Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pediatr Res ; 91(5): 1028-1035, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33767375

RESUMEN

Most individuals with autism spectrum disorder (ASD)-a complex, life-long developmental disorder-do not have access to the care required to address their diverse health needs. Here, we review: (1) common barriers to healthcare access (shortage/cost of services; physician awareness; stigma); (2) barriers encountered primarily during childhood (limited screening/diagnosis; unclear referral pathways), transition to adulthood (insufficient healthcare transition services; suboptimal physician awareness of healthcare needs) and adulthood (shortage of services/limited insurance; communication difficulties with physicians; limited awareness of healthcare needs of aging adults); and (3) advances in research/program development for better healthcare access. A robust understanding of barriers to accessing healthcare across the lifespan of autistic individuals is critical to ensuring the best use of healthcare resources to improve social, physical, and mental health outcomes. Stakeholders must strengthen healthcare service provision by coming together to: better understand healthcare needs of underserved populations; strengthen medical training on care of autistic individuals; increase public awareness of ASD; promote research into/uptake of tools for ASD screening, diagnosis, and treatment; understand specific healthcare needs of autistic individuals in lower resource countries; and conduct longitudinal studies to understand the lifetime health, social, and economic impacts of ASD and enable the evaluation of novel approaches to increasing healthcare access. IMPACT: Despite the growing body of evidence, our understanding of barriers to healthcare encountered by individuals with ASD remains limited, particularly beyond childhood and in lower resource countries. We describe current and emerging barriers to healthcare access encountered by individuals with ASD across the lifespan. We recommend that stakeholders develop evidence-informed policies, programs, and technologies that address barriers to healthcare access for individuals with ASD and consider broad, equitable implementation to maximize impact.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Transición a la Atención de Adultos , Adulto , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/psicología , Trastorno del Espectro Autista/terapia , Trastorno Autístico/diagnóstico , Comunicación , Accesibilidad a los Servicios de Salud , Humanos
2.
Nat Commun ; 12(1): 2598, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33972511

RESUMEN

The intestinal immune system is an important modulator of glucose homeostasis and obesity-associated insulin resistance. Dietary factors, the intestinal microbiota and their metabolites shape intestinal immunity during obesity. The intestinal immune system in turn affects processes such as intestinal permeability, immune cell trafficking, and intestinal hormone availability, impacting systemic insulin resistance. Understanding these pathways might identify mechanisms underlying treatments for insulin resistance, such as metformin and bariatric surgery, or aid in developing new therapies and vaccination approaches. Here, we highlight evolving concepts centered on intestinal immunity, diet, and the microbiota to provide a working model of obesity-related metabolic disease.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/metabolismo , Obesidad/metabolismo , Animales , Linfocitos B/inmunología , Citocinas/metabolismo , Dietoterapia , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Resistencia a la Insulina/inmunología , Enfermedades Metabólicas/microbiología , Enfermedades Metabólicas/terapia , Obesidad/dietoterapia , Obesidad/inmunología , Obesidad/terapia , Linfocitos T/inmunología
3.
Nat Commun ; 10(1): 3650, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31409776

RESUMEN

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


Asunto(s)
Inmunoglobulina A/inmunología , Resistencia a la Insulina , Obesidad/inmunología , Tejido Adiposo/inmunología , Animales , Linfocitos B/inmunología , Estudios de Cohortes , Heces/microbiología , Microbioma Gastrointestinal , Glucosa/metabolismo , Humanos , Intestinos/inmunología , Masculino , Ratones , Obesidad/metabolismo , Obesidad/microbiología
5.
Int J Obes (Lond) ; 43(12): 2407-2421, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30944419

RESUMEN

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


Asunto(s)
Antiinflamatorios/farmacología , Carmin de Índigo/farmacología , Resistencia a la Insulina/fisiología , Obesidad/metabolismo , Receptores de Hidrocarburo de Aril/agonistas , Animales , Citocinas/metabolismo , Dieta Alta en Grasa , Microbioma Gastrointestinal , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Extractos Vegetales/química
6.
Cell ; 176(3): 610-624.e18, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30612739

RESUMEN

Plasma cells (PC) are found in the CNS of multiple sclerosis (MS) patients, yet their source and role in MS remains unclear. We find that some PC in the CNS of mice with experimental autoimmune encephalomyelitis (EAE) originate in the gut and produce immunoglobulin A (IgA). Moreover, we show that IgA+ PC are dramatically reduced in the gut during EAE, and likewise, a reduction in IgA-bound fecal bacteria is seen in MS patients during disease relapse. Removal of plasmablast (PB) plus PC resulted in exacerbated EAE that was normalized by the introduction of gut-derived IgA+ PC. Furthermore, mice with an over-abundance of IgA+ PB and/or PC were specifically resistant to the effector stage of EAE, and expression of interleukin (IL)-10 by PB plus PC was necessary and sufficient to confer resistance. Our data show that IgA+ PB and/or PC mobilized from the gut play an unexpected role in suppressing neuroinflammation.


Asunto(s)
Inmunoglobulina A/metabolismo , Interleucina-10/metabolismo , Intestinos/inmunología , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Inmunoglobulina A/inmunología , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Neuroinmunomodulación/inmunología , Células Plasmáticas/metabolismo
7.
Cell Metab ; 28(6): 922-934.e4, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30174303

RESUMEN

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


Asunto(s)
Antígenos CD/inmunología , Inmunidad Celular/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Activación de Linfocitos/inmunología , Receptor de Insulina/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD/genética , Citocinas/inmunología , Citocinas/metabolismo , Glucólisis/inmunología , Humanos , Inflamación/inmunología , Inflamación/virología , Insulina/metabolismo , Ganglios Linfáticos , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae , Receptor de Insulina/genética , Transducción de Señal , Bazo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo
8.
Sci Immunol ; 2(10)2017 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-28567448

RESUMEN

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

9.
Cell Rep ; 16(3): 717-30, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27373163

RESUMEN

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


Asunto(s)
Inflamación/metabolismo , Inflamación/patología , Resistencia a la Insulina/fisiología , Ácidos Nucleicos/metabolismo , Obesidad/metabolismo , Obesidad/patología , Adulto , Animales , Células Dendríticas/metabolismo , Células Dendríticas/patología , Dieta Alta en Grasa/métodos , Glucosa/metabolismo , Homeostasis/fisiología , Humanos , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Hígado/metabolismo , Hígado/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Receptores Toll-Like/metabolismo
10.
Cell Metab ; 23(3): 413-26, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26853748

RESUMEN

Obesity and insulin resistance are associated with chronic inflammation in metabolic tissues such as adipose tissue and the liver. Recently, growing evidence has implicated the intestinal immune system as an important contributor to metabolic disease. Obesity predisposes to altered intestinal immunity and is associated with changes to the gut microbiota, intestinal barrier function, gut-residing innate and adaptive immune cells, and oral tolerance to luminal antigens. Accordingly, the gut immune system may represent a novel therapeutic target for systemic inflammation in insulin resistance. This review discusses the emerging field of intestinal immunity in obesity-related insulin resistance and how it affects metabolic disease.


Asunto(s)
Resistencia a la Insulina/inmunología , Obesidad/inmunología , Inmunidad Adaptativa , Animales , Tracto Gastrointestinal/metabolismo , Humanos , Inmunidad Innata , Microbiota/inmunología , Obesidad/metabolismo , Obesidad/microbiología
11.
Turk Patoloji Derg ; 31 Suppl 1: 172-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26177326

RESUMEN

Excess energy intake and a sedentary lifestyle have led to increasing incidence of obesity which is a major risk factor for the development of insulin resistance. Research in the last two decades has revealed that chronic-low grade inflammation in adipose tissue is a key link between obesity and insulin resistance. As a result, adipose tissue is now considered an active immune organ with a key role in metabolic homeostasis. In the course of obesity, cells of the immune system infiltrate visceral adipose tissue (VAT) in an active process that promotes local and systemic inflammation. This inflammatory process in VAT is driven by various subsets of immune cells and is a central mechanism connecting obesity with its metabolic complications. One key event of adipose tissue inflammation is the switching of macrophages towards a pro-inflammatory phenotype. In addition, recent research has discovered an expanding list of immune cells contributing to this inflammatory process. Pro-inflammatory immune cells are crucial to obese VAT inflammation because of their production of cytokines, which can interfere with insulin signaling in peripheral tissues. This review summarizes our current knowledge of the pathology of innate and adaptive immune cells in obese adipose tissue, with emphasis in the immunological mechanisms mediating obesity-associated insulin resistance.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Grasa Intraabdominal/patología , Síndrome Metabólico/patología , Obesidad/patología , Paniculitis/patología , Animales , Metabolismo Energético , Humanos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/metabolismo , Síndrome Metabólico/inmunología , Síndrome Metabólico/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Paniculitis/inmunología , Paniculitis/metabolismo , Transducción de Señal
12.
Cell Metab ; 21(4): 527-42, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25863246

RESUMEN

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


Asunto(s)
Antiinflamatorios/farmacología , Tracto Gastrointestinal/inmunología , Inmunidad Mucosa/inmunología , Resistencia a la Insulina/inmunología , Obesidad/inmunología , Animales , Western Blotting , Citocinas/sangre , Dieta Alta en Grasa/efectos adversos , Citometría de Flujo , Tracto Gastrointestinal/efectos de los fármacos , Técnicas Histológicas , Inmunohistoquímica , Cadenas beta de Integrinas/metabolismo , Mesalamina/farmacología , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa/citología , Membrana Mucosa/inmunología
13.
Diabetes ; 64(1): 90-103, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25048196

RESUMEN

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


Asunto(s)
Intolerancia a la Glucosa/inmunología , Resistencia a la Insulina/inmunología , Grasa Intraabdominal/inmunología , Obesidad/inmunología , Paniculitis/inmunología , Perforina/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Dieta Alta en Grasa , Células Madre Embrionarias/citología , Femenino , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Insulina/metabolismo , Grasa Intraabdominal/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Paniculitis/genética , Paniculitis/metabolismo , Perforina/genética , Perforina/metabolismo , Bazo/citología , Bazo/inmunología
14.
Endocr Pathol ; 25(1): 93-101, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24356782

RESUMEN

Obesity is a major health burden worldwide and is a major factor in the development of insulin resistance and metabolic complications such as type II diabetes. Chronic nutrient excess leads to visceral adipose tissue (VAT) expansion and dysfunction in an active process that involves the adipocytes, their supporting matrix, and immune cell infiltrates. These changes contribute to adipose tissue hypoxia, adipocyte cell stress, and ultimately cell death. Accumulation of lymphocytes, macrophages, and other immune cells around dying adipocytes forms the so-called "crown-like structure", a histological hallmark of VAT in obesity. Cross talk between immune cells in adipose tissue dictates the overall inflammatory response, ultimately leading to the production of pro-inflammatory mediators which directly induce insulin resistance in VAT. In this review, we summarize recent studies demonstrating the dramatic changes that occur in visceral adipose tissue during obesity leading to low-grade chronic inflammation and metabolic disease.


Asunto(s)
Inflamación/patología , Grasa Intraabdominal/patología , Obesidad/patología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...