Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.788
Filter
1.
Rev Invest Clin ; 76(2): 65-79, 2024.
Article in English | MEDLINE | ID: mdl-38718804

ABSTRACT

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).


Subject(s)
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
3.
Biomolecules ; 14(4)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38672413

ABSTRACT

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.


Subject(s)
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
4.
Front Immunol ; 15: 1378202, 2024.
Article in English | MEDLINE | ID: mdl-38650945

ABSTRACT

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.


Subject(s)
Adipocytes , Energy Metabolism , Homeostasis , Macrophages , Obesity , Humans , Macrophages/metabolism , Macrophages/immunology , Animals , Adipocytes/metabolism , Obesity/metabolism , Obesity/immunology , Cell Communication , Thermogenesis
5.
Trends Parasitol ; 40(5): 386-400, 2024 May.
Article in English | MEDLINE | ID: mdl-38609741

ABSTRACT

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.


Subject(s)
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
6.
Acta Med Okayama ; 78(2): 185-191, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38688836

ABSTRACT

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.


Subject(s)
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.
Eur J Immunol ; 54(5): e2350669, 2024 May.
Article in English | MEDLINE | ID: mdl-38339772

ABSTRACT

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.


Subject(s)
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
8.
Autoimmun Rev ; 23(3): 103502, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38101692

ABSTRACT

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.


Subject(s)
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
9.
Science ; 381(6662): 1092-1098, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37676935

ABSTRACT

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.


Subject(s)
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
10.
Front Immunol ; 14: 1153915, 2023.
Article in English | MEDLINE | ID: mdl-37153549

ABSTRACT

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.


Subject(s)
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
11.
Science ; 379(6639): 1298-1300, 2023 03 31.
Article in English | MEDLINE | ID: mdl-36996218

ABSTRACT

Overnutrition could lead to loss of self-tolerance by impinging on immune regulation.


Subject(s)
Autoimmune Diseases , Autoimmunity , Obesity , Overnutrition , Self Tolerance , Humans , Obesity/complications , Obesity/immunology , Overnutrition/complications , Overnutrition/immunology , Autoimmune Diseases/etiology
12.
Science ; 379(6627): 28-29, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36603093
13.
Eur J Immunol ; 53(2): e2249990, 2023 02.
Article in English | MEDLINE | ID: mdl-36433684

ABSTRACT

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.


Subject(s)
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
14.
Arch Gerontol Geriatr ; 105: 104856, 2023 02.
Article in English | MEDLINE | ID: mdl-36399890

ABSTRACT

Osteosarcopenic obesity (OSO) has been associated with increase immobility, falls, fractures, and other dysfunctions, which could increase mortality risk during aging. However, its etiology remains unknown. Recent studies revealed that sedentarism, fat gain, and epigenetic regulators are critical in its development. One effective intervention to prevent and treat OSO is exercise. Therefore, in the present study, by keeping rats in conditions of sedentarism and others under a low-intensity exercise routine, we established an experimental model of OSO. We determined the degree of sarcopenia, obesity, and osteopenia at different ages and analyzed the miRNA expression during the lifespan using miRNA microarrays from gastrocnemius muscle. Interestingly microarrays results showed that there is a set of miRNAs that changed their expression with exercise. The pathway enrichment analysis showed that these miRNAs are strongly associated with immune regulation. Further inflammatory profiles with IL-6/IL-10 and TNF-α/IL-10 ratios showed that exercised rats presented a lower pro-inflammatory profile than sedentary rats. Also, the body fat gain in the sedentary group increased the inflammatory profile, ultimately leading to muscle dysfunction. Exercise prevented strength loss over time and maintained skeletal muscle functionality over time. Differential expression of miRNAs suggests that they might participate in this process by regulating the inflammatory response associated with aging, thus preventing the development of OSO.


Subject(s)
Aging , Bone Diseases, Metabolic , Immunity , MicroRNAs , Obesity , Physical Conditioning, Animal , Sarcopenia , Animals , Rats , Interleukin-10/genetics , Interleukin-10/metabolism , MicroRNAs/metabolism , Obesity/immunology , Obesity/prevention & control , Sarcopenia/immunology , Sarcopenia/prevention & control , Bone Diseases, Metabolic/immunology , Bone Diseases, Metabolic/prevention & control , Muscle, Skeletal/metabolism , Inflammation/immunology , Inflammation/prevention & control , Sedentary Behavior , Disease Models, Animal , Cytokines/genetics , Cytokines/metabolism
15.
Science ; 378(6619): 485, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36378990
16.
Nat Commun ; 13(1): 5208, 2022 09 05.
Article in English | MEDLINE | ID: mdl-36064857

ABSTRACT

Adipose tissue macrophage (ATM) inflammation is involved with meta-inflammation and pathology of metabolic complications. Here we report that in adipocytes, elevated lactate production, previously regarded as the waste product of glycolysis, serves as a danger signal to promote ATM polarization to an inflammatory state in the context of obesity. Adipocyte-selective deletion of lactate dehydrogenase A (Ldha), the enzyme converting pyruvate to lactate, protects mice from obesity-associated glucose intolerance and insulin resistance, accompanied by a lower percentage of inflammatory ATM and reduced production of pro-inflammatory cytokines such as interleukin 1ß (IL-1ß). Mechanistically, lactate, at its physiological concentration, fosters the activation of inflammatory macrophages by directly binding to the catalytic domain of prolyl hydroxylase domain-containing 2 (PHD2) in a competitive manner with α-ketoglutarate and stabilizes hypoxia inducible factor (HIF-1α). Lactate-induced IL-1ß was abolished in PHD2-deficient macrophages. Human adipose lactate level is positively linked with local inflammatory features and insulin resistance index independent of the body mass index (BMI). Our study shows a critical function of adipocyte-derived lactate in promoting the pro-inflammatory microenvironment in adipose and identifies PHD2 as a direct sensor of lactate, which functions to connect chronic inflammation and energy metabolism.


Subject(s)
Adipocytes , Hypoxia-Inducible Factor-Proline Dioxygenases , Inflammation , Lactate Dehydrogenase 5 , Lactic Acid , Macrophages , Adipocytes/immunology , Adipose Tissue/immunology , Animals , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Insulin Resistance/genetics , Insulin Resistance/immunology , Insulin Resistance/physiology , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/immunology , Lactate Dehydrogenase 5/genetics , Lactate Dehydrogenase 5/immunology , Lactic Acid/immunology , Macrophages/immunology , Mice , Obesity/genetics , Obesity/immunology , Obesity/pathology , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/immunology , Prolyl Hydroxylases
17.
Int J Mol Sci ; 23(5)2022 Mar 06.
Article in English | MEDLINE | ID: mdl-35270015

ABSTRACT

Almost two years have passed since the outbreak reported for the first time in Wuhan of coronavirus disease 2019 (COVID-19), due to severe acute respiratory syndrome (SARS)-CoV-2 coronavirus, rapidly evolved into a pandemic. This infectious disease has stressed global health care systems. The mortality rate is higher, particularly in elderly population and in patients with comorbidities such as hypertension, diabetes mellitus, cardiovascular disease, chronic lung disease, chronic renal disease, and malignancy. Among them, subjects with diabetes have a high risk of developing severe form of COVID-19 and show increased mortality. How diabetes contributes to COVID-19 severity remains unclear. It has been hypothesized that it may be correlated with the effects of hyperglycemia on systemic inflammatory responses and immune system dysfunction. Vitamin D (VD) is a modulator of immune-response. Data from literature showed that vitamin D deficiency in COVID-19 patients increases COVID-19 severity, likely because of its negative impact on immune and inflammatory responses. Therefore, the use of vitamin D might play a role in some aspects of the infection, particularly the inflammatory state and the immune system function of patients. Moreover, a piece of evidence highlighted a link among vitamin D deficiency, obesity and diabetes, all factors associated with COVID-19 severity. Given this background, we performed an overview of the systematic reviews to assess the association between vitamin D supplementation and inflammatory markers in patients with diabetes; furthermore, vitamin D's possible role in COVID-19 patients was assessed as well. Three databases, namely MEDLINE, PubMed Central and the Cochrane Library of Systematic Reviews, were reviewed to retrieve the pertinent data. The aim of this review is to provide insight into the recent advances about the molecular basis of the relationship between vitamin D, immune response, inflammation, diabetes and COVID-19.


Subject(s)
COVID-19/immunology , Diabetes Mellitus/immunology , Immune System/immunology , Inflammation/immunology , Obesity/immunology , Vitamin D/immunology , COVID-19/virology , Humans , Immune System/drug effects , Meta-Analysis as Topic , SARS-CoV-2/physiology , Systematic Reviews as Topic , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Vitamin D/administration & dosage , Vitamins/administration & dosage , Vitamins/immunology
18.
Front Immunol ; 13: 814203, 2022.
Article in English | MEDLINE | ID: mdl-35145521

ABSTRACT

T cells and B cells have been identified in human and murine islets, but the phenotype and role of islet lymphocytes is unknown. Resident immune populations set the stage for responses to inflammation in the islets during homeostasis and diabetes. Thus, we sought to identify the phenotype and effector function of islet lymphocytes to better understand their role in normal islets and in islets under metabolic stress. Lymphocytes were located in the islet parenchyma, and were comprised of a mix of naïve, activated, and memory T cell and B cell subsets, with an enrichment for regulatory B cell subsets. Use of a Nur77 reporter indicated that CD8 T cells and B cells both received local antigen stimulus, indicating that they responded to antigens present in the islets. Analysis of effector function showed that islet T cells and B cells produced the regulatory cytokine IL-10. The regulatory phenotype of islet T cells and B cells and their response to local antigenic stimuli remained stable under conditions of metabolic stress in the diet induced obesity (DIO) model. T cells present in human islets retained a similar activated and memory phenotype in non-diabetic and T2D donors. Under steady-state conditions, islet T cells and B cells have a regulatory phenotype, and thus may play a protective role in maintaining tissue homeostasis.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Homeostasis/physiology , Islets of Langerhans/immunology , Stress, Physiological/physiology , T-Lymphocytes/immunology , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Obesity/immunology , Phenotype
19.
Physiol Rep ; 10(4): e15148, 2022 02.
Article in English | MEDLINE | ID: mdl-35179822

ABSTRACT

AIM: To reexamine the associations of NK cell number and function in the peripheral blood with overweight/obesity and the metabolic syndrome in a large, well-phenotyped human cohort. METHODS: Cross-sectional analysis of 273 women in the PPSDiab Study; measurement of absolute and relative number of NK cells in peripheral blood, and of functional parameters CD69 positivity and cytotoxicity against K562 cells; group comparison of NK cell characteristics between lean, overweight, and obese participants, as well as metabolic syndrome scores of 0, 1, 2, and ≥3; Spearman correlation analyses to clinical parameters related to the metabolic syndrome. RESULTS: We found no differences in NK cell number and function between lean, overweight, and obese women (relative NK cell number (median (Q1-Q3), [%]) 5.1(2.6-9.4) vs. 4.8 (2.9-8.4) vs. 3.8 (1.7-7.8), p = 0.187; absolute NK cell number [106 /L]: 86.9 (44.6-188.8) vs. 92.6 (52.5-154.6) vs. 85.9 (44-153.8), p = 0.632; CD69+ [%]: 27.2 (12.9-44.3) vs. 37.6 (13.2-52.8) vs. 33.6 (16.3-45), p = 0.136; cytotoxicity [%]: 11.0 (7.1-14.5) vs. 8.5 (6.4-13.2) vs. 11.3 (8.7-14.2), p = 0.094), as well as between different metabolic syndrome scores. Nonesterified fatty acids correlated with absolute and relative NK cell number and cytotoxicity (ρ [p-value]: 0.142 [0.021], 0.119 [0.049], and 0.131 [0.035], respectively). Relative NK cell number further correlated with high-density lipoprotein cholesterol (0.144 [0.018]) and cytotoxicity with 2 h glucose in oral glucose tolerance testing (0.132 [0.034]). CD69 positivity correlated with body fat (0.141 [0.021]), triglycerides (0.129 [0.033]), and plasma leptin (0.155 [0.010]). After correction for multiple testing, none of the associations remained significant. CONCLUSION: In the present study, we observed no associations of NK cell number and function in the peripheral blood with overweight/obesity and the metabolic syndrome. Extreme phenotypes of obesity and the metabolic syndrome might have caused differing results in previous studies. Further analyses with a focus on compartments other than peripheral blood may help to clarify the relation between NK cells and metabolic diseases.


Subject(s)
Killer Cells, Natural/immunology , Metabolic Syndrome/blood , Obesity/blood , Adult , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cytotoxicity, Immunologic , Female , Humans , Lectins, C-Type/metabolism , Metabolic Syndrome/epidemiology , Metabolic Syndrome/immunology , Middle Aged , Obesity/epidemiology , Obesity/immunology
20.
Gut Microbes ; 14(1): 2031696, 2022.
Article in English | MEDLINE | ID: mdl-35130127

ABSTRACT

Obesity and type 2 diabetes (T2D) are growing burdens for individuals and the health-care system. Bariatric surgery is an efficient, but drastic treatment to reduce body weight, normalize glucose values, and reduce low-grade inflammation. The gut microbiome, which is in part controlled by intestinal antibodies, such as IgA, is involved in the development of both conditions. Knowledge of the effect of bariatric surgery on systemic and intestinal antibody response is limited. Here, we determined the fecal antibody and gut microbiome response in 40 T2D and non-diabetic (ND) obese individuals that underwent bariatric surgery (N = 40). Body weight, fasting glucose concentrations and inflammatory parameters decreased after bariatric surgery, whereas pro-inflammatory bacterial species such as lipopolysaccharide (LPS), and flagellin increased in the feces. Simultaneously, concentrations of LPS- and flagellin-specific intestinal IgA levels increased with the majority of pro-inflammatory bacteria coated with IgA after surgery. Finally, serum antibodies decreased in both groups, along with a lower inflammatory tone. We conclude that intestinal rearrangement by bariatric surgery leads to expansion of typical pro-inflammatory bacteria, which may be compensated by an improved antibody response. Although further evidence and mechanistic insights are needed, we postulate that this apparent compensatory antibody response might help to reduce systemic inflammation by neutralizing intestinal immunogenic components and thereby enhance intestinal barrier function after bariatric surgery.


Subject(s)
Antibodies, Bacterial/blood , Bacteria/immunology , Diabetes Mellitus, Type 2/immunology , Gastrointestinal Microbiome , Intestines/microbiology , Obesity/immunology , Antibodies, Bacterial/immunology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bariatric Surgery , Cohort Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/surgery , Feces/chemistry , Feces/microbiology , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Intestines/immunology , Obesity/blood , Obesity/microbiology , Obesity/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...