Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Lab Invest ; 99(4): 514-527, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30568176

RESUMEN

Cutaneous fibrosis is a common complication seen in mixed connective tissue diseases. It often occurs as a result of TGF-ß-induced deposition of excessive amounts of collagen in the skin. Lysyl oxidases (LOXs), a family of extracellular matrix (ECM)-modifying enzymes responsible for collagen cross-linking, are known to be increased in dermal fibroblasts from patients with fibrotic diseases, denoting a possible role of LOXs in fibrosis. To directly study this, we have developed two bioengineered, in vitro skin-like models: human skin equivalents (hSEs), and self-assembled stromal tissues (SASs) that contain either normal or systemic sclerosis (SSc; scleroderma) patient-derived fibroblasts. These tissues provide an organ-level structure that could be combined with non-invasive, label-free, multiphoton microscopy (SHG/TPEF) to reveal alterations in the organization and cross-linking levels of collagen fibers during the development of cutaneous fibrosis, which demonstrated increased stromal rigidity and activation of dermal fibroblasts in response to TGF-ß1. Specifically, inhibition of specific LOXs isoforms, LOX and LOXL4, in foreskin fibroblasts (HFFs) resulted in antagonistic effects on TGF-ß1-induced fibrogenic hallmarks in both hSEs and SASs. In addition, a translational relevance of these models was seen as similar antifibrogenic phenotypes were achieved upon knocking down LOXL4 in tissues containing SSc patient-derived-dermal fibroblasts (SScDFs). These findings point to a pivotal role of LOXs in TGF-ß1-induced cutaneous fibrosis through impaired ECM homeostasis in skin-like tissues, and show the value of these tissue platforms in accelerating the discovery of antifibrosis therapeutics.


Asunto(s)
Fibroblastos/metabolismo , Fibrosis/metabolismo , Proteína-Lisina 6-Oxidasa/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Aminoácido Oxidorreductasas/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Fibroblastos/citología , Humanos , Modelos Biológicos , Fenotipo , Piel/citología , Piel/metabolismo
2.
Br J Nutr ; 119(12): 1393-1399, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29845904

RESUMEN

Blueberry, rich in antioxidant and anti-inflammatory phytochemicals, has been demonstrated to lower inflammatory status in adipose induced by high-fat diet (HFD) and obesity. The effect of blueberry on systemic immune functions has not been examined. C57BL/6 mice were randomised to one of three diets - low-fat diet (LFD), HFD and HFD plus 4 % (w/w) blueberry (HFD+B) - for 8 or 12 weeks. Ex vivo T-cell mitogens (concanavalin A (Con A); phytohaemagglutinin), T-cell antibody (anti-CD3; anti-CD3/CD28)-stimulated T-cell proliferation and cytokine production were assessed. After 8 weeks, both HFD groups weighed more (>4 g) than the LFD group; after 12 weeks, HFD+B-fed mice weighed more (>6 g) and had 41 % more adipose tissue than HFD-fed mice (P<0·05). After 12 weeks, T-cell proliferation was less in both HFD groups, compared with the LFD group. HFD-associated decrements in T-cell proliferation were partially (10-50 %) prevented by blueberry supplementation. At 12 weeks, splenocytes from HFD mice, but not from HFD+B mice, produced 51 % less IL-4 (CD3/CD28) and 57 % less interferon-γ (Con A) compared with splenocytes from LFD mice (P<0·05). In response to lipopolysaccharide challenge, splenocytes from both HFD groups produced 24-30 % less IL-6 and 27-33 % less TNF-α compared with splenocytes from LFD mice (P<0·05), indicating impaired acute innate immune response. By demonstrating deleterious impacts of HFD feeding on T-cell proliferation and splenocyte immune responses, our results provide insights into how HFD/obesity can disrupt systemic immune function. The protective effects of blueberry suggest that dietary blueberry can buttress T-cell and systemic immune function against HFD-obesity-associated insults.


Asunto(s)
Arándanos Azules (Planta) , Suplementos Dietéticos , Obesidad/dietoterapia , Obesidad/inmunología , Linfocitos T/inmunología , Adiposidad , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Proliferación Celular , Citocinas/biosíntesis , Dieta con Restricción de Grasas , Dieta Alta en Grasa/efectos adversos , Inmunidad Celular , Inmunosupresores/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Linfocitos T/patología , Aumento de Peso
3.
Proc Natl Acad Sci U S A ; 111(14): 5313-8, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706839

RESUMEN

The HLA-F adjacent transcript 10 (FAT10) is a member of the ubiquitin-like gene family that alters protein function/stability through covalent ligation. Although FAT10 is induced by inflammatory mediators and implicated in immunity, the physiological functions of FAT10 are poorly defined. We report the discovery that FAT10 regulates lifespan through pleiotropic actions on metabolism and inflammation. Median and overall lifespan are increased 20% in FAT10ko mice, coincident with elevated metabolic rate, preferential use of fat as fuel, and dramatically reduced adiposity. This phenotype is associated with metabolic reprogramming of skeletal muscle (i.e., increased AMP kinase activity, ß-oxidation and -uncoupling, and decreased triglyceride content). Moreover, knockout mice have reduced circulating glucose and insulin levels and enhanced insulin sensitivity in metabolic tissues, consistent with elevated IL-10 in skeletal muscle and serum. These observations suggest novel roles of FAT10 in immune metabolic regulation that impact aging and chronic disease.


Asunto(s)
Adiposidad/genética , Longevidad/genética , Ubiquitinas/genética , Adipocitos/metabolismo , Animales , Biomarcadores/metabolismo , Metabolismo Energético , Femenino , Masculino , Ratones , Ratones Noqueados , Oxidación-Reducción , Triglicéridos/metabolismo
4.
Proc Natl Acad Sci U S A ; 110(13): 5133-8, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23479618

RESUMEN

Patients with type 2 diabetes (T2D) have disease-associated changes in B-cell function, but the role these changes play in disease pathogenesis is not well established. Data herein show B cells from obese mice produce a proinflammatory cytokine profile compared with B cells from lean mice. Complementary in vivo studies show that obese B cell-null mice have decreased systemic inflammation, inflammatory B- and T-cell cytokines, adipose tissue inflammation, and insulin resistance (IR) compared with obese WT mice. Reduced inflammation in obese/insulin resistant B cell-null mice associates with an increased percentage of anti-inflammatory regulatory T cells (Tregs). This increase contrasts with the sharply decreased percentage of Tregs in obese compared with lean WT mice and suggests that B cells may be critical regulators of T-cell functions previously shown to play important roles in IR. We demonstrate that B cells from T2D (but not non-T2D) subjects support proinflammatory T-cell function in obesity/T2D through contact-dependent mechanisms. In contrast, human monocytes increase proinflammatory T-cell cytokines in both T2D and non-T2D analyses. These data support the conclusion that B cells are critical regulators of inflammation in T2D due to their direct ability to promote proinflammatory T-cell function and secrete a proinflammatory cytokine profile. Thus, B cells are potential therapeutic targets for T2D.


Asunto(s)
Linfocitos B/inmunología , Citocinas/inmunología , Diabetes Mellitus Tipo 2/inmunología , Obesidad/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B/patología , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/terapia , Femenino , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/terapia , Masculino , Ratones , Ratones Obesos , Obesidad/patología , Obesidad/terapia , Linfocitos T Reguladores/patología
5.
J Control Release ; 336: 252-261, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34175365

RESUMEN

Current therapeutic treatments improving the impaired transportation of oxygen in acute respiratory distress syndrome (ARDS) have been found to be relevant and beneficial for the therapeutic treatment of COVID-19 patients suffering from severe respiratory complications. Hence, we report the preclinical and the preliminary results of the Phase I/II clinical trial of LEAF-4L6715, a liposomal nanocarrier encapsulating the kosmotropic agent trans-crocetin (TC), which, once injected, enhance the oxygenation of vascular tissue and therefore has the potential to improve the clinical outcomes of ARDS and COVID-19 in severely impacted patients. We demonstrated that the liposomal formulation enabled to increase from 30 min to 48 h the reoxygenation properties of free TCs in vitro in endothelial cells, but also to improve the half-life of TC by 6-fold in healthy mice. Furthermore, we identified 25 mg/kg as the maximum tolerated dose in mice. This determined concentration led to the validation of the therapeutic efficacy of LEAF-4 L6715 in a sepsis mouse model. Finally, we report the preliminary outcomes of an open-label multicenter Phase I/II clinical trial (EudraCT 2020-001393-30; NCT04378920), which was aimed to define the appropriate schedule and dosage of LEAF-4L6715 and to confirm its tolerability profile and preliminary clinical activity in COVID-19 patients treated in intensive care unit.


Asunto(s)
COVID-19 , Síndrome de Dificultad Respiratoria , Animales , Carotenoides , Células Endoteliales , Humanos , Ratones , Respiración Artificial , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , SARS-CoV-2 , Vitamina A/análogos & derivados
6.
J Nutr ; 139(8): 1510-6, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19515743

RESUMEN

Adipose tissue (AT) inflammation promotes insulin resistance (IR) and other obesity complications. AT inflammation and IR are associated with oxidative stress, adipocyte death, and the scavenging of dead adipocytes by proinflammatory CD11c+ AT macrophages (ATMPhi). We tested the hypothesis that supplementation of an obesitogenic (high-fat) diet with whole blueberry (BB) powder protects against AT inflammation and IR. Male C57Bl/6j mice were maintained for 8 wk on 1 of 3 diets: low-fat (10% of energy) diet (LFD), high-fat (60% of energy) diet (HFD) or the HFD containing 4% (wt:wt) whole BB powder (1:1 Vaccinium ashei and V. corymbosum) (HFD+B). BB supplementation (2.7% of total energy) did not affect HFD-associated alterations in energy intake, metabolic rate, body weight, or adiposity. We observed an emerging pattern of gene expression in AT of HFD mice indicating a shift toward global upregulation of inflammatory genes (tumor necrosis factor-alpha, interleukin-6, monocyte chemoattractant protein 1, inducible nitric oxide synthase), increased M1-polarized ATMPhi (CD11c+), and increased oxidative stress (reduced glutathione peroxidase 3). This shift was attenuated or nonexistent in HFD+B-fed mice. Furthermore, mice fed the HFD+B were protected from IR and hyperglycemia coincident with reductions in adipocyte death. Salutary effects of BB on adipocyte physiology and ATMPhi gene expression may reflect the ability of BB anthocyanins to alter mitogen-activated protein kinase and nuclear factor-kappaB stress signaling pathways, which regulate cell fate and inflammatory genes. These results suggest that cytoprotective and antiinflammatory actions of dietary BB can provide metabolic benefits to combat obesity-associated pathology.


Asunto(s)
Adipocitos/efectos de los fármacos , Antocianinas/farmacología , Antiinflamatorios/farmacología , Arándanos Azules (Planta) , Muerte Celular/efectos de los fármacos , Resistencia a la Insulina , Preparaciones de Plantas/farmacología , Adiposidad/efectos de los fármacos , Animales , Arándanos Azules (Planta)/química , Muerte Celular/genética , Quimiocina CCL2/metabolismo , Dieta , Grasas de la Dieta/administración & dosificación , Frutas , Expresión Génica/efectos de los fármacos , Glutatión Peroxidasa/metabolismo , Hiperglucemia/prevención & control , Interleucina-6/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Preparaciones de Plantas/química , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
7.
Brain Res ; 1181: 74-82, 2007 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-17961518

RESUMEN

The environmental neurotoxin arsenic has recently been associated with altered neurofilament (NF) content in sciatic nerve. We examined herein the impact of sodium arsenite (the inorganic form of arsenic) on NF dynamics. Treatment of differentiated NB2/d1 cells and cultured dorsal root ganglion neurons decreased NF transport into axonal neurites and increased perikaryal phospho-NF immunoreactivity. Both of these effects were prevented by a pharmacological inhibitor (SP600125) of c-jun terminal kinase and by expression of a dominant-negative form of this kinase. Arsenic-induced inhibition of NF transport was prevented by treatment with lithium, a selective inhibitor of glycogen synthase kinase-3beta. Pharmacological inhibitors of cyclin-dependent kinase 5 and p38 mitogen-activated protein kinase did not attenuate the effects of arsenic on NF dynamics. These latter findings suggest that this environmental neurotoxin could contribute to peripheral neuropathy by perturbing NF dynamics.


Asunto(s)
Arsénico/toxicidad , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Neuritas/efectos de los fármacos , Proteínas de Neurofilamentos/efectos de los fármacos , Neurotoxinas/toxicidad , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Células Cultivadas , Embrión de Pollo , Contaminantes Ambientales/toxicidad , Inhibidores Enzimáticos/farmacología , Epítopos/efectos de los fármacos , Epítopos/metabolismo , Ganglios Espinales/citología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Neuritas/metabolismo , Neuroblastoma , Proteínas de Neurofilamentos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación , Purinas/farmacología , Roscovitina , Células Tumorales Cultivadas
8.
Brain Res ; 1123(1): 20-6, 2006 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-17078934

RESUMEN

c-Jun N-terminal kinase (JNK), along with its upstream activator MEKK-1, is typically thought of as a stress-activated kinase that mediates apoptosis. However, additional studies indicate that the MEKK-1/JNK pathway mediates critical aspects of neuronal survival and differentiation. Herein, we demonstrate that transfection of differentiated NB2a/d1 cells with a construct expression constitutively activated (ca) MEKK-1 increases levels of phospho-dependent neurofilament (NF) immunoreactivity within perikarya, while expression of a dominant-negative (dn) form of MEKK-1 decreases it. Steady-state levels of perikaryal phospho-NF immunoreactivity are reduced and the increase resulting from expression of caMEKK-1 is prevented, by the JNK inhibitor SP600125, suggesting that JNK is a major downstream effector of MEKK-1 on NF phosphorylation. Unexpectedly, both caMEKK-1 and dnMEKK-1 inhibited neuritogenesis as well as translocation of NFs into newly elaborated neurites. The JNK inhibitor SP600125 also inhibited NF transport in a dose-dependent manner. caMEKK-1 also prevented the increase in NF transport otherwise mediated by MAP kinase. Finally, both caMEKK-1 and dnMEKK-1 prevented initial neuritogenesis. These findings indicate that the MEKK-1/JNK pathway regulates critical aspects of initial outgrowth, and subsequent stabilization of axonal neurites.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Neuritas/enzimología , Neurofibrillas/metabolismo , Animales , Línea Celular Tumoral , Ratones , Neuroblastoma , Neuronas/citología , Neuronas/metabolismo , Fosforilación , Transporte de Proteínas/fisiología , Transducción de Señal/fisiología
9.
Obesity (Silver Spring) ; 24(1): 102-12, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26576827

RESUMEN

OBJECTIVE: T cell inflammation plays pivotal roles in obesity-associated type 2 diabetes (T2DM). The identification of dominant sources of T cell inflammation in humans remains a significant gap in understanding disease pathogenesis. It was hypothesized that cytokine profiles from circulating T cells identify T cell subsets and T cell cytokines that define T2DM-associated inflammation. METHODS: Multiplex analyses were used to quantify T cell-associated cytokines in αCD3/αCD28-stimulated PBMCs, or B cell-depleted PBMCs, from subjects with T2DM or BMI-matched controls. Cytokine measurements were subjected to multivariate (principal component and partial least squares) analyses. Flow cytometry detected intracellular TNFα in multiple immune cell subsets in the presence/absence of antibodies that neutralize T cell cytokines. RESULTS: T cell cytokines were generally higher in T2DM samples, but Th17 cytokines are specifically important for classifying individuals correctly as T2DM. Multivariate analyses indicated that B cells support Th17 inflammation in T2DM but not control samples, while monocytes supported Th17 inflammation regardless of T2DM status. Partial least squares regression analysis indicated that both Th17 and Th1 cytokines impact %HbA1c. CONCLUSIONS: Among various T cell subsets, Th17 cells are major contributors to inflammation and hyperglycemia and are uniquely supported by B cells in obesity-associated T2DM.


Asunto(s)
Citocinas/inmunología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/inmunología , Obesidad/complicaciones , Obesidad/inmunología , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Adulto , Anciano , Linfocitos B/inmunología , Células Cultivadas , Femenino , Humanos , Inflamación/complicaciones , Inflamación/inmunología , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Adulto Joven
10.
Obesity (Silver Spring) ; 22(6): 1485-94, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24616441

RESUMEN

OBJECTIVE: To investigate the role of TNF-like weak inducer of apoptosis (TWEAK) in pathological adipose tissue (AT) remodeling and complications of obesity. METHODS: Wild type (WT) and TWEAK knockout (KO) mice were fed normal diet (ND) or a high fat diet (HFD) for up to 17 weeks. Adipocyte death was induced using an established transgenic mouse model of inducible adipocyte apoptosis (FAT-ATTAC). Metabolic, biochemical, histologic, and flow cytometric analyses were performed. RESULTS: TWEAK and its receptor, fibroblast growth factor-inducible molecule 14 (Fn14) were upregulated in gonadal (g)AT of WT mice after HFD week 4 and 24 h after induction of adipocyte apoptosis. Phenotypes of KO and WT mouse were indistinguishable through HFD week 8. However, at week 17 obese KO mice had ∼30% larger gAT adipocytes and gAT mass than WT mice, coincident with reduced adipocyte death, enhanced insulin signaling, Th2/M2 immune skewing, fewer thick collagen fibers, and altered expression of extracellular matrix constituents and modulators that is consistent with reduced fibrosis and larger adipocytes. KO mice were less steatotic and became more insulin sensitive and glucose tolerant than WT mice after HFD week 12. CONCLUSION: TWEAK constrains "healthy" gAT expansion and promotes metabolic complications in severe obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Apoptosis/fisiología , Eliminación de Gen , Obesidad Mórbida/genética , Obesidad Mórbida/prevención & control , Factores de Necrosis Tumoral/genética , Adipocitos/metabolismo , Animales , Citocina TWEAK , Dieta Alta en Grasa , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba
11.
J Leukoc Biol ; 96(2): 349-57, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24782490

RESUMEN

Individuals with T2D and PD suffer significantly from the ability of one disease to intensify the other. Disease-associated inflammation is one mechanism thought to fuel this pathogenic feed-forward loop. Several lines of evidence indicate that proinflammatory B cells promote T2D and PD; thus, B cells are top candidates for a cell type that predisposes PD in T2D. To test directly the role of B cells in T2D-associated PD, we compared outcomes from oral Porphyromonas gingivalis challenge of lean WT or B cell-null mice with outcomes from mice that were obese and insulin-resistant before challenge. Obese WT mice responded to oral P. gingivalis challenge with significant periodontal bone loss, whereas obese B cell-null mice were protected completely from PD. By contrast, lean WT and B cell-null mice suffer similar periodontal bone loss in response to oral pathogen. B cells from obese/insulin-resistant hosts also support oral osteoclastogenesis and both oral and systemic production of inflammatory cytokines, including pro-osteoclastogenic TNF-α and MIP-2, an ortholog of human IL-8. B cells furthermore impact AT inflammation in obese, P. gingivalis-infected hosts. Taken together, these data show that fundamentally different mechanisms regulate PD in lean and obese hosts, with B cells able to promote PD only if the hosts are "primed" by obesity. These results justify more intense analysis of obesity-associated changes in B cells that predispose PD in human T2D.


Asunto(s)
Linfocitos B/inmunología , Obesidad/inmunología , Periodontitis/inmunología , Porphyromonas gingivalis/inmunología , Animales , Linfocitos B/patología , Quimiocina CXCL2/genética , Quimiocina CXCL2/inmunología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Ratones Mutantes , Obesidad/genética , Obesidad/patología , Periodontitis/etiología , Periodontitis/genética , Periodontitis/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
12.
Endocrinology ; 153(9): 4266-77, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22778213

RESUMEN

Menopause promotes central obesity, adipose tissue (AT) inflammation, and insulin resistance (IR). Both obesity and the loss of estrogen can activate innate and adaptive immune cells (macrophages, T cells). The respective impacts of weight gain and loss of ovarian hormones on AT inflammation and IR are poorly understood. Here we determined the temporal kinetics of fat accretion, AT inflammation, and IR over a 26-wk time course in ovariectomized (OVX) mice, a model of menopause. OVX and sham-operated (SHM) C57BL6 mice were fed a normal chow diet. Weight, body composition (magnetic resonance imaging), total and regional adiposity, activity, food intake, AT crown-like structures, biohumoral measures, and insulin sensitivity (insulin tolerance testing and homeostatic model assessment) were determined at wk 12, 20, and 26. Macrophages and T cells from perigonadal AT were immunophenotyped by fluorescence-associated cell sorting, and perigonadal adipose tissue (PGAT) gene expression was quantified by quantitative PCR. OVX mice (≈ 31 g) became fatter than SHM mice (≈ 26 g) by wk 12, but mice were equally insulin sensitive. PGAT of OVX mice contained more T cells but expressed higher levels of M2-MΦ (arginase-1) and T cell-regulatory (cytotoxic T-lymphocyte antigen 4) genes. At wk 20, both OVX and SHM mice weighed approximately 35 g and were equally insulin sensitive with comparable amounts of PGAT and total body fat. OVX mice became less insulin sensitive than SHM mice by wk 26, coincident with the down-regulation of PGAT arginase-1 (-20-fold) and cytotoxic T-lymphocyte antigen 4 (2-fold) and up-regulation of M1/Th1 genes CD11c (+2-fold), IL12p40 (+2-fold), and interferon-γ (+78-fold). Ovarian hormone loss in mice induces PGAT inflammation and IR by mechanisms that can be uncoupled from OVX-induced obesity.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Adiposidad/fisiología , Resistencia a la Insulina/inmunología , Ovariectomía , Animales , Composición Corporal/fisiología , Peso Corporal/fisiología , Femenino , Citometría de Flujo , Inmunohistoquímica , Ratones , Reacción en Cadena de la Polimerasa
13.
Obesity (Silver Spring) ; 18(10): 1918-25, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20111012

RESUMEN

The role of adaptive immunity in obesity-associated adipose tissue (AT) inflammation and insulin resistance (IR) is controversial. We employed flow cytometry and quantitative PCR to assess T-cell recruitment and activation in epididymal AT (eAT) of C57BL/6 mice during 4-22 weeks of a high-fat diet (HFD (60% energy)). By week 6, eAT mass and stromal vascular cell (SVC) number increased threefold in mice fed HFD, coincident with onset of IR. We observed no increase in the proportion of CD3(+) SVCs or in gene expression of CD3, interferon-γ (IFN-γ), or regulated upon activation, normal T-cell expressed and secreted (RANTES) during the first 16 weeks of HFD. In contrast, CD11c(+) macrophages (MΦ) were enriched sixfold by week 8 (P < 0.01). SVC enrichment for T cells (predominantly CD4(+) and CD8(+)) and elevated IFN-γ and RANTES gene expression were detected by 20-22 weeks of HFD (P < 0.01), coincident with the resolution of eAT remodeling. HFD-induced T-cell priming earlier in the obesity time course is suggested by (i) elevated (fivefold) interleukin-12 (IL-12)p40 gene expression in eAT by week 12 (P ≤ 0.01) and (ii) greater IFN-γ secretion from phorbol myristate acetate (PMA)/ionophore-stimulated eAT explants at week 6 (onefold, P = 0.08) and week 12 (fivefold, P < 0.001). In conclusion, T-cell enrichment and IFN-γ gene induction occur subsequent to AT macrophage (ATMΦ) recruitment, onset of IR and resolution of eAT remodeling. However, enhanced priming for IFN-γ production suggests the contribution of CD4(+) and/or CD8(+) effectors to cell-mediated immune responses promoting HFD-induced AT inflammation and IR.


Asunto(s)
Tejido Adiposo/inmunología , Inflamación/etiología , Resistencia a la Insulina , Macrófagos/metabolismo , Obesidad/inmunología , Linfocitos T/metabolismo , Células TH1/metabolismo , Tejido Adiposo/metabolismo , Animales , Antígeno CD11c/metabolismo , Complejo CD3/metabolismo , Linfocitos T CD8-positivos/metabolismo , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Grasas de la Dieta/efectos adversos , Expresión Génica , Inflamación/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo , Subunidad p40 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/metabolismo , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Acetato de Tetradecanoilforbol/farmacología
14.
Endocrinology ; 150(12): 5240-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19819973

RESUMEN

The control of glucose metabolism is a complex process, and dysregulation at any level can cause impaired glucose tolerance and insulin resistance. These two defects are well-known characteristics associated with obesity and onset of type 2 diabetes. Here we introduce the N-terminal dipeptidase, DPP2, as a novel regulator of the glucose metabolism. We generated mice with a neurogenin 3 (NGN3)-specific DPP2 knockdown (kd) to explore a possible role of DPP2 in maintaining metabolic homeostasis. These mice spontaneously developed hyperinsulinemia, glucose intolerance, and insulin resistance by 4 months of age. In addition, we observed an increase in food intake in DPP2 kd mice, which was associated with a significant increase in adipose tissue mass and enhanced liver steatosis but no difference in body weight. In accordance with these findings, the mutant mice had a higher rate of respiratory exchange than the control littermates. This phenotype was exacerbated with age and when challenged with a high-fat diet. We report, for the first time, that DPP2 enzyme activity is essential for preventing hyperinsulinemia and maintaining glucose homeostasis. Interestingly, the phenotype of NGN3-DPP2 kd mice is opposite that of DPP4 knockout mice with regard to glucose metabolism, namely the former have normal glucagon-like peptide 1 levels but present with glucose intolerance, whereas the latter have increased glucagon-like peptide 1, which is accompanied by augmented glucose tolerance.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Intolerancia a la Glucosa/metabolismo , Resistencia a la Insulina , Proteínas del Tejido Nervioso/metabolismo , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Glucemia/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Ingestión de Alimentos , Ayuno/sangre , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Femenino , Técnica del Anticuerpo Fluorescente , Péptido 1 Similar al Glucagón/metabolismo , Intolerancia a la Glucosa/sangre , Insulina/sangre , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética
15.
Diabetes ; 56(12): 2910-8, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17848624

RESUMEN

OBJECTIVE: We sought to determine the role of adipocyte death in obesity-induced adipose tissue (AT) inflammation and obesity complications. RESEARCH DESIGN AND METHODS: Male C57BL/6 mice were fed a high-fat diet for 20 weeks to induce obesity. Every 4 weeks, insulin resistance was assessed by intraperitoneal insulin tolerance tests, and epididymal (eAT) and inguinal subcutaneous AT (iAT) and livers were harvested for histological, immunohistochemical, and gene expression analyses. RESULTS: Frequency of adipocyte death in eAT increased from <0.1% at baseline to 16% at week 12, coincident with increases in 1) depot weight; 2) AT macrophages (ATM Phi s) expressing F4/80 and CD11c; 3) mRNA for tumor necrosis factor (TNF)-alpha, monocyte chemotactic protein (MCP)-1, and interleukin (IL)-10; and 4) insulin resistance. ATM Phi s in crown-like structures surrounding dead adipocytes expressed TNF-alpha and IL-6 proteins. Adipocyte number began to decline at week 12. At week 16, adipocyte death reached approximately 80%, coincident with maximal expression of CD11c and inflammatory genes, loss (40%) of eAT mass, widespread collagen deposition, and accelerated hepatic macrosteatosis. By week 20, adipocyte number was restored with small adipocytes, coincident with reduced adipocyte death (fourfold), CD11c and MCP-1 gene expression (twofold), and insulin resistance (35%). eAT weight did not increase at week 20 and was inversely correlated with liver weight after week 12 (r = -0. 85, P < 0.001). In iAT, adipocyte death was first detected at week 12 and remained

Asunto(s)
Adipocitos/patología , Tejido Adiposo/fisiopatología , Obesidad/complicaciones , Obesidad/fisiopatología , Tejido Adiposo/patología , Animales , Muerte Celular , Modelos Animales de Enfermedad , Inflamación/fisiopatología , Macrófagos/citología , Macrófagos/patología , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Obesidad/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA