Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nutrients ; 15(23)2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38068816

RESUMEN

Vertical transmission of obesity is a critical contributor to the unabated obesity pandemic and the associated surge in metabolic diseases. Existing experimental models insufficiently recapitulate "human-like" obesity phenotypes, limiting the discovery of how severe obesity in pregnancy instructs vertical transmission of obesity. Here, via utility of thermoneutral housing and obesogenic diet feeding coupled to syngeneic mating of WT obese female and lean male mice on a C57BL/6 background, we present a tractable, more "human-like" approach to specifically investigate how maternal obesity contributes to offspring health. Using this model, we found that maternal obesity decreased neonatal survival, increased offspring adiposity, and accelerated offspring predisposition to obesity and metabolic disease. We also show that severe maternal obesity was sufficient to skew offspring microbiome and create a proinflammatory gestational environment that correlated with inflammatory changes in the offspring in utero and adulthood. Analysis of a human birth cohort study of mothers with and without obesity and their infants was consistent with mouse study findings of maternal inflammation and offspring weight gain propensity. Together, our results show that dietary induction of obesity in female mice coupled to thermoneutral housing can be used for future mechanistic interrogations of obesity and metabolic disease in pregnancy and vertical transmission of pathogenic traits.


Asunto(s)
Enfermedades Metabólicas , Obesidad Materna , Efectos Tardíos de la Exposición Prenatal , Humanos , Femenino , Masculino , Ratones , Embarazo , Animales , Estudios de Cohortes , Vivienda , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Enfermedades Metabólicas/etiología
2.
Mucosal Immunol ; 16(6): 843-858, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37730122

RESUMEN

Influenza virus-induced respiratory pneumonia remains a major public health concern. Obesity, metabolic diseases, and female sex are viewed as independent risk factors for worsened influenza virus-induced lung disease severity. However, lack of experimental models of severe obesity in female mice limits discovery-based studies. Here, via utility of thermoneutral housing (30 °C) and high-fat diet (HFD) feeding, we induced severe obesity and metabolic disease in female C57BL/6 mice and compared their responses to severely obese male C57BL/6 counterparts during influenza virus infection. We show that lean male and female mice have similar lung edema, inflammation, and immune cell infiltration during influenza virus infection. At standard housing conditions, HFD-fed male, but not female, mice exhibit severe obesity, metabolic disease, and exacerbated influenza disease severity. However, combining thermoneutral housing and HFD feeding in female mice induces severe obesity and metabolic disease, which is sufficient to amplify influenza virus-driven disease severity to a level comparable to severely obese male counterparts. Lastly, increased total body weights of male and female mice at time of infection correlated with worsened influenza virus-driven disease severity metrics. Together, our findings confirm the impact of obesity and metabolic disease as key risk factors to influenza disease severity and present a novel mouse experimental model suitable for future mechanistic interrogation of sex, obesity, and metabolic disease traits in influenza virus-driven disease severity.


Asunto(s)
Gripe Humana , Enfermedades Metabólicas , Obesidad Mórbida , Infecciones por Orthomyxoviridae , Orthomyxoviridae , Masculino , Femenino , Animales , Ratones , Humanos , Obesidad Mórbida/complicaciones , Ratones Endogámicos C57BL , Obesidad , Gravedad del Paciente
3.
Front Immunol ; 14: 1095132, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36875069

RESUMEN

Introduction: Inflammation is a common unifying factor in experimental models of non-alcoholic fatty liver disease (NAFLD) progression. Recent evidence suggests that housing temperature-driven alterations in hepatic inflammation correlate with exacerbated hepatic steatosis, development of hepatic fibrosis, and hepatocellular damage in a model of high fat diet-driven NAFLD. However, the congruency of these findings across other, frequently employed, experimental mouse models of NAFLD has not been studied. Methods: Here, we examine the impact of housing temperature on steatosis, hepatocellular damage, hepatic inflammation, and fibrosis in NASH diet, methionine and choline deficient diet, and western diet + carbon tetrachloride experimental models of NAFLD in C57BL/6 mice. Results: We show that differences relevant to NAFLD pathology uncovered by thermoneutral housing include: (i) augmented NASH diet-driven hepatic immune cell accrual, exacerbated serum alanine transaminase levels and increased liver tissue damage as determined by NAFLD activity score; (ii) augmented methionine choline deficient diet-driven hepatic immune cell accrual and increased liver tissue damage as indicated by amplified hepatocellular ballooning, lobular inflammation, fibrosis and overall NAFLD activity score; and (iii) dampened western diet + carbon tetrachloride driven hepatic immune cell accrual and serum alanine aminotransferase levels but similar NAFLD activity score. Discussion: Collectively, our findings demonstrate that thermoneutral housing has broad but divergent effects on hepatic immune cell inflammation and hepatocellular damage across existing experimental NAFLD models in mice. These insights may serve as a foundation for future mechanistic interrogations focused on immune cell function in shaping NAFLD progression.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Ratones Endogámicos C57BL , Tetracloruro de Carbono , Vivienda , Cirrosis Hepática , Metionina , Alanina Transaminasa , Colina , Modelos Animales de Enfermedad , Inflamación
4.
Obes Rev ; 23(5): e13415, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34989117

RESUMEN

Type 2 diabetes (T2D) and obesity are independent risk factors for increased morbidity and mortality associated with influenza and SARS-CoV-2 infection. Skewed cellular metabolism shapes immune cell inflammatory responsiveness and function in obesity, T2D, and infection. However, altered immune cell responsiveness and levels of systemic proinflammatory mediators, partly independent of peripheral immune cell contribution, are linked with SARS-CoV-2-associated disease severity. Despite such knowledge, the role of tissue parenchymal cell-driven inflammatory responses, and specifically those dominantly modified in obesity (e.g., adipocytes), in influenza and SARS-CoV-2 infection pathogenesis remain poorly defined. Whether obesity-dependent skewing of adipocyte cellular metabolism uncovers inflammatory clades and promotes the existence of a 'pathogenic-inflammatory' adipocyte phenotype that amplifies SARS-CoV-2 infection diseases severity in individuals with obesity and individuals with obesity and T2D has not been examined. Here, using the knowledge gained from studies of immune cell responses in obesity, T2D, and infection, we highlight the key knowledge gaps underlying adipocyte cellular functions that may sculpt and grease pathogenic processes associated with influenza and SARS-CoV-2 disease severity in diabetes.


Asunto(s)
COVID-19 , Diabetes Mellitus Tipo 2 , Gripe Humana , Neumonía Viral , Diabetes Mellitus Tipo 2/etiología , Humanos , Gripe Humana/complicaciones , Gripe Humana/patología , Obesidad/metabolismo , Neumonía Viral/complicaciones , SARS-CoV-2
5.
STAR Protoc ; 2(4): 100937, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34778849

RESUMEN

Isolation of viable immune cells from human tissues is critical for the characterization of cellular and molecular processes underlying disease pathogenesis. Here, we describe protocols for the isolation of highly viable immune cells from liver wedges and mesenteric white adipose tissue resections from obese persons. Notably, characterization of the isolated single-immune cell suspensions, via utility of basic immunological interrogations and genetic approaches, promises to generate an improved understanding of altered immunological pathways in obese individuals with or without metabolic diseases. For complete details on the use and execution of this protocol, please refer to Moreno-Fernandez et al. (2021).


Asunto(s)
Separación Celular/métodos , Sistema Inmunológico/citología , Hígado , Mesenterio , Análisis de la Célula Individual/métodos , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/patología , Adolescente , Biopsia , Células Cultivadas , Femenino , Humanos , Hígado/citología , Hígado/patología , Masculino , Mesenterio/citología , Mesenterio/patología , Obesidad Infantil
6.
Int J Obes (Lond) ; 45(11): 2377-2387, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34302121

RESUMEN

OBJECTIVE: The risks of excess sugar intake in addition to high-fat diet consumption on immunopathogenesis of obesity-associated metabolic diseases are poorly defined. Interleukin-4 (IL-4) and IL-13 signaling via IL-4Rα regulates adipose tissue lipolysis, insulin sensitivity, and liver fibrosis in obesity. However, the contribution of IL-4Rα to sugar rich diet-driven obesity and metabolic sequelae remains unknown. METHODS: WT, IL-4Rα-deficient (IL-4Rα-/-) and STAT6-deficient mice (STAT6-/-) male mice were fed low-fat chow, high fat (HF) or HF plus high carbohydrate (HC/fructose) diet (HF + HC). Analysis included quantification of: (i) body weight, adiposity, energy expenditure, fructose metabolism, fatty acid oxidation/synthesis, glucose dysmetabolism and hepatocellular damage; (ii) the contribution of the hematopoietic or non-hematopoietic IL-4Rα expression; and (iii) the relevance of IL-4Rα downstream canonical STAT6 signaling pathway in this setting. RESULTS: We show that IL-4Rα regulated HF + HC diet-driven weight gain, whole body adiposity, adipose tissue inflammatory gene expression, energy expenditure, locomotor activity, glucose metabolism, hepatic steatosis, hepatic inflammatory gene expression and hepatocellular damage. These effects were potentially, and in part, dependent on non-hematopoietic IL-4Rα expression but were independent of direct STAT6 activation. Mechanistically, hepatic ketohexokinase-A and C expression was dependent on IL-4Rα, as it was reduced in IL-4Rα-deficient mice. KHK activity was also affected by HF + HC dietary challenge. Further, reduced expression/activity of KHK in IL-4Rα mice had a significant effect on fatty acid oxidation and fatty acid synthesis pathways. CONCLUSION: Our findings highlight potential contribution of non-hematopoietic IL-4Rα activation of a non-canonical signaling pathway that regulates the HF + HC diet-driven induction of obesity and severity of obesity-associated sequelae.


Asunto(s)
Metabolismo Energético/fisiología , Interleucina-4/metabolismo , Obesidad/metabolismo , Animales , Modelos Animales de Enfermedad , Fructosa/efectos adversos , Resistencia a la Insulina/fisiología , Interleucina-4/análisis , Ratones , Obesidad/inmunología
7.
Nutr Diabetes ; 11(1): 15, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099626

RESUMEN

BACKGROUND: Understanding gender-associated bias in aging and obesity-driven metabolic derangements has been hindered by the inability to model severe obesity in female mice. METHODS: Here, using chow- or high fat diet (HFD)-feeding regimens at standard (TS) and thermoneutral (TN) housing temperatures, the latter to model obesity in female mice, we examined the impact of gender and aging on obesity-associated metabolic derangements and immune responsiveness. Analysis included quantification of: (i) weight gain and adiposity; (ii) the development and severity of glucose dysmetabolism and non-alcoholic fatty liver disease (NAFLD); and (iii) induction of inflammatory pathways related to metabolic dysfunction. RESULTS: We show that under chow diet feeding regimen, aging was accompanied by increased body weight and white adipose tissue (WAT) expansion in a gender independent manner. HFD feeding regimen in aged, compared to young, male mice at TS, resulted in attenuated glucose dysmetabolism and hepatic steatosis. However, under TS housing conditions only aged, but not young, HFD fed female mice developed obesity. At TN however, both young and aged HFD fed female mice developed severe obesity. Independent of gender or housing conditions, aging attenuated the severity of metabolic derangements in HFD-fed obese mice. Tempered severity of metabolic derangements in aged mice was associated with increased splenic frequency of regulatory T (Treg) cells, Type I regulatory (Tr1)-like cells and circulating IL-10 levels and decreased vigor of HFD-driven induction of inflammatory pathways in adipose and liver tissues. CONCLUSION: Our findings suggest that aging-associated altered immunological profile and inflammatory vigor may play a dominant role in the attenuation of obesogenic diet-driven metabolic dysfunction.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Metabólicas/metabolismo , Obesidad/metabolismo , Tejido Adiposo Blanco/metabolismo , Adiposidad , Animales , Dieta Alta en Grasa/métodos , Femenino , Glucosa/metabolismo , Humanos , Inflamación/metabolismo , Interleucina-10/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Factores Sexuales , Linfocitos T Reguladores/metabolismo , Aumento de Peso
8.
Cell Metab ; 33(6): 1187-1204.e9, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34004162

