Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.279
Filtrar
1.
Cell Biochem Funct ; 42(5): e4089, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38978329

RESUMEN

Adipose tissue in the obese state can lead to low-grade chronic inflammation while inducing or exacerbating obesity-related metabolic diseases and impairing overall health.T cells, which are essential immune cells similar to macrophages, are widely distributed in adipose tissue and perform their immunomodulatory function; they also cross-talk with other cells in the vascular stromal fraction. Based on a large number of studies, it has been found that N6 methyl adenine (m6A) is one of the most representative of epigenetic modifications, which affects the crosstalk between T cells, as well as other immune cells, in several ways and plays an important role in the development of adipose tissue inflammation and related metabolic diseases. In this review, we first provide an overview of the widespread presence of T cells in adipose tissue and summarize the key role of T cells in adipose tissue inflammation. Next, we explored the effects of m6A modifications on T cells in adipose tissue from the perspective of adipose tissue inflammation. Finally, we discuss the impact of m6a-regulated crosstalk between T cells and immune cells on the prospects for improving adipose tissue inflammation research, providing additional new ideas for the treatment of obesity.


Asunto(s)
Tejido Adiposo , Inflamación , Linfocitos T , Humanos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Tejido Adiposo/inmunología , Inflamación/metabolismo , Inflamación/patología , Inflamación/inmunología , Linfocitos T/metabolismo , Linfocitos T/inmunología , Animales , Obesidad/metabolismo , Obesidad/patología , Obesidad/inmunología , Epigénesis Genética , Adenosina/metabolismo
2.
Int J Mol Sci ; 25(13)2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38999932

RESUMEN

The World Health Organization (WHO) highlights a greater susceptibility of males to tuberculosis (TB), a vulnerability attributed to sex-specific variations in body fat and dietary factors. Our study delves into the unexplored terrain of how alterations in body fat influence Mycobacterium tuberculosis (Mtb) burden, lung pathology, immune responses, and gene expression, with a focus on sex-specific dynamics. Utilizing a low-dose Mtb-HN878 clinical strain infection model, we employ transgenic FAT-ATTAC mice with modulable body fat to explore the impact of fat loss (via fat ablation) and fat gain (via a medium-fat diet, MFD). Firstly, our investigation unveils that Mtb infection triggers severe pulmonary pathology in males, marked by shifts in metabolic signaling involving heightened lipid hydrolysis and proinflammatory signaling driven by IL-6 and localized pro-inflammatory CD8+ cells. This stands in stark contrast to females on a control regular diet (RD). Secondly, our findings indicate that both fat loss and fat gain in males lead to significantly elevated (1.6-fold (p ≤ 0.01) and 1.7-fold (p ≤ 0.001), respectively) Mtb burden in the lungs compared to females during Mtb infection (where fat loss and gain did not alter Mtb load in the lungs). This upsurge is associated with impaired lung lipid metabolism and intensified mitochondrial oxidative phosphorylation-regulated activity in lung CD8+ cells during Mtb infection. Additionally, our research brings to light that females exhibit a more robust systemic IFNγ (p ≤ 0.001) response than males during Mtb infection. This heightened response may either prevent active disease or contribute to latency in females during Mtb infection. In summary, our comprehensive analysis of the interplay between body fat changes and sex bias in Mtb infection reveals that alterations in body fat critically impact pulmonary pathology in males. Specifically, these changes significantly reduce the levels of pulmonary CD8+ T-cells and increase the Mtb burden in the lungs compared to females. The reduction in CD8+ cells in males is linked to an increase in mitochondrial oxidative phosphorylation and a decrease in TNFα, which are essential for CD8+ cell activation.


Asunto(s)
Tejido Adiposo , Pulmón , Mycobacterium tuberculosis , Animales , Femenino , Masculino , Ratones , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Pulmón/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/inmunología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología , Tuberculosis Pulmonar/microbiología , Ratones Transgénicos , Factores Sexuales , Modelos Animales de Enfermedad , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Caracteres Sexuales , Ratones Endogámicos C57BL
3.
J Am Heart Assoc ; 13(12): e034096, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38888318

RESUMEN

BACKGROUND: Recent studies suggest that immune-mediated inflammation of perivascular adipose tissue of abdominal aortic aneurysms (AAAs) contributes to disease development and progression. Whether the perivascular adipose tissue of AAA is characterized by a specific adaptive immune signature remains unknown. METHODS AND RESULTS: To investigate this hypothesis, we sequenced the T-cell receptor ß-chain in the perivascular adipose tissue of patients with AAA and compared it with patients with aortic occlusive disease, who share the former anatomical site of the lesion and risk factors but differ in pathogenic mechanisms. Our results demonstrate that patients with AAA have a lower repertoire diversity than those with aortic occlusive disease and significant differences in variable/joining gene segment usage. Furthermore, we identified a set of 7 public T-cell receptor ß-chain clonotypes that distinguished AAA and aortic occlusive disease with very high accuracy. We also found that the T-cell receptor ß-chain repertoire differentially characterizes small and large AAAs (aortic diameter<55 mm and ≥55 mm, respectively). CONCLUSIONS: This work supports the hypothesis that T cell-mediated immunity is fundamental in AAA pathogenesis and opens up new clinical perspectives.


Asunto(s)
Aneurisma de la Aorta Abdominal , Humanos , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Masculino , Anciano , Femenino , Linfocitos T/inmunología , Tejido Adiposo/patología , Tejido Adiposo/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Persona de Mediana Edad , Aorta Abdominal/patología , Aorta Abdominal/inmunología
5.
Ann Endocrinol (Paris) ; 85(3): 226-230, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38871498

RESUMEN

IMPORTANCE: Epicardial adipose tissue (EAT) is a biologically active organ surrounding myocardium and coronary arteries that has been associated with coronary artery disease (CAD) and atrial fibrillation. Previous work has shown that EAT exhibits beige features. OBJECTIVE: Our objective was to determine whether the stromal vascular fraction of the human EAT contains innate or adaptive lymphoid cells compared to thoracic subcutaneous (thSAT), visceral abdominal (VAT) and subcutaneous abdominal (abSAT). PARTICIPANTS: New pangenomic microarray analysis was performed on previous transcriptomic dataset using significance analysis of microarray and ingenuity pathway analysis (n=41) to identify specific immune signature and its link with browning genes. EAT, thSAT, VAT and abSAT samples from explanted patients with severe cardiomyopathies and multi-organ donor patients (n=17) were used for flow cytometry (FC) immunophenotyping assay. Patients were on average 55±16 years-old; 47% had hypertension and 6% CAD. Phenotypic adaptive and innate immune profiles were performed using a TBNK panel and a specific ILC1-2-3 panel including CD127, CD117, CRTH2 (CD294) and activation markers such as CD25 and CD69. RESULTS: Transcriptomic analysis showed a significant positive correlation between the TH2 immune pathway (IL-4, IL-5, IL-13, IL-25, IL-33) and browning genes (UCP-1, PRDM16, TMEM26, CITED1, TBX1) in EAT versus thSAT (R=0.82, P<0.0001). Regarding adaptive immune cells, a preponderance of CD8T cells, a contingent of CD4T cells, and a few B cells were observed in all ATs (P<0.0001). In innate lymphoid cells (ILCs), an increase was observed in visceral ATs (i.e. EAT; VAT 35±8ILCs/g of tissue) compared to their subcutaneous counterpart (i.e. thSAT+abSAT: 8±3 ILCs/g of AT, P=0.002), with a difference in the proportion of the 3 subtypes of ILCs (ILC1>ILC3>ILC2). In addition, we observed an increase in EAT-ILC2 compared to other ATs and almost all these EAT-ILC2 expressed CD69 and/or CD25 activation markers (99.75±0.16%; P<0.0001). We also observed more NKs in EAT and VAT (1520±71 cells/g of AT) than in SATs (562±17 cells/g of AT); P=0.01. CONCLUSION: This is the first study to provide a comparison between innate and adaptive lymphoid cells in human epicardial versus abdominal or thoracic adipose tissues. Further studies are ongoing to decipher whether these cells could be involved in EAT beiging. TRIAL REGISTRATION: CODECOH No. DC-2021-4518 The French agency of biomedicine PFS21-005.


Asunto(s)
Inmunidad Adaptativa , Tejido Adiposo , Inmunidad Innata , Pericardio , Humanos , Pericardio/inmunología , Pericardio/patología , Masculino , Persona de Mediana Edad , Femenino , Tejido Adiposo/inmunología , Anciano , Adulto , Linfocitos/inmunología , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Transcriptoma , Tejido Adiposo Epicárdico
7.
Front Immunol ; 15: 1375138, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38812501

RESUMEN

Objectives: The effects of cold exposure on whole-body metabolism in humans have gained increasing attention. Brown or beige adipose tissues are crucial in cold-induced thermogenesis to dissipate energy and thus have the potential to combat metabolic disorders. Despite the immune regulation of thermogenic adipose tissues, the overall changes in vital immune cells during distinct cold periods remain elusive. This study aimed to discuss the overall changes in immune cells under different cold exposure periods and to screen several potential immune cell subpopulations on thermogenic regulation. Methods: Cibersort and mMCP-counter algorithms were employed to analyze immune infiltration in two (brown and beige) thermogenic adipose tissues under distinct cold periods. Changes in some crucial immune cell populations were validated by reanalyzing the single-cell sequencing dataset (GSE207706). Flow cytometry, immunofluorescence, and quantitative real-time PCR assays were performed to detect the proportion or expression changes in mouse immune cells of thermogenic adipose tissues under cold challenge. Results: The proportion of monocytes, naïve, and memory T cells increased, while the proportion of NK cells decreased under cold exposure in brown adipose tissues. Conclusion: Our study revealed dynamic changes in immune cell profiles in thermogenic adipose tissues and identified several novel immune cell subpopulations, which may contribute to thermogenic activation of adipose tissues under cold exposure.


Asunto(s)
Tejido Adiposo Pardo , Frío , Termogénesis , Termogénesis/inmunología , Animales , Ratones , Tejido Adiposo Pardo/inmunología , Tejido Adiposo Pardo/metabolismo , Ratones Endogámicos C57BL , Masculino , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Beige/inmunología , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Monocitos/inmunología , Monocitos/metabolismo
8.
Rev Invest Clin ; 76(2): 65-79, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-38718804

RESUMEN

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).


Asunto(s)
Tejido Adiposo , Inflamación , Enfermedades Metabólicas , Obesidad , Humanos , Tejido Adiposo/metabolismo , Tejido Adiposo/inmunología , Obesidad/inmunología , Obesidad/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/etiología , Metabolismo Energético/fisiología , Adipocitos/metabolismo , Adipocitos/inmunología , Metabolismo de los Lípidos/fisiología , Animales , Homeostasis
9.
Biochem Pharmacol ; 225: 116324, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38815633

RESUMEN

Obesity is characterized by adipose tissue expansion, extracellular matrix remodelling and unresolved inflammation that contribute to insulin resistance and fibrosis. Adipose tissue macrophages represent the most abundant class of immune cells in adipose tissue inflammation and could be key mediators of adipocyte dysfunction and fibrosis in obesity. Although macrophage activation states are classically defined by the M1/M2 polarization nomenclature, novel studies have revealed a more complex range of macrophage phenotypes in response to external condition or the surrounding microenvironment. Here, we discuss the plasticity of adipose tissue macrophages (ATMs) in response to their microenvironment in obesity, with special focus on macrophage infiltration and polarization, and their contribution to adipose tissue fibrosis. A better understanding of the role of ATMs as regulators of adipose tissue remodelling may provide novel therapeutic strategies against obesity and associated metabolic diseases.


Asunto(s)
Tejido Adiposo , Fibrosis , Macrófagos , Obesidad , Humanos , Obesidad/metabolismo , Obesidad/patología , Obesidad/inmunología , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Macrófagos/fisiología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Tejido Adiposo/inmunología , Animales
10.
Immunity ; 57(6): 1289-1305.e9, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38772366

RESUMEN

Adipose tissue group 2 innate lymphoid cells (ILC2s) help maintain metabolic homeostasis by sustaining type 2 immunity and promoting adipose beiging. Although impairment of the ILC2 compartment contributes to obesity-associated insulin resistance, the underlying mechanisms have not been elucidated. Here, we found that ILC2s in obese mice and humans exhibited impaired liver kinase B1 (LKB1) activation. Genetic ablation of LKB1 disrupted ILC2 mitochondrial metabolism and suppressed ILC2 responses, resulting in exacerbated insulin resistance. Mechanistically, LKB1 deficiency induced aberrant PD-1 expression through activation of NFAT, which in turn enhanced mitophagy by suppressing Bcl-xL expression. Blockade of PD-1 restored the normal functions of ILC2s and reversed obesity-induced insulin resistance in mice. Collectively, these data present the LKB1-PD-1 axis as a promising therapeutic target for the treatment of metabolic disease.


Asunto(s)
Tejido Adiposo , Homeostasis , Resistencia a la Insulina , Linfocitos , Mitocondrias , Obesidad , Receptor de Muerte Celular Programada 1 , Proteínas Serina-Treonina Quinasas , Animales , Resistencia a la Insulina/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Mitocondrias/metabolismo , Humanos , Tejido Adiposo/metabolismo , Tejido Adiposo/inmunología , Obesidad/inmunología , Obesidad/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Inmunidad Innata , Masculino , Mitofagia/inmunología , Quinasas de la Proteína-Quinasa Activada por el AMP
11.
Am J Clin Nutr ; 120(1): 257-268, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38705359

RESUMEN

The rapidly evolving field of immunometabolism explores how changes in local immune environments may affect key metabolic and cellular processes, including that of adipose tissue. Importantly, these changes may contribute to low-grade systemic inflammation. In turn, chronic low-grade inflammation affecting adipose tissue may exacerbate the outcome of metabolic diseases. Novel advances in our understanding of immunometabolic processes may critically lead to interventions to reduce disease severity and progression. An important example in this regard relates to obesity, which has a multifaceted effect on immunity, activating the proinflammatory pathways such as the inflammasome and disrupting cellular homeostasis. This multifaceted effect of obesity can be investigated through study of downstream conditions using cellular and systemic investigative techniques. To further explore this field, the National Institutes of Health P30 Nutrition Obesity Research Center at Harvard, in partnership with Harvard Medical School, assembled experts to present at its 24th Annual Symposium entitled "Adiposity, Immunity, and Inflammation: Interrelationships in Health and Disease" on 7 June, 2023. This manuscript seeks to synthesize and present key findings from the symposium, highlighting new research and novel disease-specific advances in the field. Better understanding the interaction between metabolism and immunity offers promising preventative and treatment therapies for obesity-related immunometabolic diseases.


Asunto(s)
Adiposidad , Inflamación , Obesidad , Humanos , Inflamación/inmunología , Obesidad/inmunología , Tejido Adiposo/metabolismo , Tejido Adiposo/inmunología , Inmunidad
12.
PeerJ ; 12: e17267, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38699186

RESUMEN

Exercise has many beneficial effects that provide health and metabolic benefits. Signaling molecules are released from organs and tissues in response to exercise stimuli and are widely termed exerkines, which exert influence on a multitude of intricate multi-tissue processes, such as muscle, adipose tissue, pancreas, liver, cardiovascular tissue, kidney, and bone. For the metabolic effect, exerkines regulate the metabolic homeostasis of organisms by increasing glucose uptake and improving fat synthesis. For the anti-inflammatory effect, exerkines positively influence various chronic inflammation-related diseases, such as type 2 diabetes and atherosclerosis. This review highlights the prospective contribution of exerkines in regulating metabolism, augmenting the anti-inflammatory effects, and providing additional advantages associated with exercise. Moreover, a comprehensive overview and analysis of recent advancements are provided in this review, in addition to predicting future applications used as a potential biomarker or therapeutic target to benefit patients with chronic diseases.


Asunto(s)
Ejercicio Físico , Inflamación , Humanos , Inflamación/metabolismo , Ejercicio Físico/fisiología , Diabetes Mellitus Tipo 2/metabolismo , Músculo Esquelético/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/inmunología
13.
Ann Endocrinol (Paris) ; 85(3): 211-213, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38575108

RESUMEN

Adipose tissue is a highly immunologically active tissue that can be involved in many inflammatory diseases. In this presentation, only adipose tissue disorders associated with inflammatory diseases in children will be described, with the exception of obesity.


Asunto(s)
Tejido Adiposo , Inflamación , Humanos , Niño , Tejido Adiposo/inmunología , Tejido Adiposo/patología , Inflamación/inmunología , Obesidad/inmunología , Obesidad/complicaciones
15.
Int J Obes (Lond) ; 48(7): 993-1002, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38538853

RESUMEN

BACKGROUND: T-Lymphocyte activation is modulated by the adipokine leptin and serum concentrations of this hormone can be reduced with short-term calorie restriction. The aim of this study was to understand whether leptin per se is important in determining levels of T-lymphocyte activation in humans, by investigating whether the reduction in leptin concentration following calorie restriction is associated with a decrease in T-Lymphocyte activation in blood and adipose tissue. METHODS: Twelve men with overweight and obesity (age 35-55 years, waist circumference 95-115 cm) reduced their calorie intake by 50% for 3 consecutive days. Blood and subcutaneous adipose tissue were obtained for isolation of immune cells and cytokine analysis. CD4+ and CD8 + T-Lymphocytes were identified and characterised according to their expression of activation markers CD25 and CD69 by flow cytometry. RESULTS: Serum leptin was reduced by (mean ± SEM) 31 ± 16% (p < 0.001) following calorie restriction. The percentage of blood CD4 + CD25 + T-lymphocytes and level of CD25 expression on these lymphocytes were significantly reduced by 8 ± 10% (p = 0.016) and 8 ± 4% (p = 0.058), respectively. After calorie restriction, ex vivo leptin secretion from abdominal subcutaneous adipose tissue explants was not changed, and this corresponded with a lack of change in adipose tissue resident T-Lymphocyte activation. CONCLUSIONS: Serum leptin was reduced after calorie restriction and this was temporally associated with a reduction in activation of blood CD4 + CD25 + T-Lymphocytes. In abdominal subcutaneous adipose tissue, however, leptin secretion was unaltered, and there were no observed changes in adipose resident T-Lymphocyte activation.


Asunto(s)
Restricción Calórica , Leptina , Activación de Linfocitos , Obesidad , Sobrepeso , Humanos , Masculino , Leptina/sangre , Leptina/metabolismo , Adulto , Persona de Mediana Edad , Obesidad/sangre , Obesidad/inmunología , Obesidad/metabolismo , Sobrepeso/sangre , Sobrepeso/inmunología , Citometría de Flujo , Tejido Adiposo/metabolismo , Tejido Adiposo/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Pérdida de Peso/fisiología
16.
Eur J Immunol ; 54(5): e2350669, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38339772

RESUMEN

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.


Asunto(s)
Tejido Adiposo , Dieta Alta en Grasa , Inflamación , Macrófagos , Obesidad , Receptor Notch1 , Receptor Notch2 , Transducción de Señal , Animales , Macrófagos/inmunología , Macrófagos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/inmunología , Ratones , Dieta Alta en Grasa/efectos adversos , Inflamación/inmunología , Inflamación/metabolismo , Transducción de Señal/inmunología , Obesidad/metabolismo , Obesidad/inmunología , Receptor Notch1/metabolismo , Receptor Notch1/genética , Receptor Notch2/metabolismo , Receptor Notch2/genética , Ratones Noqueados , Ratones Endogámicos C57BL , Masculino
17.
Front Med ; 18(2): 205-236, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38165533

RESUMEN

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.


Asunto(s)
Tejido Adiposo , Macrófagos , Obesidad , Humanos , Obesidad/inmunología , Macrófagos/inmunología , Macrófagos/fisiología , Tejido Adiposo/inmunología , Inflamación/inmunología , Resistencia a la Insulina , Animales , Termogénesis/fisiología , Adipocitos
18.
Autoimmun Rev ; 23(3): 103502, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38101692

RESUMEN

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.


Asunto(s)
Adipocitos , Tejido Adiposo , Autoinmunidad , Humanos , Adipocitos/inmunología , Adipocitos/metabolismo , Autoinmunidad/inmunología , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Animales , Síndrome Metabólico/inmunología , Síndrome Metabólico/metabolismo , Resistencia a la Insulina/inmunología , Adiposidad/inmunología
19.
Front Immunol ; 14: 1153915, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37153549

RESUMEN

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.


Asunto(s)
Tejido Adiposo , Macrófagos , Enfermedades Metabólicas , Obesidad , Tejido Adiposo/inmunología , Macrófagos/clasificación , Macrófagos/inmunología , Obesidad/inmunología , Obesidad/terapia , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/terapia , Humanos , Inflamación/inmunología , Inflamación/terapia , Adipogénesis/inmunología , Polaridad Celular
20.
Immunity ; 56(4): 723-741, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37044062

RESUMEN

The immune response is tailored to the environment in which it takes place. Immune cells sense and adapt to changes in their surroundings, and it is now appreciated that in addition to cytokines made by stromal and epithelial cells, metabolic cues provide key adaptation signals. Changes in immune cell activation states are linked to changes in cellular metabolism that support function. Furthermore, metabolites themselves can signal between as well as within cells. Here, we discuss recent progress in our understanding of how metabolic regulation relates to type 2 immunity firstly by considering specifics of metabolism within type 2 immune cells and secondly by stressing how type 2 immune cells are integrated more broadly into the metabolism of the organism as a whole.


Asunto(s)
Sistema Inmunológico , Citocinas/inmunología , Humanos , Animales , Células Th2/inmunología , Macrófagos/inmunología , Adaptación Fisiológica , Tejido Adiposo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...