Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Intervalo de año de publicación
1.
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
2.
Front Immunol ; 12: 678355, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34322117

RESUMEN

Chronic inflammation plays an important role in the development of metabolic diseases. These include obesity, type 2 diabetes mellitus, and metabolic dysfunction-associated fatty liver disease. The proinflammatory environment maintained by the innate immunity, including macrophages and related cytokines, can be influenced by adaptive immunity. The function of T helper 17 (Th17) and regulatory T (Treg) cells in this process has attracted attention. The Th17/Treg balance is regulated by inflammatory cytokines and various metabolic factors, including those associated with cellular energy metabolism. The possible underlying mechanisms include metabolism-related signaling pathways and epigenetic regulation. Several studies conducted on human and animal models have shown marked differences in and the important roles of Th17/Treg in chronic inflammation associated with obesity and metabolic diseases. Moreover, Th17/Treg seems to be a bridge linking the gut microbiota to host metabolic disorders. In this review, we have provided an overview of the alterations in and the functions of the Th17/Treg balance in metabolic diseases and its role in regulating immune response-related glucose and lipid metabolism.


Asunto(s)
Susceptibilidad a Enfermedades , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Enfermedades Metabólicas/etiología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Adipogénesis/genética , Adipogénesis/inmunología , Animales , Biomarcadores , Microambiente Celular/inmunología , Citocinas/metabolismo , Humanos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Enfermedades Metabólicas/diagnóstico , Enfermedades Metabólicas/metabolismo , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Células Th17/metabolismo
3.
Platelets ; 32(7): 950-959, 2021 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-32835568

RESUMEN

Steroid-associated necrosis of the femoral head (SANFH) is one of the most common and refractory chronic diseases with increasing incidence. The typical pathological changes of SANFH include decreased osteogenic differentiation, enhanced intramedullary adipocytes deposition and impaired osseous circulation. In this study, we investigated the effects and potential mechanisms of Platelet-rich plasma (PRP) on SANFH. Sixty Sprague-Dawley rats were randomly divided into the control, PRP donor, model, and PRP groups. Compared to the model group, PRP treatment significantly increased the hemorheological indexes and serum levels of bone gla-protein (BGP) and vascular endothelial growth factor (VEGF), while decreased the levels of triglyceride (TG) and total cholesterol (TC). Meanwhile, Micro-CT and histopathological stain (Hematoxylin-eosin and Alcian blue-hematoxylin/orange G staining) were performed on the femoral head for morphological and histopathological evaluation, indicating that bone trabecular microstructure and bone mineral density (BMD) were significantly improved after PRP treatment. Immunohistochemical analysis revealed that PRP remarkably up-regulated the expression of osteogenic markers including ß-catenin and alkaline phosphatase (ALP), angiogenic markers containing VEGF and platelet endothelial cell adhesion molecule-1 (CD31), while down-regulated adipogenic markers involving fatty acid-binding protein (FABP-4), and peroxisome proliferator-activated receptor gamma (PPAR-γ) in SANFH rat models. In summary, for the first time, PRP was demonstrated to prevent the development of SANFH through stimulating bone formation and vascularization as well as retarding adipogenesis.


Asunto(s)
Adipogénesis/inmunología , Cabeza Femoral/patología , Osteogénesis/inmunología , Osteonecrosis/inducido químicamente , Plasma Rico en Plaquetas/metabolismo , Animales , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
4.
Int Immunol ; 32(6): 407-419, 2020 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-32147688

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are type 2 cytokine-producing cells that have important roles in helminth infection and allergic inflammation. ILC2s are tissue-resident cells, and their phenotypes and roles are regulated by tissue-specific environmental factors. While the role of ILC2s in the lung, intestine and bone marrow has been elucidated in many studies, their role in adipose tissues is still unclear. Here, we report on the role of ILC2-derived bone morphogenetic protein 7 (BMP7) in adipocyte differentiation and lipid accumulation. Co-culture of fat-derived ILC2s with pluripotent mesenchymal C3H10T1/2 cells and committed white preadipocyte 3T3-L1 cells resulted in their differentiation to adipocytes and induced lipid accumulation. Co-culture experiments using BMP7-deficient ILC2s revealed that BMP7, produced by ILC2s, induces differentiation into brown adipocytes. Our results demonstrate that BMP7, produced by ILC2s, affects adipocyte differentiation, particularly in brown adipocytes.


Asunto(s)
Adipogénesis/inmunología , Proteína Morfogenética Ósea 7/biosíntesis , Inmunidad Innata , Linfocitos/inmunología , Células 3T3-L1 , Animales , Células Cultivadas , Técnicas de Cocultivo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Curr Osteoporos Rep ; 18(1): 32-37, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31970653

RESUMEN

PURPOSE OF REVIEW: We reviewed recent progress on the role of sclerostin (SOST) and its effects on the immune system in order to summarize the current state of knowledge in osteoimmunology, in regard to hematopoiesis, lymphopoiesis, and inflammation. RECENT FINDINGS: Changes in sclerostin levels affect distinct niches within the bone marrow that support hematopoietic stem cells and B cell development. Sclerostin's regulation of adipogenesis could also be important for immune cell maintenance with age. Surprisingly, B cell development in the bone marrow is influenced by Sost produced by mesenchymal stem cells and osteoblasts, but not by osteocytes. Additionally, extramedullary hematopoiesis in the spleen and increased pro-inflammatory cytokine levels in the bone marrow are observed in global Sost-/- mice. In addition to changes in bone marrow density, sclerostin depletion affects B lymphopoiesis and myelopoiesis, as well as other changes within the bone marrow cavity that could affect hematopoiesis. It is therefore important to monitor for hematopoietic changes in patients receiving sclerostin-depleting therapies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Adipogénesis/inmunología , Hematopoyesis Extramedular/inmunología , Linfopoyesis/inmunología , Animales , Linfocitos B , Médula Ósea/inmunología , Citocinas/inmunología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas , Humanos , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Noqueados , Mielopoyesis/inmunología , Osteoblastos/inmunología , Osteocitos/inmunología
6.
Fish Shellfish Immunol ; 91: 343-349, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31042574

RESUMEN

Excessive body fat is a chronic inflammatory disorder. In this process, white adipose tissue (WAT) performs immune activities because of the dysregulated expression of adipokines. Excessive fat is accumulated in farmed fish, thereby threatening fish health. Studies have shown that adipose tissue is also an active immune organ in fish, capable of participating in and influencing immune responses. Adipocytes are the main cellular component of adipose tissue; however, little is known about the relationship between adipocyte and inflammation in fish. In this study, we analyzed transcriptome changes during adipogenesis in the primary culture of grass carp adipocytes using bioinformatics. The results showed that inflammatory signaling pathway may be activated during grass carp adipocyte differentiation, such as NFκB signaling pathway, Toll-like receptor signaling pathway and Adipocytokine signaling pathway, indicating that grass carp adipocytes have immune activities. Exposure to LPS induced expression of adipokines genes in adipocytes and preadipocytes, including NF-kB, IL-6, MCP-1 and TNFα, suggesting that preadipocytes and adipocytes both have immune response and the immune activity is conserved in vertebrates white adipocytes. Further study found that these immune marker genes were higher expressed in adipocytes compared with preadipocytes in LPS-induced inflammation. In summary, adipocyte should be considered as an active immune site in fish. Adipocytes have greater potency compared with preadipocytes in LPS-induced inflammation. This study indicated that adipocytes and preadipocytes may have different contribution in inflammation.


Asunto(s)
Adipocitos/inmunología , Adipogénesis/inmunología , Carpas/inmunología , Inmunidad Innata/fisiología , Lipopolisacáridos/farmacología , Adipocitos/metabolismo , Animales
7.
Methods Mol Biol ; 1773: 137-146, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29687386

RESUMEN

Murine models of obesity or reduced adiposity are a valuable resource for understanding the role of adipocyte dysfunction in metabolic disorders. Adipose tissue stromal vascular cells or primary adipocytes derived from murine adipose tissue and grown in culture are essential tools for studying the mechanisms underlying adipocyte development and function. Herein, we describe methods for the isolation, expansion, and long-term storage of murine adipose-derived stromal/stem cells along with protocols for inducing adipogenesis in this cell population or isolating the adipose stromal vascular fraction cells for flow cytometric analysis.


Asunto(s)
Adipocitos/citología , Adipocitos/inmunología , Adipogénesis/inmunología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/inmunología , Animales , Colagenasas/farmacología , Femenino , Citometría de Flujo , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Rosiglitazona/farmacología , Factores de Tiempo
8.
Biochem Biophys Res Commun ; 495(1): 740-748, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29137982

RESUMEN

OBJECTIVES: It is well-known that the complement system plays an essential role in host immunity. Observational studies have indicated that complement system-related molecules such as complement factor B (CfB) and other components are correlated with obesity and/or insulin resistance parameters. In this study, we investigated the role of adipocyte-derived CfB in adipose tissue metabolism. METHODS: We investigated the expression level of complement system-related genes in adipocytes. To understand the role of CfB in adipocyte, we performed Cfb overexpression in 3T3-L1 preadipocytes and generated adipocyte-specific Cfb transgenic mice. RESULTS: Cfb expression was markedly enhanced in 3T3-L1 adipocytes co-cultured with macrophages following endotoxin stimulation. In Cfb-overexpressing cells, the expression of adipocyte differentiation/maturation-related genes encoding peroxisome proliferator-activated receptor γ (Pparγ), adipocyte Protein 2 and perilipin was significantly enhanced. Cfb transgenic mice showed a marked increase in the expression of genes encoding Pparγ, perilipin, sterol regulatory element-binding protein 1 c, and Cd36 in the subcutaneous adipose tissue. CONCLUSIONS: CfB plays a crucial role in late-phase of adipocyte differentiation and subsequent lipid droplet formation.


Asunto(s)
Adipocitos/inmunología , Tejido Adiposo/inmunología , Diferenciación Celular/inmunología , Factor B del Complemento/inmunología , Inmunidad Innata/inmunología , Gotas Lipídicas/inmunología , Células 3T3-L1 , Adipocitos/citología , Adipogénesis/inmunología , Tejido Adiposo/citología , Animales , Proliferación Celular , Células Cultivadas , Masculino , Ratones , Ratones Transgénicos
9.
J Clin Endocrinol Metab ; 102(11): 4273-4283, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28938397

RESUMEN

Context: T helper (Th)17 cells are correlated with many human autoimmune disorders, including Graves disease, and may play key roles in the pathogenesis of Graves orbitopathy (GO). Objective: To study the phenotype of Th17 cells in patients with GO and healthy subjects, investigate the fibrosis and adipogenesis in orbital fibroblasts (OFs) modulated by interleukin (IL)-17A, and determine the interaction between Th17 cells and OFs. Design/Setting/Participants: Blood samples and orbital tissues from GO patients and healthy controls were collected. Main Outcome Measures: We conducted multicolor flow cytometry, immunohistochemical and immunofluorescent stainings, Western blotting, a PathScan intracellular signaling assay, Luminex and enzyme-linked immunosorbent assays, and protein mass spectrum. Results: Interferon-γ- and IL-22-expressing Th17 cells are increased in GO patients, which are positively related to clinical activity score. Costimulatory molecules are highly expressed in GO orbits and most GO OFs are CD90+. IL-17A promotes TGF-ß-induced fibrosis in CD90+ OFs but impedes 15-deoxy-Δ12,14-prostaglandin J2-induced adipogenesis in CD90- OFs. Th17 cells promote proinflammatory cytokine secretion in both CD90+ and CD90- OFs. Meanwhile, both CD90+ and CD90- OFs contribute to Th17 cell differentiation through prostaglandin E2 production, which can be attenuated by indomethacin. Furthermore, Th17 cells upregulate costimulatory molecule expression on OFs. Conclusion: Our findings unravel the pathogenicity of IL-17A in the initiation and progression of GO. In-depth interpretation of the molecular basis of OFs delineated by CD90 and Th17-OF interaction will help to afford a novel approach to better therapeutic strategies for GO.


Asunto(s)
Adipogénesis/inmunología , Oftalmopatía de Graves/complicaciones , Órbita/inmunología , Órbita/patología , Células Th17/fisiología , Adipocitos/fisiología , Estudios de Casos y Controles , Diferenciación Celular , Células Cultivadas , Fibroblastos/inmunología , Fibroblastos/patología , Fibrosis , Oftalmopatía de Graves/inmunología , Oftalmopatía de Graves/metabolismo , Oftalmopatía de Graves/patología , Humanos , Receptores de Hialuranos/metabolismo , Órbita/metabolismo , Células Th17/inmunología
10.
Obesity (Silver Spring) ; 25(11): 1932-1940, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28944626

RESUMEN

OBJECTIVE: Caveolin-1 (Cav-1) is expressed abundantly in adipose tissue and involved in many physiological processes. While Cav-1 has been reported to be secreted in pancreatic acinar cells and LNCaP prostate cancer cells, its secretion from adipose tissue awaits investigation. METHODS: Cav-1 secretion from 3T3-L1 adipocytes and fat tissues from normal chow diet- and high-fat diet (HFD)-fed mice was measured. Functions and uptake of secreted Cav-1 proteins were assessed by adding Cav-1 back to preadipocytes and LNCaP cells. RESULTS: Cav-1 secretion was evident in adipose tissues and were substantially promoted in HFD-fed mice. Cav-1 was detectable in the conditioned media of 3T3-L1 adipocytes but not preadipocytes. Hypertrophied adipocytes induced by glucose and fatty acids secreted more Cav-1, suggesting that hypertrophied adipocytes were responsible for enhanced Cav-1 secretion in obese mice. Secreted Cav-1 was taken up by preadipocytes and LNCaP cells. 3T3-L1 preadipocytes overexpressing Cav-1 were better differentiated, suggesting that secreted Cav-1 may promote adipogenesis. Hypertrophied 3T3-L1 adipocytes enhanced ERK1/2 activation, and the attenuation of ERK1/2 activity by PD98059 inhibited Cav-1 secretion. CONCLUSIONS: Cav-1 is actively secreted from adipocytes as a putative adipogenesis enhancer. Hypertrophied adipocytes secrete Cav-1 via ERK1/2-dependent mechanisms to promote adipogenesis, thus establishing a vicious cycle.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/inmunología , Tejido Adiposo/metabolismo , Caveolina 1/metabolismo , Animales , Técnicas de Cultivo de Célula , Masculino , Ratones
11.
Nat Immunol ; 18(6): 654-664, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28414311

RESUMEN

In obesity, inflammation of white adipose tissue (AT) is associated with diminished generation of beige adipocytes ('beige adipogenesis'), a thermogenic and energy-dissipating function mediated by beige adipocytes that express the uncoupling protein UCP1. Here we delineated an inflammation-driven inhibitory mechanism of beige adipogenesis in obesity that required direct adhesive interactions between macrophages and adipocytes mediated by the integrin α4 and its counter-receptor VCAM-1, respectively; expression of the latter was upregulated in obesity. This adhesive interaction reciprocally and concomitantly modulated inflammatory activation of macrophages and downregulation of UCP1 expression dependent on the kinase Erk in adipocytes. Genetic or pharmacological inactivation of the integrin α4 in mice resulted in elevated expression of UCP1 and beige adipogenesis of subcutaneous AT in obesity. Our findings, established in both mouse systems and human systems, reveal a self-sustained cycle of inflammation-driven impairment of beige adipogenesis in obesity.


Asunto(s)
Adipocitos Beige , Adipogénesis/inmunología , Tejido Adiposo Blanco/inmunología , Diferenciación Celular/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Obesidad/inmunología , Células 3T3-L1 , Adipocitos/inmunología , Adipocitos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Adhesión Celular/inmunología , Dieta Alta en Grasa , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Retroalimentación , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Immunoblotting , Integrina alfa4/genética , Macrófagos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Monocitos/inmunología , Obesidad/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Grasa Subcutánea , Linfocitos T/inmunología , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Adulto Joven
12.
Biochem Biophys Res Commun ; 484(4): 871-877, 2017 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-28167279

RESUMEN

Osteoporosis is one of the most common skeletal disease featured by osteopenia and adipose accumulation in bone tissue. NLRP3 inflammasome activation is an essential player in aging-related chronic diseases like osteoporosis, particularly due to the causal caspase-1 activation and its correlation to adipose accumulation in bone tissue. Moreover, the expression of anti-aging/senescence SIRT1 was reported to decline along with aging. As the major cellular contributor of bone formation, mesenchymal stem cells (MSCs) are multipotent stem cells processing mutually exclusive differentiatability toward osteocytes or adipocytes. Therefore, we hypothesized that NLRP3 inflammasome activation promotes adipogenesis and repress osteogenesis in MSCs via inhibiting SIRT1 expression. We activated NLRP3 inflammasome in human MSCs via lipopolysaccharide and palmitic acid (LPS/PA) treatment for self-renewal maintenance, adipogenic differentiation or osteogenic differentiation. LPS/PA treatment significantly increased NLRP3 expression, decreased SIRT1 expression and promoted caspase-1 activity in MSCs. LPS/PA treatment also boosted adipogenesis of MSCs and suppressed osteogenesis. Moreover, inhibition of caspase-1 activity repressed adipogenic differentiation and partially improved osteogenic differentiation of MSCs with LPS/PA treatment. Our study demonstrated the pivotal roles of NLRP3 inflammasome and downstream mediator caspase-1 for the progress of osteo-differentiation MSCs, and offered novel therapeutic target of treatment for osteoporosis.


Asunto(s)
Adipocitos/inmunología , Adipogénesis/inmunología , Inflamasomas/inmunología , Células Madre Mesenquimatosas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Osteogénesis/inmunología , Adipocitos/citología , Diferenciación Celular/inmunología , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología
13.
Curr Drug Targets ; 18(14): 1664-1675, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28056748

RESUMEN

BACKGROUND: Obesity was traditionally considered as a positive regulator on the strength of bone. With the in-depth study, obesity is considered as a major risk factor for osteoporosis. Some proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) are the factors that fat uses to negatively regulate bone metabolism. OBJECTIVE: This review was aimed to summarize and critically discuss the convincing evidence for the therapeutic effectiveness of pro-inflammatory cytokines for the treatment of obesity-related bone disorders. RESULTS: Obese people and animals show a higher level of serum TNF-α and IL-6, which are produced by macrophages derived from adipose tissue. These pro-inflammatory cytokines regulate the proliferation and apoptosis of adipocyte, promote lipolysis, inhibit lipid synthesis and decrease blood lipids through autocrine and paracrine way. On the other hand, TNF-α and IL-6 regulate bone metabolism through the endocrine way. Several reports suggest that TNF-α is a negative regulator of osteoblast at some stages of differentiation and positively regulates osteoclast proliferation and differentiation. In contrast, IL-6 influences osteoblasts and osteoclasts through complex mechanisms, which reflect dual effects. In addition, TNF-α and IL-6 may regulate bone metabolism indirectly by regulating adiponectin and leptin released from adipocytes. CONCLUSION: In this review, we first summarize the role of TNF-α and IL-6 in lipid and bone metabolisms. We further discuss how TNF-α and IL-6 regulate the communication between fat and bone, and their pathological roles in obesity-related bone disorders. Lastly, we discuss the possibility of using pro-inflammatory signaling pathway as a therapeutic target to develop drug for obesity-related bone disorders.


Asunto(s)
Interleucina-6/metabolismo , Obesidad/complicaciones , Osteoporosis/etiología , Factor de Necrosis Tumoral alfa/metabolismo , Adipogénesis/inmunología , Huesos/inmunología , Humanos , Metabolismo de los Lípidos/inmunología , Terapia Molecular Dirigida , Obesidad/tratamiento farmacológico , Obesidad/inmunología , Obesidad/metabolismo , Osteoblastos/inmunología , Osteoclastos/inmunología , Osteoporosis/tratamiento farmacológico , Osteoporosis/inmunología , Osteoporosis/metabolismo
14.
Curr Stem Cell Res Ther ; 12(3): 197-206, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27306400

RESUMEN

Mesenchymal stem cells (MSCs) are stromal origin cells with multilineage differentiation capacity. The immunoregulatory properties of MSCs can be interfered effectively by cytokines. Cytokines, produced by a broad range of cells, act at the systemic level to influence biological phenomena such as inflammation, wound healing, organogenesis and oncogenesis. Cytokines also play vital roles in the differentiation of MSCs into several cell lineages. This review summarizes on how cytokines can affect MSCs differentiation and their relative signaling pathways, which may serve to understand the possible underlying mechanisms. Also, this review reveals the potential clinical use of MSCs as promising therapeutic agents due to their special characteristics such as multipotent differentiation, immunomodulatory properties, and selfrestoration.


Asunto(s)
Adipogénesis/inmunología , Condrogénesis/inmunología , Citocinas/inmunología , Células Madre Mesenquimatosas/citología , Odontogénesis/inmunología , Osteogénesis/inmunología , Adipogénesis/genética , Animales , Diferenciación Celular , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Proliferación Celular , Condrogénesis/genética , Citocinas/genética , Expresión Génica , Humanos , Inmunomodulación , Células Madre Mesenquimatosas/inmunología , Odontogénesis/genética , Osteogénesis/genética , Transducción de Señal
15.
Biochem Biophys Res Commun ; 482(1): 87-92, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27816451

RESUMEN

In this work, we aimed to correlate the expression of sortilin with the production of sortilin-derived propeptide (PE) during adipocyte differentiation, insulin resistance and inflammation. We also investigated the effect of spadin, a shorter analogue of PE that exerts a potent antidepressant in mice, on adipocyte functions. During adipogenesis, insulin resistance and inflammation, we measured the mRNA and protein expression of sortilin, by quantitative PCR and Western-blot, and quantified the expression of PE by a specific dosing method. We observed that the production of PE was correlated with the sortilin expression during adipogenesis. Immunostaining experiments allowed to visualize the co-localization of sortilin, PE and VAMP2 in 3T3-L1 adipocytes. TNFα treatment induced insulin resistance and a decrease of sortilin expression (mRNA and protein), correlated with the decrease of the PE production. By contrast, treatment with dexamethasone, which also induced insulin resistance, was without effect on sortilin expression and PE production. As a putative bioactive peptide, we have evaluated its autocrine effect by the use of spadin on 3T3-L1 adipocytes by performing glucose uptake and signalling experiments. Any effect was measured on adipocytes indicating that the use of spadin as an antidepressant would have no side effects on adipocyte physiology.


Asunto(s)
Adipocitos/inmunología , Adipocitos/patología , Adipogénesis/inmunología , Paniculitis/inmunología , Paniculitis/patología , Péptidos/inmunología , Células 3T3-L1 , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Diferenciación Celular/inmunología , Ratones
16.
Cell Death Dis ; 7(11): e2458, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27831565

RESUMEN

Ribonuclease L (RNase-L) is an endoribonuclease well known for its roles in innate immunity. Recently it has been shown to regulate several cellular functions by modulating the levels of specific mRNAs. In this study, we investigated whether RNase-L may regulate adipocyte functions. We showed that knockdown of RNase-L reduced 3T3-L1 adipocyte differentiation and lipid accumulation. After mRNA profiling, we found that upregulation of Pref-1 mRNA, an inhibitory regulator of adipogenesis, could explain the reduced adipocyte differentiation with RNase-L downregulation. The signaling molecules downstream to Pref-1, including focal adhesion kinase, extracellular signal-regulated kinases and SRY-box 9, were activated by RNase-L suppression. The presence of Pref-1 mRNA was detected in the mRNP complexes precipitated by anti-RNase-L antibody. Moreover, the Pref-1 mRNA decay rate was raised by elevated RNase-L ribonuclease activity. Finally, in stable cell clones with RNase-L silencing, suppression of Pref-1 mRNA by specific siRNA partially recovered the adipocyte differentiation phenotype. Consistent with our findings, meta-analysis of 45 public array datasets from seven independent studies showed a significant negative relationship between RNase-L and Pref-1 mRNA levels in mouse adipose tissues. Higher RNase-L and lower Pref-1 mRNAs were found in the adipose tissues of high-fat diet mice compared to those of ND mice. In line with this, our animal data also showed that the adipose tissues of obese rats contained higher RNase-L and lower Pref-1 expression in comparison to that of lean rats. This study demonstrated that Pref-1 mRNA is a novel substrate of RNase-L. RNase-L is involved in adipocyte differentiation through destabilizing Pref-1 mRNA, thus offering a new link among RNA metabolism, innate immunity and adipogenesis in obesity progression.


Asunto(s)
Adipogénesis/inmunología , Endorribonucleasas/metabolismo , Inmunidad Innata , Péptidos y Proteínas de Señalización Intercelular/genética , Estabilidad del ARN , Células 3T3-L1 , Adipogénesis/genética , Tejido Adiposo/metabolismo , Animales , Proteínas de Unión al Calcio , Diferenciación Celular/genética , Dieta Alta en Grasa , Regulación hacia Abajo/genética , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Inmunidad Innata/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metabolismo de los Lípidos/genética , Masculino , Ratones , Modelos Biológicos , Obesidad/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Ribonucleoproteínas/metabolismo , Transducción de Señal/genética
17.
Can J Diabetes ; 40(5): 411-418, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27157387

RESUMEN

OBJECTIVES: Adipose tissue expands via differentiation of preadipocytes into adipocytes (adipogenesis) and/or hypertrophy of existing adipocytes. A low adipogenic capacity promotes adipocyte hypertrophy, causing inflammatory macrophage accumulation and insulin resistance. Macrophage-conditioned medium (MacCM) inhibits adipogenesis and promotes adipocyte inflammation, but it is unknown whether these effects are altered by high glucose (HG) versus normal glucose (NG) concentrations. Our aim was to compare the effect of HG-MacCM versus NG-MacCM on human adipogenesis and adipocyte inflammation. METHODS: Human monocyte-derived macrophages (MDMs) were placed in 5 mmol/L (NG) or 25 mmol/L (HG) glucose for 24 hours. MacCM was collected and its effect on differentiation of human subcutaneous abdominal preadipocytes and adipocyte inflammation was evaluated. RESULTS: HG-MacCM, but not NG-MacCM, inhibited triacylglycerol (TG) accumulation and protein expression of peroxisome proliferator-activated receptor γ (PPARgamma) during human adipogenesis. Preadipocytes differentiated in HG-MacCM displayed a more pro-inflammatory phenotype, as assessed by increased interleukin-6 and monocyte chemotactic protein-1 (MCP-1), as well as reduced adiponectin mRNA expression. In MDMs, HG increased phosphorylation of inhibitor of kappaB kinase (IKK)-beta and decreased protein expression of inhibitor of kappaB alpha. HG also reduced protein expression of PPARgamma in MDMs. However, no MDM changes in mRNA expression of MCP-1, interleukin-1beta or tumor necrosis factor-alpha were detected. The stimulatory effect of HG-MacCM on MCP-1 expression in adipocytes was partially inhibited when MDMs were treated with sc-514 (IKKbeta inhibitor). CONCLUSIONS: High glucose concentration accentuates the anti-adipogenic and pro-inflammatory effects of MacCM on human preadipocytes.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Glucosa/farmacología , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/inmunología , Adulto , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Humanos , Macrófagos/fisiología , Masculino , Persona de Mediana Edad
18.
Immunity ; 44(2): 355-67, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26872699

RESUMEN

Normal repair of skeletal muscle requires local expansion of a special population of Foxp3(+)CD4(+) regulatory T (Treg) cells. Such cells failed to accumulate in acutely injured muscle of old mice, known to undergo ineffectual repair. This defect reflected reduced recruitment of Treg cells to injured muscle, as well as less proliferation and retention therein. Interleukin-33 (IL-33) regulated muscle Treg cell homeostasis in young mice, and its administration to old mice ameliorated their deficits in Treg cell accumulation and muscle regeneration. The major IL-33-expressing cells in skeletal muscle displayed a constellation of markers diagnostic of fibro/adipogenic progenitor cells and were often associated with neural structures, including nerve fibers, nerve bundles, and muscle spindles, which are stretch-sensitive mechanoreceptors important for proprioception. IL-33(+) cells were more frequent after muscle injury and were reduced in old mice. IL-33 is well situated to relay signals between the nervous and immune systems within the muscle context.


Asunto(s)
Envejecimiento/inmunología , Interleucina-33/metabolismo , Músculo Esquelético/fisiología , Células Madre/inmunología , Linfocitos T Reguladores/inmunología , Adipogénesis/inmunología , Animales , Biomarcadores/metabolismo , Proliferación Celular , Células Cultivadas , Homeostasis , Humanos , Mecanotransducción Celular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/inervación , Fibras Nerviosas/fisiología , Neuroinmunomodulación , Regeneración/inmunología , Cicatrización de Heridas
19.
Rev Endocr Metab Disord ; 17(1): 41-9, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-26910560

RESUMEN

The adipose tissue (AT) is multifunctional, acting as an endocrine tissue and participating in the regulation of the organism's homeostasis. Metabolic, endocrine and inflammatory mechanisms are tightly intertwined within the AT, regulating its function. Disruption of the equilibrium among these mechanisms leads to pathologies, the most common being obesity-related insulin resistance. Two types of AT exist, the white and the brown AT. Traditionally the white AT (WAT) was thought to store energy in the form of lipids, while the brown AT (BAT) was known to mediate heat generation. Recently, the 'brite' or 'beige' AT was identified, which is localized predominantly in subcutaneous WAT, but shares functional features with the BAT and is capable of heat production. The major stimulus triggering beige and brown adipogenesis is cold exposure and catecholamine signalling. However, several further signals and mechanisms exist, which can orchestrate and fine-tune beige and brown AT function. Immune cells and inflammation have emerged as regulators of beige and brown AT function. The present review will focus on the recently identified crosstalk between innate immunity and the regulation of beige and brown adipogenesis.


Asunto(s)
Adipogénesis/inmunología , Tejido Adiposo Beige/inmunología , Tejido Adiposo Pardo/inmunología , Inmunidad Innata/inmunología , Animales , Humanos
20.
J Gerontol A Biol Sci Med Sci ; 71(9): 1131-40, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26450946

RESUMEN

The infrapatellar fat pad (IFP) secretes inflammatory mediators in osteoarthritic knees, but the effect of aging on IFP inflammation is unknown. We tested the hypothesis that aging increases basal and interleukin-1ß (IL-1ß)-stimulated IFP inflammation in 10-, 20-, and 30-month-old male F344BN F1-hybrid rats. IFPs were cultured ex vivo for 24 hours and treated ±1ng/mL IL-1ß to simulate injury-induced inflammation. IFP inflammation was evaluated by measuring secreted cytokine concentrations and by quantitative expression of immunoregulatory and pro- and anti-adipogenic genes. With age, osteoarthritis pathology increased and IFP mass decreased. Although adipocyte size did not change with age, variation in adipocyte size was positively associated with synovial thickness independent of age whereas associations with cartilage damage were age dependent. In the absence of IL-1ß, aging was associated with a significant increase in IFP secretion of tumor necrosis factor α by 67% and IL-13 by 35% and a reduction in the expression of immunoregulatory M2 macrophage genes. However, following an IL-1ß challenge, adipogenesis markers decreased and pro- and anti-inflammatory cytokines increased independent of age. The lone exception was leptin, which decreased >70% with age. Thus, although aging promotes osteoarthritis risk by increasing basal inflammation, our findings also revealed a potentially protective effect of aging by decreasing IL-1ß-stimulated leptin production.


Asunto(s)
Tejido Adiposo/inmunología , Envejecimiento , Interleucina-1beta/inmunología , Articulación de la Rodilla/inmunología , Osteoartritis de la Rodilla/inmunología , Adipogénesis/inmunología , Tejido Adiposo/patología , Animales , Biomarcadores/sangre , Técnicas In Vitro , Inflamación/inmunología , Interleucina-13/inmunología , Interleucina-1beta/deficiencia , Articulación de la Rodilla/patología , Leptina/inmunología , Masculino , Osteoartritis de la Rodilla/diagnóstico , Valor Predictivo de las Pruebas , Ratas , Ratas Endogámicas F344 , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...