RESUMEN

Emerging evidence suggests a key contribution to non-alcoholic fatty liver disease (NAFLD) pathogenesis by Th17 cells. The pathogenic characteristics and mechanisms of hepatic Th17 cells, however, remain unknown. Here, we uncover and characterize a distinct population of inflammatory hepatic CXCR3+Th17 (ihTh17) cells sufficient to exacerbate NAFLD pathogenesis. Hepatic ihTh17 cell accrual was dependent on the liver microenvironment and CXCR3 axis activation. Mechanistically, the pathogenic potential of ihTh17 cells correlated with increased chromatin accessibility, glycolytic output, and concomitant production of IL-17A, IFNγ, and TNFα. Modulation of glycolysis using 2-DG or cell-specific PKM2 deletion was sufficient to reverse ihTh17-centric inflammatory vigor and NAFLD severity. Importantly, ihTh17 cell characteristics, CXCR3 axis activation, and hepatic expression of glycolytic genes were conserved in human NAFLD. Together, our data show that the steatotic liver microenvironment regulates Th17 cell accrual, metabolism, and competence toward an ihTh17 fate. Modulation of these pathways holds potential for development of novel therapeutic strategies for NAFLD.


Asunto(s)
Proteínas Portadoras/inmunología , Proteínas de la Membrana/inmunología , Enfermedad del Hígado Graso no Alcohólico/inmunología , Piruvato Quinasa/inmunología , Receptores CXCR3/inmunología , Células Th17/inmunología , Hormonas Tiroideas/inmunología , Animales , Línea Celular , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Th17/citología , Proteínas de Unión a Hormona Tiroide
9.
Nat Commun ; 12(1): 2911, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34006859

RESUMEN

The impact of immune mediators on weight homeostasis remains underdefined. Interrogation of resistance to diet-induced obesity in mice lacking a negative regulator of Toll-like receptor signaling serendipitously uncovered a role for B cell activating factor (BAFF). Here we show that overexpression of BAFF in multiple mouse models associates with protection from weight gain, approximating a log-linear dose response relation to BAFF concentrations. Gene expression analysis of BAFF-stimulated subcutaneous white adipocytes unveils upregulation of lipid metabolism pathways, with BAFF inducing white adipose tissue (WAT) lipolysis. Brown adipose tissue (BAT) from BAFF-overexpressing mice exhibits increased Ucp1 expression and BAFF promotes brown adipocyte respiration and in vivo energy expenditure. A proliferation-inducing ligand (APRIL), a BAFF homolog, similarly modulates WAT and BAT lipid handling. Genetic deletion of both BAFF and APRIL augments diet-induced obesity. Lastly, BAFF/APRIL effects are conserved in human adipocytes and higher BAFF/APRIL levels correlate with greater BMI decrease after bariatric surgery. Together, the BAFF/APRIL axis is a multifaceted immune regulator of weight gain and adipose tissue function.


Asunto(s)
Factor Activador de Células B/genética , Obesidad/genética , Transducción de Señal/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Aumento de Peso/genética , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Factor Activador de Células B/metabolismo , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Obesidad/etiología , Obesidad/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
10.
Mucosal Immunol ; 14(6): 1224-1234, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33958704

RESUMEN

Epidemiological evidence establishes obesity as an independent risk factor for increased susceptibility and severity to viral respiratory pneumonias associated with H1N1 influenza and SARS-CoV-2 pandemics. Given the global obesity prevalence, a better understanding of the mechanisms behind obese susceptibility to infection is imperative. Altered immune cell metabolism and function are often perceived as a key causative factor of dysregulated inflammation. However, the contribution of adipocytes, the dominantly altered cell type in obesity with broad inflammatory properties, to infectious disease pathogenesis remains largely ignored. Thus, skewing of adipocyte-intrinsic cellular metabolism may lead to the development of pathogenic inflammatory adipocytes, which shape the overall immune responses by contributing to either premature immunosenescence, delayed hyperinflammation, or cytokine storm in infections. In this review, we discuss the underappreciated contribution of adipocyte cellular metabolism and adipocyte-produced mediators on immune system modulation and how such interplay may modify disease susceptibility and pathogenesis of influenza and SARS-CoV-2 infections in obese individuals.


Asunto(s)
Adipocitos/metabolismo , COVID-19/metabolismo , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , SARS-CoV-2/metabolismo , Adipocitos/patología , Adipocitos/virología , COVID-19/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/virología , Gripe Humana/patología
11.
Nat Commun ; 11(1): 2745, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488081

RESUMEN

White adipose tissue inflammation, in part via myeloid cell contribution, is central to obesity pathogenesis. Mechanisms regulating adipocyte inflammatory potential and consequent impact of such inflammation in disease pathogenesis remain poorly defined. We show that activation of the type I interferon (IFN)/IFNα receptor (IFNAR) axis amplifies adipocyte inflammatory vigor and uncovers dormant gene expression patterns resembling inflammatory myeloid cells. IFNß-sensing promotes adipocyte glycolysis, while glycolysis inhibition impeded IFNß-driven intra-adipocyte inflammation. Obesity-driven induction of the type I IFN axis and activation of adipocyte IFNAR signaling contributes to obesity-associated pathogenesis in mice. Notably, IFNß effects are conserved in human adipocytes and detection of the type I IFN/IFNAR axis-associated signatures positively correlates with obesity-driven metabolic derangements in humans. Collectively, our findings reveal a capacity for the type I IFN/IFNAR axis to regulate unifying inflammatory features in both myeloid cells and adipocytes and hint at an underappreciated contribution of adipocyte inflammation in disease pathogenesis.


Asunto(s)
Adipocitos/metabolismo , Inflamación/metabolismo , Interferón Tipo I/metabolismo , Obesidad/metabolismo , Animales , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Interferón beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Receptor de Interferón alfa y beta/metabolismo
12.
PLoS Negl Trop Dis ; 14(2): e0008029, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32023240

RESUMEN

Interleukin-32 is a novel inflammatory mediator that has been described to be important in the immunopathogenesis and control of infections caused by Leishmania parasites. By performing experiments with primary human cells in vitro, we demonstrate that the expression of IL-32 isoforms is dependent on the time exposed to L. amazonensis and L. braziliensis antigens. Moreover, for the first time we show the functional consequences of three different genetic variations in the IL32 (rs4786370, rs4349147, rs1555001) modulating IL-32γ expression, influencing innate and adaptive cytokine production after Leishmania exposure. Using a Brazilian cohort of 107 American Tegumentary Leishmaniasis patients and a control cohort of 245 healthy individuals, the IL32 rs4786370 genetic variant was associated with protection against ATL, whereas the IL32 rs4349147 was associated with susceptibility to the development of localized cutaneous and mucosal leishmaniasis. These novel insights may help improve therapeutic strategies and lead to benefits for patients suffering from Leishmania infections.


Asunto(s)
Predisposición Genética a la Enfermedad , Variación Genética , Interleucinas/genética , Leishmania/clasificación , Leishmaniasis Cutánea/genética , Adulto , Anciano , Brasil/epidemiología , Citocinas/genética , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Leishmaniasis Cutánea/epidemiología , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/metabolismo , Masculino , Persona de Mediana Edad , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Isoformas de Proteínas , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
13.
Cytokine ; 125: 154793, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31398626

RESUMEN

Low-grade inflammation is associated with the development of insulin resistance in obese individuals. The present study aims to provide additional evidence strengthening the role of interleukin (IL)-32 in this key process. Using an IL-32 transgenic (IL-32tg) mouse model, we observed that IL-32tg fed a normal diet had greater body weight, due to greater accumulation of white adipose tissue (WAT) along with larger sized adipocytes. This led to metabolic consequences, with significant higher leptin levels and a trend towards hyperinsulinemia, indicating a phenotype resembling the metabolic syndrome. Adipocytes of IL-32tg mice were more prone to induce a pro-inflammatory response locally, which would be expected when predisposed to insulin resistance and type2 diabetes mellitus (T2D). In conclusion, our study provides novel evidence of a direct contribution of IL-32 to pathophysiological perturbations within the adipose tissue, possibly contributing to the metabolic syndrome that precedes frank insulin resistance and T2D. Future research should focus on the role of IL-32 in the obesity epidemic.


Asunto(s)
Adipocitos/citología , Adipoquinas/metabolismo , Tejido Adiposo Blanco/metabolismo , Interleucinas/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo Blanco/fisiopatología , Animales , Peso Corporal/genética , Peso Corporal/fisiología , Citocinas/metabolismo , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Inflamación/genética , Inflamación/metabolismo , Interleucinas/genética , Leptina/metabolismo , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética
14.
Cell Rep ; 28(10): 2659-2672.e6, 2019 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-31484076

RESUMEN

American tegumentary leishmaniasis is a vector-borne parasitic disease caused by Leishmania protozoans. Innate immune cells undergo long-term functional reprogramming in response to infection or Bacillus Calmette-Guérin (BCG) vaccination via a process called trained immunity, conferring non-specific protection from secondary infections. Here, we demonstrate that monocytes trained with the fungal cell wall component ß-glucan confer enhanced protection against infections caused by Leishmania braziliensis through the enhanced production of proinflammatory cytokines. Mechanistically, this augmented immunological response is dependent on increased expression of interleukin 32 (IL-32). Studies performed using a humanized IL-32 transgenic mouse highlight the clinical implications of these findings in vivo. This study represents a definitive characterization of the role of IL-32γ in the trained phenotype induced by ß-glucan or BCG, the results of which improve our understanding of the molecular mechanisms governing trained immunity and Leishmania infection control.


Asunto(s)
Inmunidad , Interleucinas/metabolismo , Leishmania braziliensis/fisiología , Leishmaniasis Cutánea/prevención & control , beta-Glucanos/farmacología , Adulto , Anciano , Animales , Vacuna BCG/inmunología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunidad/efectos de los fármacos , Interleucina-1/metabolismo , Leishmania braziliensis/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/parasitología , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Vacunación , Adulto Joven
15.
J Interferon Cytokine Res ; 39(8): 459-471, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30920343

RESUMEN

Comprehension of adipocyte function has evolved beyond a long-held belief of their inert nature, as simple energy storing and releasing cells. Adipocytes, including white, brown, and beige, are capable mediators of global metabolic health, but their intersection with inflammation is a budding field of exploration. Evidence hints at a reciprocal relationship adipocytes share with immune cells. Adipocyte's capacity to behave in an "immune-like" manner and ability to sense inflammatory cues that subsequently alter core adipocyte function might play an important role in shaping immune responses. Clarifying this intricate relationship could uncover previously underappreciated contribution of adipocytes to inflammation-driven human health and disease. In this review, we highlight the potential of largely underappreciated adipocyte "immune-like" function and how it may contribute to inflammation, immunity, and pathology of various diseases.


Asunto(s)
Adipocitos/inmunología , Inflamación/inmunología , Animales , Humanos
16.
Front Immunol ; 10: 2893, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921154

RESUMEN

Obesity is a prevalent predisposing factor to non-alcoholic fatty liver disease (NAFLD), the most common chronic liver disease in the developed world. NAFLD spectrum of disease involves progression from steatosis (NAFL), to steatohepatitis (NASH), cirrhosis and hepatocellular carcinoma (HCC). Despite clinical and public health significance, current FDA approved therapies for NAFLD are lacking in part due to insufficient understanding of pathogenic mechanisms driving disease progression. The etiology of NAFLD is multifactorial. The induction of both systemic and tissue inflammation consequential of skewed immune cell metabolic state, polarization, tissue recruitment, and activation are central to NAFLD progression. Here, we review the current understanding of the above stated cellular and molecular processes that govern macrophage contribution to NAFLD pathogenesis and how adipose tissue and liver crosstalk modulates macrophage function. Notably, the manipulation of such events may lead to the development of new therapies for NAFLD.


Asunto(s)
Susceptibilidad a Enfermedades , Macrófagos/inmunología , Macrófagos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Biomarcadores , Movimiento Celular , Plasticidad de la Célula/inmunología , Citocinas/metabolismo , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Metabolismo Energético , Humanos , Mediadores de Inflamación/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/terapia , Transducción de Señal
17.
Sci Rep ; 8(1): 14050, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30232372

RESUMEN

About 60% of RA patients don't achieve good response with biological disease-modifying anti-rheumatic drugs bDMARD treatment (including TNF inhibitors, TNFi's). Previously, a link between TNFα and interleukin (IL)-32 was reported in RA. However, the exact mechanism linking IL-32 to response to treatment as not been studied yet. Therefore, we explored the influence of a promoter single nucleotide polymorphism (SNP) rs4786370 in IL-32 on clinical responsiveness to TNFi's in RA patients, potentially serving as new biomarker in RA. Expression of pro-inflammatory cytokines by peripheral mononuclear cells (PBMCs) from RA patients and healthy individuals were studied. Moreover, "ex vivo response" and clinical response to anti-TNFα therapy (etanercept, adalimumab) were measured and stratified for the IL-32 SNP. Higher IL-32 protein production was observed in RA patients. Additionally, patients bearing the CC genotype showed higher IL-32 protein and cytokine expression. DAS28 was independent of the promoter SNP, however, the "ex vivo" cytokine response was not. IL-32 mRNA and protein production was higher in RA patients, with a trend towards higher concentrations in patients bearing the CC genotype. Furthermore, genotype dependent IL-1 beta production might predict clinical response to etanercept/adalimumab. This indicates that IL-32 could play a role in predicting response to treatment in RA.


Asunto(s)
Antirreumáticos/farmacología , Artritis Reumatoide/genética , Citocinas/metabolismo , Interleucinas/genética , Leucocitos Mononucleares/efectos de los fármacos , Polimorfismo de Nucleótido Simple , Adalimumab/farmacología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Estudios de Casos y Controles , Etanercept/farmacología , Femenino , Genotipo , Humanos , Interleucina-6/metabolismo , Interleucina-9/metabolismo , Interleucinas/metabolismo , Leucocitos Mononucleares/inmunología , Masculino , Regiones Promotoras Genéticas , Regulación hacia Arriba
18.
Semin Immunol ; 38: 15-23, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29551246

RESUMEN

Interleukin 32 (IL-32) is an intracellular cytokine produced by immune and non immune cells after different stimuli. It contributes to inflammation and control of intracellular pathogens mainly by inducing proinflammatory cytokines and microbicidal molecules. Evidence is rising showing that IL-32 can be considered an endogenous danger signal after tissue injury, amplifying the inflammatory process and acquired immune responses. It seems to be a master regulator of intracellular infectious diseases. In this review, first the general properties of IL-32 are described followed by its role in the immunopathogenesis of inflammatory and infectious diseases. Roles of IL-32 in the control of infectious diseases caused by intracellular pathogens are reported, and later a focus on IL-32 in leishmaniases, diseases caused by an intracellular protozoan, is presented.


Asunto(s)
Mediadores de Inflamación/inmunología , Interleucinas/inmunología , Espacio Intracelular/inmunología , Leishmania/inmunología , Leishmaniasis/inmunología , Transducción de Señal/inmunología , Animales , Citocinas/inmunología , Citocinas/metabolismo , Interacciones Huésped-Parásitos/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Interleucinas/metabolismo , Espacio Intracelular/parasitología , Leishmania/fisiología , Leishmaniasis/metabolismo , Leishmaniasis/parasitología
19.
Atherosclerosis ; 271: 193-202, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29524862

RESUMEN

BACKGROUND AND AIMS: The role of interleukin (IL-)32 in inflammatory conditions is well-established, however, the mechanism behind its role in atherosclerosis remains unexplained. Our group reported a promoter single nucleotide polymorphism in IL-32 associated with higher high-density lipoprotein (HDL) concentrations. We hypothesize that endogenous IL-32 in liver cells, a human monocytic cell line and carotid plaque tissue, can affect atherosclerosis by regulating (HDL) cholesterol homeostasis via expression of cholesterol transporters/mediators. METHODS: Human primary liver cells were stimulated with recombinant human (rh)TNFα and poly I:C to study the expression of IL-32 and mediators in cholesterol pathways. Additionally, IL-32 was overexpressed in HepG2 cells and overexpressed and silenced in THP-1 cells to study the direct effect of IL-32 on cholesterol transporters expression and function. RESULTS: Stimulation of human primary liver cells resulted in induction of IL-32α, IL-32ß and IL-32γ mRNA expression (p < 0.01). A strong correlation between the expression of IL-32γ and ABCA1, ABCG1, LXRα and apoA1 was observed (p < 0.01), and intracellular lipid concentrations were reduced in the presence of endogenous IL-32 (p < 0.05). Finally, IL32γ and ABCA1 mRNA expression was upregulated in carotid plaque tissue and when IL-32 was silenced in THP-1 cells, mRNA expression of ABCA1 was strongly reduced. CONCLUSIONS: Regulation of IL-32 in human primary liver cells, HepG2 and THP-1 cells strongly influences the mRNA expression of ABCA1, ABCG1, LXRα and apoA1 and affects intracellular lipid concentrations in the presence of endogenous IL-32. These data, for the first time, show an important role for IL32 in cholesterol homeostasis.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , HDL-Colesterol/metabolismo , Hepatocitos/metabolismo , Interleucinas/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Células Hep G2 , Hepatocitos/efectos de los fármacos , Homeostasis , Humanos , Interleucinas/genética , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Poli I-C/farmacología , Cultivo Primario de Células , Células THP-1 , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
20.
Sci Rep ; 7(1): 15219, 2017 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-29123157

RESUMEN

American Tegumentary Leishmaniasis is a chronic infection caused by Leishmania protozoan. It is not known whether genetic variances in NOD-like receptor (NLR) family members influence the immune response towards Leishmania parasites and modulate intracellular killing. Using functional genomics, we investigated whether genetic variants in NOD1 or NOD2 influence the production of cytokines by human PBMCs exposed to Leishmania. In addition, we examined whether recognition of Leishmania by NOD2 contributes to intracellular killing. Polymorphisms in the NOD2 gene decreased monocyte- and lymphocyte-derived cytokine production after stimulation with L. amazonensis or L. braziliensis compared to individuals with a functional NOD2 receptor. The phagolysosome formation is important for Leishmania-induced cytokine production and upregulation of NOD2 mRNA expression. NOD2 is crucial to control intracellular infection caused by Leishmania spp. NOD2 receptor is important for Leishmania recognition, the control of intracellular killing, and the induction of innate and adaptive immune responses.


Asunto(s)
Leishmania/inmunología , Leishmaniasis Cutánea/inmunología , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Polimorfismo Genético , Adulto , Anciano , Supervivencia Celular , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Países Bajos , Fagosomas/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA