Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell Mol Life Sci ; 78(1): 227-247, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32157317

RESUMEN

Chronic inflammation that affects primarily metabolic organs, such as white adipose tissue (WAT), is considered as a major cause of human obesity-associated co-morbidities. However, the molecular mechanisms initiating this inflammation in WAT are poorly understood. By combining transcriptomics, ChIP-seq and modeling approaches, we studied the global early and late responses to a high-fat diet (HFD) in visceral (vWAT) and subcutaneous (scWAT) AT, the first being more prone to obesity-induced inflammation. HFD rapidly triggers proliferation of adipocyte precursors within vWAT. However, concomitant antiadipogenic signals limit vWAT hyperplastic expansion by interfering with the differentiation of proliferating adipocyte precursors. Conversely, in scWAT, residing beige adipocytes lose their oxidizing properties and allow storage of excessive fatty acids. This phase is followed by tissue hyperplastic growth and increased angiogenic signals, which further enable scWAT expansion without generating inflammation. Our data indicate that scWAT and vWAT differential ability to modulate adipocyte number and differentiation in response to obesogenic stimuli has a crucial impact on the different susceptibility to obesity-related inflammation of these adipose tissue depots.


Asunto(s)
Adipogénesis , Tejido Adiposo Blanco/metabolismo , Diferenciación Celular , Inflamación/patología , Obesidad/patología , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/patología , Animales , Dieta Alta en Grasa , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Inflamación/etiología , Inflamación/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Grasa Intraabdominal/citología , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones , Transducción de Señal/genética , Células Madre/citología , Células Madre/metabolismo , Grasa Subcutánea/citología , Grasa Subcutánea/metabolismo , Grasa Subcutánea/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Wnt/metabolismo
2.
Int J Mol Sci ; 22(18)2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34576136

RESUMEN

Men with nonalcoholic fatty liver disease (NAFLD) are more exposed to nonalcoholic steatohepatitis (NASH) and liver fibrosis than women. However, the underlying molecular mechanisms of NALFD sex dimorphism are unclear. We combined gene expression, histological and lipidomic analyses to systematically compare male and female liver steatosis. We characterized hepatosteatosis in three independent mouse models of NAFLD, ob/ob and lipodystrophic fat-specific (PpargFΔ/Δ) and whole-body PPARγ-null (PpargΔ/Δ) mice. We identified a clear sex dimorphism occurring only in PpargΔ/Δ mice, with females showing macro- and microvesicular hepatosteatosis throughout their entire life, while males had fewer lipid droplets starting from 20 weeks. This sex dimorphism in hepatosteatosis was lost in gonadectomized PpargΔ/Δ mice. Lipidomics revealed hepatic accumulation of short and highly saturated TGs in females, while TGs were enriched in long and unsaturated hydrocarbon chains in males. Strikingly, sex-biased genes were particularly perturbed in both sexes, affecting lipid metabolism, drug metabolism, inflammatory and cellular stress response pathways. Most importantly, we found that the expression of key sex-biased genes was severely affected in all the NAFLD models we tested. Thus, hepatosteatosis strongly affects hepatic sex-biased gene expression. With NAFLD increasing in prevalence, this emphasizes the urgent need to specifically address the consequences of this deregulation in humans.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/patología , PPAR gamma/deficiencia , Caracteres Sexuales , Animales , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Femenino , Regulación de la Expresión Génica , Hormonas Esteroides Gonadales/metabolismo , Inflamación/patología , Gotas Lipídicas/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , PPAR gamma/metabolismo , Fenotipo , Transducción de Señal , Triglicéridos/metabolismo
3.
Am J Physiol Renal Physiol ; 314(6): F1154-F1165, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29488390

RESUMEN

Different complex mechanisms control the morphology of podocyte foot processes and their interactions with the underlying basement membrane. Injuries to this system often cause glomerular dysfunction and albuminuria. The present study aimed at identifying early markers of glomerular damage in diabetic nephropathy. For this purpose, we performed a microarray analysis on kidneys of 3-wk-old peroxisome proliferator-activated receptor-γ (PPARγ)-null and AZIP/F1 mice, which are two models of diabetic nephropathy due to lipodystrophy. This was followed by functional annotation of the enriched clusters of genes. One of the significant changes in the early stages of glomerular damage was the increase of hemicentin 1 (HMCN1). Its expression and distribution were then studied by real-time PCR and immunofluorescence in various models of glomerular damage and on podocyte cell cultures. HMCN1 progressively increased in the glomeruli of diabetic mice, according to disease severity, as well as in puromycin aminonucleoside (PA)-treated rats. Studies on murine and human podocytes showed an increased HMCN1 deposition upon different pathological stimuli, such as hyperglycemia, transforming growth factor-ß (TGF-ß), and PA. In vitro silencing studies showed that HMCN1 mediated the rearrangements of podocyte cytoskeleton induced by TGF-ß. Finally, we demonstrated an increased expression of HMCN1 in the kidneys of patients with proteinuric nephropathies. In summary, our studies identified HMCN1 as a new molecule involved in the dynamic changes of podocyte foot processes. Its increased expression associated with podocyte dysfunction points to HMCN1 as a possible marker for the early glomerular damage occurring in different proteinuric nephropathies.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Nefropatías Diabéticas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Inmunoglobulinas/metabolismo , Nefrosis/metabolismo , Podocitos/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/patología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Femenino , Glucosa/farmacología , Humanos , Inmunoglobulinas/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrosis/genética , Nefrosis/patología , PPAR gamma/genética , PPAR gamma/metabolismo , Podocitos/efectos de los fármacos , Podocitos/patología , Proteinuria/genética , Proteinuria/metabolismo , Proteinuria/patología , Ratas Sprague-Dawley , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba
4.
Int J Mol Sci ; 19(7)2018 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-30011852

RESUMEN

Skeletal muscle is a regenerative tissue which can repair damaged myofibers through the activation of tissue-resident muscle stem cells (MuSCs). Many muscle diseases with impaired regeneration cause excessive adipose tissue accumulation in muscle, alter the myogenic fate of MuSCs, and deregulate the cross-talk between MuSCs and fibro/adipogenic progenitors (FAPs), a bi-potent cell population which supports myogenesis and controls intra-muscular fibrosis and adipocyte formation. In order to better characterize the interaction between adipogenesis and myogenesis, we studied muscle regeneration and MuSC function in whole body Pparg null mice generated by epiblast-specific Cre/lox deletion (PpargΔ/Δ). We demonstrate that deletion of PPARγ completely abolishes ectopic muscle adipogenesis during regeneration and impairs MuSC expansion and myogenesis after injury. Ex vivo assays revealed that perturbed myogenesis in PpargΔ/Δ mice does not primarily result from intrinsic defects of MuSCs or from perturbed myogenic support from FAPs. The immune transition from a pro- to anti-inflammatory MuSC niche during regeneration is perturbed in PpargΔ/Δ mice and suggests that PPARγ signaling in macrophages can interact with ectopic adipogenesis and influence muscle regeneration. Altogether, our study demonstrates that a PPARγ-dependent adipogenic response regulates muscle fat infiltration during regeneration and that PPARγ is required for MuSC function and efficient muscle repair.


Asunto(s)
Adipogénesis/genética , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , PPAR gamma/genética , Regeneración/genética , Adipocitos/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Mioblastos/citología , Mioblastos/metabolismo , PPAR gamma/metabolismo
5.
Am J Physiol Endocrinol Metab ; 309(4): E357-69, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26081281

RESUMEN

PPARγ-deficient mice die at E9.5 due to placental abnormalities. The mechanism by which this occurs is unknown. We demonstrated that the new endocrine factor EG-VEGF controls the same processes as those described for PPARγ, suggesting potential regulation of EG-VEGF by PPARγ. EG-VEGF exerts its functions via prokineticin receptor 1 (PROKR1) and 2 (PROKR2). This study sought to investigate whether EG-VEGF mediates part of PPARγ effects on placental development. Three approaches were used: 1) in vitro, using human primary isolated cytotrophoblasts and the extravillous trophoblast cell line (HTR-8/SVneo); 2) ex vivo, using human placental explants (n = 46 placentas); and 3) in vivo, using gravid wild-type PPARγ(+/-) and PPARγ(-/-) mice. Major processes of placental development that are known to be controlled by PPARγ, such as trophoblast proliferation, migration, and invasion, were assessed in the absence or presence of PROKR1 and PROKR2 antagonists. In both human trophoblast cell and placental explants, we demonstrated that rosiglitazone, a PPARγ agonist, 1) increased EG-VEGF secretion, 2) increased EG-VEGF and its receptors mRNA and protein expression, 3) increased placental vascularization via PROKR1 and PROKR2, and 4) inhibited trophoblast migration and invasion via PROKR2. In the PPARγ(-/-) mouse placentas, EG-VEGF levels were significantly decreased, supporting an in vivo control of EG-VEGF/PROKRs system during pregnancy. The present data reveal EG-VEGF as a new mediator of PPARγ effects during pregnancy and bring new insights into the fine mechanism of trophoblast invasion.


Asunto(s)
PPAR gamma/fisiología , Placentación , Resultado del Embarazo/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Animales , Benzamidas/farmacología , Células Cultivadas , Cricetinae , Implantación del Embrión/efectos de los fármacos , Implantación del Embrión/genética , Embrión de Mamíferos , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , Placenta/metabolismo , Embarazo , Piridinas/farmacología , Rosiglitazona , Tiazolidinedionas/farmacología , Activación Transcripcional/efectos de los fármacos , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo
6.
Sci Rep ; 11(1): 7847, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33846376

RESUMEN

The thioredoxin system plays key roles in regulating cancer cell malignancy. Here we identify the Thioredoxin-interacting protein (TXNIP) as a gene, which expression is regulated by PPARγ in melanoma cells. We show that high TXNIP expression levels associate with benign melanocytic lesions, with tumor regression in patients on MAP kinase targeted therapy, with decreased proliferation in patients' melanoma biopsies, and with cell cycle arrest in human melanoma cell lines. In contrast, reduced TXNIP expression associates with advanced melanoma and with disease progression in patients. TXNIP depletion in human melanoma cells altered the expression of integrin beta-3 and the localization of the integrin alpha-v/beta-3 dimer at their surface. Moreover, TXNIP depletion affected human melanoma cell motility and improved their capacity to colonize mouse lungs in an in vivo assay. This study establishes TXNIP as a PPARγ-regulated gene in melanoma cells, thereby suggesting a link between these two proteins both involved in the regulation of cancer and of energy metabolism. It also reveals that the decrease in TXNIP expression, which is observed in advanced patient tumors, likely favors lung metastatic seeding of malignant cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Neoplasias Pulmonares , Melanoma , PPAR gamma/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Melanoma/metabolismo , Melanoma/patología , Ratones
7.
Front Cell Dev Biol ; 9: 627153, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33869176

RESUMEN

Long bones from mammals host blood cell formation and contain multiple cell types, including adipocytes. Physiological functions of bone marrow adipocytes are poorly documented. Herein, we used adipocyte-deficient PPARγ-whole body null mice to investigate the consequence of total adipocyte deficiency on bone homeostasis in mice. We first highlighted the dual bone phenotype of PPARγ null mice: one the one hand, the increased bone formation and subsequent trabecularization extending in the long bone diaphysis, due to the well-known impact of PPARγ deficiency on osteoblasts formation and activity; on the other hand, an increased osteoclastogenesis in the cortical bone. We then further explored the cause of this unexpected increased osteoclastogenesis using two independent models of lipoatrophy, which recapitulated this phenotype. This demonstrates that hyperosteoclastogenesis is not intrinsically linked to PPARγ deficiency, but is a consequence of the total lipodystrophy. We further showed that adiponectin, a cytokine produced by adipocytes and mesenchymal stromal cells is a potent inhibitor of osteoclastogenesis in vitro and in vivo. Moreover, pharmacological activation of adiponectin receptors by the synthetic agonist AdipoRon inhibited mature osteoclast activity both in mouse and human cells by blocking podosome formation through AMPK activation. Finally, we demonstrated that AdipoRon treatment blocks bone erosion in vivo in a murine model of inflammatory bone loss, providing potential new approaches to treat osteoporosis.

8.
FEBS Open Bio ; 9(2): 328-334, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30761257

RESUMEN

Obesity is a condition characterized by adipose tissue hypertrophy; it is estimated that the obesity epidemic accounted for 4 million deaths in 2015 and that 70% of these were due to cardiovascular disease (CVD). One of the mechanisms linking obesity to CVD is the ability of adipose tissue to secrete circulating factors. We hypothesized that adipose tissue and its secretory products may influence mineralocorticoid receptor (MR) expression. Here, we showed that expression of MR and its downstream targets (Cnksr3, Scnn1b, and Sgk1) were significantly reduced in the kidneys of peroxisome proliferator-activated receptor-γ null (PpargΔ/Δ ) and A-ZIP/F-1 (AZIPtg/+) lipoatrophic mice with respect to their controls. Intriguingly, MR expression was also found to be significantly reduced in the kidneys of genetically obese ob/ob mice. Our data suggest that adipose tissue contributes to the regulation of MR expression. Given that leptin deficiency seems to be the major feature shared by PpargΔ/Δ , AZIPtg/+, and ob/ob mice, we speculate that adipose tissue modulates MR expression through the leptin system.


Asunto(s)
Lipodistrofia/metabolismo , Receptores de Mineralocorticoides/genética , Animales , Modelos Animales de Enfermedad , Femenino , Leptina/deficiencia , Leptina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Obesidad/metabolismo , Receptores de Mineralocorticoides/metabolismo
9.
Metabolism ; 95: 8-20, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30878493

RESUMEN

BACKGROUND: The peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-dependent transcription factor involved in many aspects of metabolism, immune response and development. Numerous studies relying on tissue-specific invalidation of the Pparg gene have shown distinct facets of its activity, whereas the effects of its systemic inactivation remain unexplored due to embryonic lethality. By maintaining PPARγ expression in the placenta, we recently generated a mouse model carrying Pparg full body deletion (PpargΔ/Δ), which in contrast to a previously published model is totally deprived of any form of adipose tissue. Herein, we propose an in-depth study of the metabolic alterations observed in this new model. METHODS: Young adult mice, both males and females analyzed separately, were first phenotyped for their gross anatomical alterations. Systemic metabolic parameters were analyzed in the blood, in static and in dynamic conditions. A full exploration of energy metabolism was performed in calorimetric cages as well as in metabolic cages. Our study was completed by expression analyses of a set of specific genes. MAIN FINDINGS: PpargΔ/Δ mice show a striking complete absence of any form of adipose tissue, which triggers a complex metabolic phenotype including increased lean mass with organomegaly, hypermetabolism, urinary energy loss, hyperphagia, and increased amino acid metabolism. PpargΔ/Δ mice develop severe type 2 diabetes, characterized by hyperglycemia, hyperinsulinemia, polyuria and polydispsia. They show a remarkable metabolic inflexibility, as indicated by the inability to shift substrate oxidation between glucose and lipids, in both ad libitum fed state and fed/fasted/refed transitions. Moreover, upon fasting PpargΔ/Δ mice enter a severe hypometabolic state. CONCLUSIONS: Our data comprehensively describe the impact of lipoatrophy on metabolic homeostasis. As such, the presented data on PpargΔ/Δ mice gives new clues on what and how to explore severe lipodystrophy and its subsequent metabolic complications in human.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Trastornos del Metabolismo de los Lípidos/genética , Tamaño de los Órganos/genética , PPAR gamma/genética , Tejido Adiposo/anatomía & histología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/genética , Femenino , Eliminación de Gen , Glucosa/metabolismo , Trastornos del Metabolismo de los Lípidos/metabolismo , Lipodistrofia/genética , Lipodistrofia/metabolismo , Ratones , Embarazo
10.
Front Immunol ; 9: 2573, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30483254

RESUMEN

Adult hematopoiesis takes place in the perivascular zone of the bone cavity, where endothelial cells, mesenchymal stromal/stem cells and their derivatives such as osteoblasts are key components of bone marrow (BM) niches. Defining the contribution of BM adipocytes to the hematopoietic stem cell niche remains controversial. While an excess of medullar adiposity is generally considered deleterious for hematopoiesis, an active role for adipocytes in shaping the niche has also been proposed. We thus investigated the consequences of total adipocyte deletion, including in the BM niche, on adult hematopoiesis using mice carrying a constitutive deletion of the gene coding for the nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ). We show that PpargΔ/Δ lipodystrophic mice exhibit severe extramedullary hematopoiesis (EMH), which we found to be non-cell autonomous, as it is reproduced when wild-type donor BM cells are transferred into PpargΔ/Δ recipients. This phenotype is not due to a specific alteration linked to Pparg deletion, such as chronic inflammation, since it is also found in AZIPtg/+ mice, another lipodystrophic mouse model with normal PPARγ expression, that display only very moderate levels of inflammation. In both models, the lack of adipocytes alters subpopulations of both myeloid and lymphoid cells. The CXCL12/CXCR4 axis in the BM is also dysregulated in an adipocyte deprived environment supporting the hypothesis that adipocytes are required for normal hematopoietic stem cell mobilization or retention. Altogether, these data suggest an important role for adipocytes, and possibly for the molecular interactions they provide within the BM, in maintaining the appropriate microenvironment for hematopoietic homeostasis.


Asunto(s)
Adipocitos/fisiología , Hematopoyesis/fisiología , Adipocitos/metabolismo , Adipogénesis/fisiología , Animales , Médula Ósea/metabolismo , Médula Ósea/fisiología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Huesos/metabolismo , Huesos/fisiología , Quimiocina CXCL12/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Femenino , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Osteoblastos/metabolismo , Osteoblastos/fisiología , PPAR gamma/metabolismo , Receptores CXCR4/metabolismo , Nicho de Células Madre/fisiología
11.
J Invest Dermatol ; 138(3): 500-510, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28964716

RESUMEN

PPARγ regulates multiple aspects of skin physiology, including sebocyte differentiation, keratinocyte proliferation, epithelial stem cell survival, adipocyte biology, and inflammatory skin responses. However, the effects of its global deletion, namely of nonredundant key functions of PPARγ signaling in mammalian skin, are yet unknown because of embryonic lethality. Here, we describe the skin and hair phenotype of a whole-body PPARγ-null mouse (PpargΔ/Δ), obtained by preserving PPARγ expression in the placenta. PpargΔ/Δ mice exhibited total lipoatrophy and complete absence of sebaceous glands. Right after birth, hair follicle (HF) morphogenesis was transiently delayed, along with reduced expression of HF differentiation markers and of transcriptional regulators necessary for HF development. Later, adult PpargΔ/Δ mice developed scarring alopecia and severe perifollicular inflammation. Skin analyses in other models of lipodystrophy, AZIPtg/+ and Adipoq-Cretg/+Ppargfl/fl mice, coupled with skin graft experiments, showed that the early defects observed in hair morphogenesis were caused by the absence of adipose tissue. In contrast, the late alteration of HF cycle and appearance of inflammation were observed only in PpargΔ/Δ mice and likely were due to the lack sebaceous glands. Our findings underscore the increasing appreciation for the importance of adipose tissue-mediated signals in HF development and function.


Asunto(s)
Folículo Piloso/crecimiento & desarrollo , Lipodistrofia/patología , Morfogénesis , PPAR gamma/fisiología , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Homeostasis , Ratones , Ratones Noqueados , PPAR gamma/genética
12.
PLoS One ; 12(2): e0171474, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28182703

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-dependent transcription factor involved in many aspects of metabolism, immune response, and development. Total-body deletion of the two Pparg alleles provoked generalized lipoatrophy along with severe type 2 diabetes. Herein, we explore the appearance and development of structural and functional alterations of the kidney, comparing Pparg null-mice to their littermate controls (carrying Pparg floxed alleles). We show that renal hypertrophy and functional alterations with increased glucosuria and albuminuria are already present in 3 weeks-old Pparg null-mice. Renal insufficiency with decreased creatinine clearance progress at 7 weeks of age, with the advance of the type 2 diabetes. At 52 weeks of age, these alterations are accompanied by signs of fibrosis and mesangial expansion. More intriguingly, aged Pparg null-mice concomitantly present an anti-phospholipid syndrome (APS), characterized by the late appearance of microthrombi and a mesangioproliferative pattern of glomerular injury, associated with significant plasmatic levels of anti-ß2- glycoprotein1 antibodies and renal deposition of IgG, IgM, and C3. Thus, in line with the role of PPARγ in metabolic homeostasis, Pparg null-mice first represent a potent model for studying the initiation and the development of diabetic nephropathy. Second, and in relation with the important PPARγ activity in inflammation and in immune system, these mice also highlight a new role for PPARγ signaling in the promotion of APS, a syndrome whose pathogenesis is poorly known and whose current treatment is limited to prevention of thrombosis events.


Asunto(s)
Síndrome Antifosfolípido/genética , Nefropatías Diabéticas/metabolismo , PPAR gamma/genética , Animales , Anticuerpos/inmunología , Síndrome Antifosfolípido/inmunología , Síndrome Antifosfolípido/patología , Complemento C3/inmunología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Homeostasis , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Ratones , PPAR gamma/metabolismo , Transducción de Señal , beta 2 Glicoproteína I/inmunología
13.
Environ Health Perspect ; 118(2): 234-41, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20123618

RESUMEN

BACKGROUND: The modulation of energetic homeostasis by pollutants has recently emerged as a potential contributor to the onset of metabolic disorders. Diethylhexyl phthalate (DEHP) is a widely used industrial plasticizer to which humans are widely exposed. Phthalates can activate the three peroxisome proliferator-activated receptor (PPAR) isotypes on cellular models and induce peroxisome proliferation in rodents. OBJECTIVES: In this study, we aimed to evaluate the systemic and metabolic consequences of DEHP exposure that have remained so far unexplored and to characterize the underlying molecular mechanisms of action. METHODS: As a proof of concept and mechanism, genetically engineered mouse models of PPARs were exposed to high doses of DEHP, followed by metabolic and molecular analyses. RESULTS: DEHP-treated mice were protected from diet-induced obesity via PPARalpha-dependent activation of hepatic fatty acid catabolism, whereas the activity of neither PPARbeta nor PPARgamma was affected. However, the lean phenotype observed in response to DEHP in wild-type mice was surprisingly abolished in PPARalpha-humanized mice. These species differences are associated with a different pattern of coregulator recruitment. CONCLUSION: These results demonstrate that DEHP exerts species-specific metabolic actions that rely to a large extent on PPARalpha signaling and highlight the metabolic importance of the species-specific activation of PPARalpha by xenobiotic compounds.


Asunto(s)
Dietilhexil Ftalato/farmacología , Metabolismo Energético/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Obesidad/prevención & control , PPAR alfa/metabolismo , Plastificantes/farmacología , Animales , Línea Celular , Ácidos Grasos/metabolismo , Prueba de Tolerancia a la Glucosa , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/inducido químicamente , Oxidación-Reducción/efectos de los fármacos , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA