independent mechanisms which require cell viability. Video Abstract.


Adaptive Immunity , Bacteroides , Gastrointestinal Microbiome , Obesity , Weight Gain , Animals , Mice , Obesity/immunology , Obesity/microbiology , Diet, High-Fat/adverse effects , Mice, Obese , T-Lymphocytes, Regulatory/immunology , Mice, Inbred C57BL , Male , Humans , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Probiotics/administration & dosage , Mice, Knockout , Glucose/metabolism
3.
Nat Commun ; 15(1): 4232, 2024 May 18.
Article En | MEDLINE | ID: mdl-38762479

Toll-like receptor 9 (TLR9) recognizes bacterial, viral and self DNA and play an important role in immunity and inflammation. However, the role of TLR9 in obesity is less well-studied. Here, we generate B-cell-specific Tlr9-deficient (Tlr9fl/fl/Cd19Cre+/-, KO) B6 mice and model obesity using a high-fat diet. Compared with control mice, B-cell-specific-Tlr9-deficient mice exhibited increased fat tissue inflammation, weight gain, and impaired glucose and insulin tolerance. Furthermore, the frequencies of IL-10-producing-B cells and marginal zone B cells were reduced, and those of follicular and germinal center B cells were increased. This was associated with increased frequencies of IFNγ-producing-T cells and increased follicular helper cells. In addition, gut microbiota from the KO mice induced a pro-inflammatory state leading to immunological and metabolic dysregulation when transferred to germ-free mice. Using 16 S rRNA gene sequencing, we identify altered gut microbial communities including reduced Lachnospiraceae, which may play a role in altered metabolism in KO mice. We identify an important network involving Tlr9, Irf4 and Il-10 interconnecting metabolic homeostasis, with the function of B and T cells, and gut microbiota in obesity.


B-Lymphocytes , Diet, High-Fat , Dysbiosis , Gastrointestinal Microbiome , Inflammation , Interleukin-10 , Mice, Knockout , Obesity , Toll-Like Receptor 9 , Animals , Obesity/immunology , Obesity/microbiology , Obesity/metabolism , Dysbiosis/immunology , Dysbiosis/microbiology , Toll-Like Receptor 9/metabolism , Toll-Like Receptor 9/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Inflammation/metabolism , Mice , Diet, High-Fat/adverse effects , Interleukin-10/metabolism , Male , Mice, Inbred C57BL , Disease Models, Animal , Interferon Regulatory Factors
4.
Front Immunol ; 15: 1358341, 2024.
Article En | MEDLINE | ID: mdl-38807605

Background: Higher prevalence of obesity has been observed among women compared to men, which can be explained partly by the higher consumption of sweets and physical inactivity. Obesity can alter immune cell infiltration, and therefore increase the susceptibility to develop chronic inflammation and metabolic disorders. In this study, we aimed to explore the association between free sugar intake and other unhealthy lifestyle habits in relation to the proportion of circulating iNKT cells among women with healthy weight and women experiencing overweight and obesity. Methods: A cross-sectional study was conducted on 51 Saudi women > 18 years, wherein their daily free sugar intake was assessed using the validated Food Frequency Questionnaire. Data on smoking status, physical activity, and supplement use were also collected. Anthropometric data including height, weight, waist circumference were objectively measured from each participants. The proportion of circulating iNKT cells was determined using flow cytometry. Results: Smoking, physical activity, supplement use, and weight status were not associated with proportion of circulating iNKT cells. Significant association was found between proportion of circulating iNKT cells and total free sugar intake and free sugar intake coming from solid food sources only among women experiencing overweight and obesity (Beta: -0.10: Standard Error: 0.04 [95% Confidence Interval: -0.18 to -0.01], p= 0.034) and (Beta: -0.15: Standard Error: 0.05 [95% Confidence Interval: -0.25 to -0.05], p= 0.005), respectively. Conclusion: Excessive free sugar consumption may alter iNKT cells and consequently increase the risk for chronic inflammation and metabolic disorders.


Natural Killer T-Cells , Obesity , Overweight , Humans , Female , Obesity/immunology , Obesity/blood , Adult , Natural Killer T-Cells/immunology , Cross-Sectional Studies , Overweight/immunology , Middle Aged , Dietary Sugars/adverse effects , Dietary Sugars/administration & dosage , Young Adult
5.
Rev Invest Clin ; 76(2): 65-79, 2024 02 15.
Article En | MEDLINE | ID: mdl-38718804

UNASSIGNED: Excess body weight has become a global epidemic and a significant risk factor for developing chronic diseases, which are the leading causes of worldwide morbidities. Adipose tissue (AT), primarily composed of adipocytes, stores substantial amounts of energy and plays a crucial role in maintaining whole-body glucose and lipid metabolism. This helps prevent excessive body fat accumulation and lipotoxicity in peripheral tissues. In addition, AT contains endothelial cells and a substantial population of immune cells (constituting 60-70% of non-adipocyte cells), including macrophages, T and B lymphocytes, and natural killer cells. These resident immune cells engage in crosstalk with adipocytes, contributing to the maintenance of metabolic and immune homeostasis in AT. An exacerbated inflammatory response or inadequate immune resolution can lead to chronic systemic low-grade inflammation, triggering the development of metabolic alterations and the onset of chronic diseases. This review aims to elucidate the regulatory mechanisms through which immune cells influence AT function and energy homeostasis. We also focus on the interactions and functional dynamics of immune cell populations, highlighting their role in maintaining the delicate balance between metabolic health and obesity-related inflammation. Finally, understanding immunometabolism is crucial for unraveling the pathogenesis of metabolic diseases and developing targeted immunotherapeutic strategies. These strategies may offer innovative avenues in the rapidly evolving field of immunometabolism. (Rev Invest Clin. 2024;76(2):65-79).


Adipose Tissue , Inflammation , Metabolic Diseases , Obesity , Humans , Adipose Tissue/metabolism , Adipose Tissue/immunology , Obesity/immunology , Obesity/metabolism , Inflammation/immunology , Inflammation/metabolism , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Metabolic Diseases/etiology , Energy Metabolism/physiology , Adipocytes/metabolism , Adipocytes/immunology , Lipid Metabolism/physiology , Animals , Homeostasis
6.
Acta Med Okayama ; 78(2): 185-191, 2024 Apr.
Article En | MEDLINE | ID: mdl-38688836

The global pandemic of coronavirus infection 2019 (COVID-19) was an unprecedented public health emergency. Several clinical studies reported that heart disease, lung disease, diabetes, hypertension, dyslipidemia, and obesity are critical risk factors for increased severity of and hospitalization for COVID-19. This is largely because patients with these underlying medical conditions can show poor immune responses to the COVID-19 vaccinations. Diabetes is one of the underlying conditions most highly associated with COVID-19 susceptibility and is considered a predictor of poor prognosis of COVID-19. We therefore investigated factors that influence the anti-SARS-CoV-2 spike IgG antibody titer after three doses of vaccination in patients with type 2 diabetes. We found that obesity was associated with low anti-SARS-CoV-2 spike IgG antibody titers following three-dose vaccination in type 2 diabetics. Obese patients with type 2 diabetes may have attenuated vaccine efficacy and require additional vaccination; continuous infection control should be considered in such patients.


COVID-19 Vaccines , COVID-19 , Diabetes Mellitus, Type 2 , Obesity , SARS-CoV-2 , Humans , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/complications , Obesity/immunology , Obesity/complications , COVID-19 Vaccines/immunology , Cross-Sectional Studies , COVID-19/immunology , COVID-19/prevention & control , COVID-19/complications , Male , Female , Middle Aged , Aged , SARS-CoV-2/immunology , Antibodies, Viral/blood , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunogenicity, Vaccine
7.
Trends Parasitol ; 40(5): 386-400, 2024 May.
Article En | MEDLINE | ID: mdl-38609741

Obesity is a worldwide pandemic and major risk factor for the development of metabolic syndrome (MetS) and type 2 diabetes (T2D). T2D requires lifelong medical support to limit complications and is defined by impaired glucose tolerance, insulin resistance (IR), and chronic low-level systemic inflammation initiating from adipose tissue. The current preventative strategies include a healthy diet, controlled physical activity, and medication targeting hyperglycemia, with underexplored underlying inflammation. Studies suggest a protective role for helminth infection in the prevention of T2D. The mechanisms may involve induction of modified type 2 and regulatory immune responses that suppress inflammation and promote insulin sensitivity. In this review, the roles of helminths in counteracting MetS, and prospects for harnessing these protective mechanisms for the development of novel anti-diabetes drugs are discussed.


Diabetes Mellitus, Type 2 , Helminths , Metabolic Syndrome , Animals , Humans , Helminths/immunology , Helminths/physiology , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Metabolic Syndrome/parasitology , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Helminthiasis/immunology , Helminthiasis/parasitology , Obesity/immunology , Obesity/metabolism , Host-Parasite Interactions/immunology , Insulin Resistance
8.
Biomolecules ; 14(4)2024 Mar 25.
Article En | MEDLINE | ID: mdl-38672413

Individuals who are overweight or obese are at increased risk of developing prediabetes and type 2 diabetes, yet the direct molecular mechanisms that connect diabetes to obesity are not clear. Chronic, sustained inflammation is considered a strong risk factor in these interactions, directed in part by the short-lived gene expression programs encoding for cytokines and pro-inflammatory mediators. In this study, we show that triptolide administration in the C57BL/6 diet-induced obese mice at up to 10 µg/kg/day for 10 weeks attenuated the development of insulin resistance and diabetes, but not obesity, in these animals. Significant reductions in adipose tissue inflammation and improved insulin sensitivity were observed in the absence of changes in food intake, body weight, body composition, or energy expenditure. Analysis of the core cluster of biomarkers that drives pro-inflammatory responses in the metabolic tissues suggested TNF-α as a critical point that affected the co-development of inflammation and insulin resistance, but also pointed to the putatively protective roles of increased COX-2 and IL-17A signaling in the mediation of these pathophysiological states. Our results show that reduction of diet-induced inflammation confers partial protection against insulin resistance, but not obesity, and suggest the possibility of achieving overweight phenotypes that are accompanied by minimal insulin resistance if inflammation is controlled.


Diterpenes , Epoxy Compounds , Insulin Resistance , Mice, Inbred C57BL , Obesity , Phenanthrenes , Animals , Epoxy Compounds/pharmacology , Epoxy Compounds/administration & dosage , Diterpenes/pharmacology , Diterpenes/administration & dosage , Phenanthrenes/pharmacology , Phenanthrenes/administration & dosage , Obesity/metabolism , Obesity/immunology , Mice , Male , Inflammation/metabolism , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Interleukin-17/metabolism , Interleukin-17/genetics , Diet, High-Fat/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/immunology , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Energy Metabolism/drug effects
9.
Front Immunol ; 15: 1378202, 2024.
Article En | MEDLINE | ID: mdl-38650945

Interactions between macrophages and adipocytes in adipose tissue are critical for the regulation of energy metabolism and obesity. Macrophage polarization induced by cold or other stimulations can drive metabolic reprogramming of adipocytes, browning, and thermogenesis. Accordingly, investigating the roles of macrophages and adipocytes in the maintenance of energy homeostasis is critical for the development of novel therapeutic approaches specifically targeting macrophages in metabolic disorders such as obesity. Current review outlines macrophage polarization not only regulates the release of central nervous system and inflammatory factors, but controls mitochondrial function, and other factor that induce metabolic reprogramming of adipocytes and maintain energy homeostasis. We also emphasized on how the adipocytes conversely motivate the polarization of macrophage. Exploring the interactions between adipocytes and macrophages may provide new therapeutic strategies for the management of obesity-related metabolic diseases.


Adipocytes , Energy Metabolism , Homeostasis , Macrophages , Obesity , Humans , Macrophages/metabolism , Macrophages/immunology , Animals , Adipocytes/metabolism , Obesity/metabolism , Obesity/immunology , Cell Communication , Thermogenesis
10.
Nat Microbiol ; 9(6): 1593-1606, 2024 Jun.
Article En | MEDLINE | ID: mdl-38637722

Metabolic disease is epidemiologically linked to severe complications upon influenza virus infection, thus vaccination is a priority in this high-risk population. Yet, vaccine responses are less effective in these same hosts. Here we examined how the timing of diet switching from a high-fat diet to a control diet affected influenza vaccine efficacy in diet-induced obese mice. Our results demonstrate that the systemic meta-inflammation generated by high-fat diet exposure limited T cell maturation to the memory compartment at the time of vaccination, impacting the recall of effector memory T cells upon viral challenge. This was not improved with a diet switch post-vaccination. However, the metabolic dysfunction of T cells was reversed if weight loss occurred 4 weeks before vaccination, restoring a functional recall response. This corresponded with changes in the systemic obesity-related biomarkers leptin and adiponectin, highlighting the systemic and specific effects of diet on influenza vaccine immunogenicity.


Diet, High-Fat , Influenza Vaccines , Obesity , Orthomyxoviridae Infections , Animals , Mice , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Diet, High-Fat/adverse effects , Obesity/immunology , Obesity/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Mice, Inbred C57BL , Vaccination , Mice, Obese , Leptin/metabolism , Male , Female , Adiponectin/metabolism , T-Lymphocytes/immunology
11.
J Reprod Immunol ; 163: 104234, 2024 Jun.
Article En | MEDLINE | ID: mdl-38479054

Combined oral contraceptives (COCs) are known to cause weight gain and alter metabolic and immunological pathways. However, modifications in arterial or venous thrombotic risk profiles of women of reproductive ages on COC remain unclear. The study aimed at assessing the impact of COC on immune activation in diet-induced obesity. We further established whether the dietary intervention of switching from a high-fat diet (HFD) to a low-fat diet (LFD) attenuates immunological responses. Twenty (n=20) five-week-old female Sprague Dawley rats were randomly divided into two diet groups of HFD (n=15) and LFD (n=5) and were monitored for eight weeks. After eight weeks, animals in the HFD group switched diets to LFD and were randomly assigned to receive high-dose COC (HCOC) or low-dose COC (LCOC) for six weeks. Animals on HFD significantly gained weight and had a higher lee index when compared to the LFD group (p < 0.05). Moreover, the triglyceride-glucose index, insulin, and other metabolic parameters also increased in the HFD group compared to the LFD group (p < 0.001). Consistently, the levels of interleukin (IL)-6 and tumor necrosis factor-alpha (TNF-α), were elevated in the HFD group when compared to the LFD group (p < 0.05). Upon switching from a high-fat to a low-fat diet, insulin levels persistently increased in animals receiving HCOC treatment compared to the LFD and HFD/LFD groups (p < 0.05). Thus, in a rat model of HFD-feeding, short-term HCOC treatment induces long-term metabolic dysregulation, which persists despite dietary intervention. However, further studies are recommended to confirm these findings.


Diet, Fat-Restricted , Diet, High-Fat , Hyperinsulinism , Obesity , Rats, Sprague-Dawley , Animals , Female , Obesity/immunology , Rats , Diet, High-Fat/adverse effects , Hyperinsulinism/immunology , Hyperinsulinism/chemically induced , Humans , Insulin/blood , Insulin/metabolism , Contraceptives, Oral, Combined/administration & dosage , Contraceptives, Oral, Combined/adverse effects , Interleukin-6/metabolism , Interleukin-6/blood
12.
Eur J Immunol ; 54(5): e2350669, 2024 May.
Article En | MEDLINE | ID: mdl-38339772

The importance of macrophages in adipose tissue (AT) homeostasis and inflammation is well established. However, the potential cues that regulate their function remain incompletely understood. To bridge this important gap, we sought to characterize novel pathways involved using a mouse model of diet-induced obesity. By performing transcriptomics analysis of AT macrophages (ATMs), we found that late-stage ATMs from high-fat diet mice presented with perturbed Notch signaling accompanied by robust proinflammatory and metabolic changes. To explore the hypothesis that the deregulated Notch pathway contributes to the development of AT inflammation and diet-induced obesity, we employed a genetic approach to abrogate myeloid Notch1 and Notch2 receptors. Our results revealed that the combined loss of Notch1 and Notch2 worsened obesity-related metabolic dysregulation. Body and AT weight gain was higher, blood glucose levels increased and metabolic parameters were substantially worsened in deficient mice fed high-fat diet. Moreover, serum insulin and leptin were elevated as were triglycerides. Molecular analysis of ATMs showed that deletion of Notch receptors escalated inflammation through the induction of an M1-like pro-inflammatory phenotype. Our findings thus support a protective role of myeloid Notch signaling in adipose tissue inflammation and metabolic dysregulation.


Adipose Tissue , Diet, High-Fat , Inflammation , Macrophages , Obesity , Receptor, Notch1 , Receptor, Notch2 , Signal Transduction , Animals , Macrophages/immunology , Macrophages/metabolism , Adipose Tissue/metabolism , Adipose Tissue/immunology , Mice , Diet, High-Fat/adverse effects , Inflammation/immunology , Inflammation/metabolism , Signal Transduction/immunology , Obesity/metabolism , Obesity/immunology , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Receptor, Notch2/metabolism , Receptor, Notch2/genetics , Mice, Knockout , Mice, Inbred C57BL , Male
13.
Clin Exp Immunol ; 216(3): 280-292, 2024 May 16.
Article En | MEDLINE | ID: mdl-38334487

Gestational diabetes mellitus (GDM) is a frequent and serious complication of pregnancy, often associated with obesity. Metabolic dysfunction and metainflammation are evident in both obesity and GDM. In this cross-sectional study, we aimed at defining the direct contribution of the immune system in GDM, across the main metabolic tissues, specifically focussing on elucidating the roles of obesity and GDM to the clinical outcome. Using immunoassays and multicolour flow cytometry, cytokine profiles and immune cell frequencies were measured in maternal circulation and central metabolic tissues [placenta and visceral adipose tissue (VAT)] in GDM-diagnosed (n = 28) and normal glucose tolerant (n = 32) women undergoing caesarean section. Participants were sub-grouped as non-obese [body mass index (BMI) < 30 kg/m2] or obese (BMI ≥ 30 kg/m2). Unsupervised data analysis was performed on the flow cytometry data set to identify functional alterations. GDM obese participants had significantly elevated circulating IL-6 and IL-17A levels. GDM non-obese participants had elevated circulating IL-12p70, elevated placental IL-17A, and VAT IFN-γ production. Unsupervised clustering of immune populations across the three biological sites simultaneously, identified different NK- and T-cell phenotypes that were altered in NGT obese and GDM non-obese participants, while a classical tissue monocyte cluster was increased in GDM obese participants. In this study, there was significant evidence of subclinical inflammation, and significant alterations in clusters of NK cells, T cells, and tissue monocyte populations in GDM. While increased adiposity assimilates with increased inflammation in the non-pregnant state, this overt relationship may not be as evident during pregnancy and warrants further examination in future longitudinal studies.


Diabetes, Gestational , Inflammation , Obesity , Humans , Female , Pregnancy , Diabetes, Gestational/immunology , Diabetes, Gestational/blood , Adult , Obesity/immunology , Inflammation/immunology , Cross-Sectional Studies , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Placenta/immunology , Placenta/metabolism , Killer Cells, Natural/immunology , Interleukin-17/blood , Cytokines/blood , Cytokines/metabolism , Interleukin-6/blood , Body Mass Index , T-Lymphocytes/immunology , Interferon-gamma/blood
14.
Front Med ; 18(2): 205-236, 2024 Apr.
Article En | MEDLINE | ID: mdl-38165533

Macrophages, a heterogeneous population of innate immune cells, exhibit remarkable plasticity and play pivotal roles in coordinating immune responses and maintaining tissue homeostasis within the context of metabolic diseases. The activation of inflammatory macrophages in obese adipose tissue leads to detrimental effects, inducing insulin resistance through increased inflammation, impaired thermogenesis, and adipose tissue fibrosis. Meanwhile, adipose tissue macrophages also play a beneficial role in maintaining adipose tissue homeostasis by regulating angiogenesis, facilitating the clearance of dead adipocytes, and promoting mitochondrial transfer. Exploring the heterogeneity of macrophages in obese adipose tissue is crucial for unraveling the pathogenesis of obesity and holds significant potential for targeted therapeutic interventions. Recently, the dual effects and some potential regulatory mechanisms of macrophages in adipose tissue have been elucidated using single-cell technology. In this review, we present a comprehensive overview of the intricate activation mechanisms and diverse functions of macrophages in adipose tissue during obesity, as well as explore the potential of drug delivery systems targeting macrophages, aiming to enhance the understanding of current regulatory mechanisms that may be potentially targeted for treating obesity or metabolic diseases.


Adipose Tissue , Macrophages , Obesity , Humans , Obesity/immunology , Macrophages/immunology , Macrophages/physiology , Adipose Tissue/immunology , Inflammation/immunology , Insulin Resistance , Animals , Thermogenesis/physiology , Adipocytes
15.
Autoimmun Rev ; 23(3): 103502, 2024 Mar.
Article En | MEDLINE | ID: mdl-38101692

Metabolic syndrome poses a great worldwide threat to the health of the patients. Increased visceral adiposity is recognized as the main determinant of the detrimental clinical effects of insulin resistance. Inflammation and immune system activation in the adipose tissue (AT) have a central role in the pathophysiology of metabolic syndrome, but the mechanisms linking increased adiposity to immunity in the AT remain in part elusive. In this review, we support the central role of adipocyte overload and relative adipose failure as key determinants in triggering immune aggression to AT. This provides a mechanistic explanation of the relative metabolic wellness of metabolically normal obese people and the disruption in insulin signaling in metabolically obese lean people.


Adipocytes , Adipose Tissue , Autoimmunity , Humans , Adipocytes/immunology , Adipocytes/metabolism , Autoimmunity/immunology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Obesity/immunology , Obesity/metabolism , Animals , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Insulin Resistance/immunology , Adiposity/immunology
16.
Science ; 381(6662): 1092-1098, 2023 09 08.
Article En | MEDLINE | ID: mdl-37676935

Dietary fiber improves metabolic health, but host-encoded mechanisms for digesting fibrous polysaccharides are unclear. In this work, we describe a mammalian adaptation to dietary chitin that is coordinated by gastric innate immune activation and acidic mammalian chitinase (AMCase). Chitin consumption causes gastric distension and cytokine production by stomach tuft cells and group 2 innate lymphoid cells (ILC2s) in mice, which drives the expansion of AMCase-expressing zymogenic chief cells that facilitate chitin digestion. Although chitin influences gut microbial composition, ILC2-mediated tissue adaptation and gastrointestinal responses are preserved in germ-free mice. In the absence of AMCase, sustained chitin intake leads to heightened basal type 2 immunity, reduced adiposity, and resistance to obesity. These data define an endogenous metabolic circuit that enables nutrient extraction from an insoluble dietary constituent by enhancing digestive function.


Adaptation, Physiological , Chitin , Chitinases , Dietary Fiber , Obesity , Stomach , Animals , Mice , Chitin/metabolism , Immunity, Innate , Lymphocytes/enzymology , Lymphocytes/immunology , Obesity/immunology , Stomach/immunology , Adaptation, Physiological/immunology , Chitinases/metabolism , Digestion/immunology
17.
Front Immunol ; 14: 1153915, 2023.
Article En | MEDLINE | ID: mdl-37153549

Macrophage infiltration into adipose tissue is a key pathological factor inducing adipose tissue dysfunction and contributing to obesity-induced inflammation and metabolic disorders. In this review, we aim to present the most recent research on macrophage heterogeneity in adipose tissue, with a focus on the molecular targets applied to macrophages as potential therapeutics for metabolic diseases. We begin by discussing the recruitment of macrophages and their roles in adipose tissue. While resident adipose tissue macrophages display an anti-inflammatory phenotype and promote the development of metabolically favorable beige adipose tissue, an increase in pro-inflammatory macrophages in adipose tissue has negative effects on adipose tissue function, including inhibition of adipogenesis, promotion of inflammation, insulin resistance, and fibrosis. Then, we presented the identities of the newly discovered adipose tissue macrophage subtypes (e.g. metabolically activated macrophages, CD9+ macrophages, lipid-associated macrophages, DARC+ macrophages, and MFehi macrophages), the majority of which are located in crown-like structures within adipose tissue during obesity. Finally, we discussed macrophage-targeting strategies to ameliorate obesity-related inflammation and metabolic abnormalities, with a focus on transcriptional factors such as PPARγ, KLF4, NFATc3, and HoxA5, which promote macrophage anti-inflammatory M2 polarization, as well as TLR4/NF-κB-mediated inflammatory pathways that activate pro-inflammatory M1 macrophages. In addition, a number of intracellular metabolic pathways closely associated with glucose metabolism, oxidative stress, nutrient sensing, and circadian clock regulation were examined. Understanding the complexities of macrophage plasticity and functionality may open up new avenues for the development of macrophage-based treatments for obesity and other metabolic diseases.


Adipose Tissue , Macrophages , Metabolic Diseases , Obesity , Adipose Tissue/immunology , Macrophages/classification , Macrophages/immunology , Obesity/immunology , Obesity/therapy , Metabolic Diseases/immunology , Metabolic Diseases/therapy , Humans , Inflammation/immunology , Inflammation/therapy , Adipogenesis/immunology , Cell Polarity
18.
Science ; 379(6639): 1298-1300, 2023 03 31.
Article En | MEDLINE | ID: mdl-36996218
20.
Eur J Immunol ; 53(2): e2249990, 2023 02.
Article En | MEDLINE | ID: mdl-36433684

Adipose tissue inflammation is a driving factor for the development of obesity-associated metabolic disturbances, and a role of adipose tissue T cells in initiating the pro-inflammatory signaling is emerging. However, data on human adipose tissue T cells in obesity are limited, reflected by the lack of phenotypic markers to define tissue-resident T cell subsets. In this study, we performed a deep characterization of T cells in blood and adipose tissue depots using multicolor flow cytometry and RNA sequencing. We identified distinct subsets of T cells associated with obesity expressing the activation markers, CD26 and CCR5, and obesity-specific genes that are potentially engaged in activating pro-inflammatory pathway, including ceramide signaling, autophagy, and IL-6 signaling. These findings increase our knowledge on the heterogeneity of T cells in adipose tissue and on subsets that may play a role in obesity-related pathogenesis.


Adipose Tissue , Inflammation , Insulin Resistance , Obesity , T-Lymphocyte Subsets , Humans , Adipose Tissue/immunology , Adipose Tissue/pathology , Autophagy/immunology , Ceramides/immunology , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Insulin Resistance/genetics , Insulin Resistance/immunology , Obesity/blood , Obesity/genetics , Obesity/immunology , Obesity/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology
...