Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters











Publication year range
1.
Cell ; 187(6): 1335-1342, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38490175

ABSTRACT

Gender inequality in STEM fields remains pervasive and undermines the ability for talented individuals to excel. Despite advances, women still encounter obstacles in pursuing academic careers and reaching leadership positions. This commentary discusses the "scissor-shaped curve" and examines effective strategies to fix it, including data-driven initiatives that we have implemented at our university.


Subject(s)
Academia , Gender Equity , Humans , Female , Leadership , Universities
2.
Nat Commun ; 12(1): 2538, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33953160

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) play a critical role in protection against helminths and in diverse inflammatory diseases by responding to soluble factors such as the alarmin IL-33, that is often overexpressed in cancer. Nonetheless, regulatory factors that dictate ILC2 functions remain poorly studied. Here, we show that peroxisome proliferator-activated receptor gamma (PPARγ) is selectively expressed in ILC2s in humans and in mice, acting as a central functional regulator. Pharmacologic inhibition or genetic deletion of PPARγ in ILC2s significantly impair IL-33-induced Type-2 cytokine production and mitochondrial fitness. Further, PPARγ blockade in ILC2s disrupts their pro-tumoral effect induced by IL-33-secreting cancer cells. Lastly, genetic ablation of PPARγ in ILC2s significantly suppresses tumor growth in vivo. Our findings highlight a crucial role for PPARγ in supporting the IL-33 dependent pro-tumorigenic role of ILC2s and suggest that PPARγ can be considered as a druggable pathway in ILC2s to inhibit their effector functions. Hence, PPARγ targeting might be exploited in cancer immunotherapy and in other ILC2-driven mediated disorders, such as asthma and allergy.


Subject(s)
Immunity, Innate/immunology , Interleukin-33/metabolism , Lymphocytes/metabolism , Neoplasms/therapy , PPAR gamma/metabolism , Animals , Asthma , Cytokines/pharmacology , Gene Knockdown Techniques , Humans , Hypersensitivity , Immunotherapy , Lymphocytes/drug effects , Mice , Mice, Inbred C57BL , Mitochondria , Neoplasms/pathology , PPAR gamma/genetics
3.
Sci Rep ; 11(1): 7847, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846376

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Lung Neoplasms , Melanoma , PPAR gamma/metabolism , Animals , Cell Line, Tumor , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma/metabolism , Melanoma/pathology , Mice
4.
Cancer Res ; 78(22): 6447-6461, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30185551

ABSTRACT

In addition to improving insulin sensitivity in type 2 diabetes, the thiazolidinedione family of compounds and the pharmacologic activation of their best-characterized target PPARγ have been proposed as a therapeutic option for cancer treatment. In this study, we reveal a new mode of action for the thiazolidinedione rosiglitazone that can contribute to tumorigenesis. Rosiglitazone activated a tumorigenic paracrine communication program in a subset of human melanoma cells that involves the secretion of cytokines, chemokines, and angiogenic factors. This complex blend of paracrine signals activated nonmalignant fibroblasts, endothelial cells, and macrophages in a tumor-friendly way. In agreement with these data, rosiglitazone promoted human melanoma development in xenografts, and tumors exposed to rosiglitazone exhibited enhanced angiogenesis and inflammation. Together, these findings establish an important tumorigenic action of rosiglitazone in a subset of melanoma cells. Although studies conducted on cohorts of diabetic patients report overall benefits of thiazolidinediones in cancer prevention, our data suggest that exposure of established tumors to rosiglitazone may be deleterious.Significance: These findings uncover a novel mechanism by which the thiazolidinedione compound rosiglitazone contributes to tumorigenesis, thus highlighting a potential risk associated with its use in patients with established tumors. Cancer Res; 78(22); 6447-61. ©2018 AACR.


Subject(s)
Melanoma/metabolism , PPAR gamma/agonists , Rosiglitazone/pharmacology , Skin Neoplasms/metabolism , Stromal Cells/metabolism , Angiogenesis Inducing Agents/metabolism , Animals , Carcinogenesis , Cell Line, Tumor , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Inflammation , Leukocytes, Mononuclear/cytology , Macrophages/drug effects , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Monocytes/metabolism , Neoplasm Metastasis , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , PPAR gamma/metabolism , Paracrine Communication , Skin Neoplasms/pathology , T-Lymphocytes/cytology
5.
J Invest Dermatol ; 138(3): 500-510, 2018 03.
Article in English | MEDLINE | ID: mdl-28964716

ABSTRACT

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.


Subject(s)
Hair Follicle/growth & development , Lipodystrophy/pathology , Morphogenesis , PPAR gamma/physiology , Animals , Cell Differentiation , Disease Models, Animal , Homeostasis , Mice , Mice, Knockout , PPAR gamma/genetics
6.
Oncotarget ; 9(102): 37614-37615, 2018 Dec 28.
Article in English | MEDLINE | ID: mdl-30701018
7.
EMBO Mol Med ; 8(8): 919-36, 2016 08.
Article in English | MEDLINE | ID: mdl-27250636

ABSTRACT

Although excessive exposure to UV is widely recognized as a major factor leading to skin perturbations and cancer, the complex mechanisms underlying inflammatory skin disorders resulting from UV exposure remain incompletely characterized. The nuclear hormone receptor PPARß/δ is known to control mouse cutaneous repair and UV-induced skin cancer development. Here, we describe a novel PPARß/δ-dependent molecular cascade involving TGFß1 and miR-21-3p, which is activated in the epidermis in response to UV exposure. We establish that the passenger miRNA miR-21-3p, that we identify as a novel UV-induced miRNA in the epidermis, plays a pro-inflammatory function in keratinocytes and that its high level of expression in human skin is associated with psoriasis and squamous cell carcinomas. Finally, we provide evidence that inhibition of miR-21-3p reduces UV-induced cutaneous inflammation in ex vivo human skin biopsies, thereby underlining the clinical relevance of miRNA-based topical therapies for cutaneous disorders.


Subject(s)
MicroRNAs/metabolism , PPAR delta/metabolism , PPAR-beta/metabolism , Radiodermatitis/pathology , Signal Transduction , Skin/radiation effects , Ultraviolet Rays , Animals , Humans , Mice
8.
J Allergy Clin Immunol ; 135(6): 1625-35.e5, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25556996

ABSTRACT

BACKGROUND: Remodeling of quiescent vessels with increases in permeability, vasodilatation, and edema are hallmarks of inflammatory disorders. Factors involved in this type of remodeling represent potential therapeutic targets. OBJECTIVES: We investigated whether the nuclear hormone receptor peroxisome proliferator-activated receptor (PPAR) ß/δ, a regulator of metabolism, fibrosis, and skin homeostasis, is involved in regulation of this type of remodeling. METHODS: Wild-type and various Pparb/d mutant mice were used to monitor dermal acute vascular hyperpermeability (AVH) and passive systemic anaphylaxis-induced hypothermia and edema. PPARß/δ-dependent kinase activation and remodeling of endothelial cell-cell junctions were addressed by using human endothelial cells. RESULTS: AVH and dilatation of dermal microvessels stimulated by vascular endothelial growth factor A, histamine, and thrombin are severely compromised in PPARß/δ-deficient mice. Selective deletion of the Pparb/d-encoding gene in endothelial cells in vivo similarly limits dermal AVH and vasodilatation, providing evidence that endothelial PPARß/δ is the major player in regulating acute dermal microvessel remodeling. Furthermore, endothelial PPARß/δ regulatory functions are not restricted to the skin vasculature because its deletion in the endothelium, but not in smooth muscle cells, also leads to reduced systemic anaphylaxis, the most severe form of allergic reaction, in which an acute vascular response plays a key role. PPARß/δ-dependent AVH activation likely involves the activation of mitogen-activated protein kinase and Akt pathways and leads to downstream destabilization of endothelial cell-cell junctions. CONCLUSION: These results unveil not only a novel function of PPARß/δ as a direct regulator of acute vessel permeability and dilatation but also provide evidence that antagonizing PPARß/δ represents an important strategy to consider for moderating diseases with altered endothelial integrity, such as acute inflammatory and allergic disorders.


Subject(s)
Anaphylaxis/immunology , Capillary Permeability/immunology , Endothelial Cells/immunology , PPAR delta/immunology , PPAR-beta/immunology , Skin/immunology , Anaphylaxis/genetics , Anaphylaxis/pathology , Animals , Capillary Permeability/drug effects , Edema/genetics , Edema/immunology , Edema/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Gene Expression Regulation , Histamine/pharmacology , Hypothermia/genetics , Hypothermia/immunology , Hypothermia/pathology , Intercellular Junctions/drug effects , Intercellular Junctions/immunology , Intercellular Junctions/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , PPAR delta/deficiency , PPAR delta/genetics , PPAR-beta/deficiency , PPAR-beta/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction , Skin/blood supply , Skin/drug effects , Skin/pathology , Thrombin/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
9.
Diabetologia ; 57(10): 2126-35, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25063273

ABSTRACT

AIM/HYPOTHESIS: Endoplasmic reticulum (ER) stress, which is involved in the link between inflammation and insulin resistance, contributes to the development of type 2 diabetes mellitus. In this study, we assessed whether peroxisome proliferator-activated receptor (PPAR)ß/δ prevented ER stress-associated inflammation and insulin resistance in skeletal muscle cells. METHODS: Studies were conducted in mouse C2C12 myotubes, in the human myogenic cell line LHCN-M2 and in skeletal muscle from wild-type and PPARß/δ-deficient mice and mice exposed to a high-fat diet. RESULTS: The PPARß/δ agonist GW501516 prevented lipid-induced ER stress in mouse and human myotubes and in skeletal muscle of mice fed a high-fat diet. PPARß/δ activation also prevented thapsigargin- and tunicamycin-induced ER stress in human and murine skeletal muscle cells. In agreement with this, PPARß/δ activation prevented ER stress-associated inflammation and insulin resistance, and glucose-intolerant PPARß/δ-deficient mice showed increased phosphorylated levels of inositol-requiring 1 transmembrane kinase/endonuclease-1α in skeletal muscle. Our findings demonstrate that PPARß/δ activation prevents ER stress through the activation of AMP-activated protein kinase (AMPK), and the subsequent inhibition of extracellular-signal-regulated kinase (ERK)1/2 due to the inhibitory crosstalk between AMPK and ERK1/2, since overexpression of a dominant negative AMPK construct (K45R) reversed the effects attained by PPARß/δ activation. CONCLUSIONS/INTERPRETATION: Overall, these findings indicate that PPARß/δ prevents ER stress, inflammation and insulin resistance in skeletal muscle cells by activating AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endoplasmic Reticulum Stress/physiology , Inflammation/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , PPAR delta/physiology , PPAR-beta/physiology , Animals , Cell Line , Diet, High-Fat/adverse effects , Endoplasmic Reticulum Stress/genetics , Humans , In Vitro Techniques , Inflammation/etiology , Inflammation/genetics , Insulin Resistance/genetics , Mice , Muscle Fibers, Skeletal/metabolism , PPAR delta/deficiency , PPAR delta/genetics , PPAR-beta/deficiency , PPAR-beta/genetics
10.
Int J Cardiol ; 174(1): 110-8, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24767130

ABSTRACT

BACKGROUND: Chronic endoplasmic reticulum (ER) stress contributes to the apoptotic cell death in the myocardium, thereby playing a critical role in the development of cardiomyopathy. ER stress has been reported to be induced after high-fat diet feeding in mice and also after saturated fatty acid treatment in vitro. Therefore, since several studies have shown that peroxisome proliferator-activated receptor (PPAR)ß/δ inhibits ER stress, the main goal of this study consisted in investigating whether activation of this nuclear receptor was able to prevent lipid-induced ER stress in cardiac cells. METHODS AND RESULTS: Wild-type and transgenic mice with reduced PPARß/δ expression were fed a standard diet or a high-fat diet for two months. For in vitro studies, a cardiomyocyte cell line of human origin, AC16, was treated with palmitate and the PPARß/δ agonist GW501516. Our results demonstrate that palmitate induced ER stress in AC16 cells, a fact which was prevented after PPARß/δ activation with GW501516. Interestingly, the effect of GW501516 on ER stress occurred in an AMPK-independent manner. The most striking result of this study is that GW501516 treatment also upregulated the protein levels of beclin 1 and LC3II, two well-known markers of autophagy. In accordance with this, feeding on a high-fat diet or suppression of PPARß/δ in knockout mice induced ER stress in the heart. Moreover, PPARß/δ knockout mice also displayed a reduction in autophagic markers. CONCLUSION: Our data indicate that PPARß/δ activation might be useful to prevent the harmful effects of ER stress induced by saturated fatty acids in the heart by inducing autophagy.


Subject(s)
Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Myocytes, Cardiac/drug effects , PPAR delta/pharmacology , PPAR-beta/pharmacology , Palmitates/pharmacology , Animals , Cells, Cultured , Humans , Male , Mice , Mice, Knockout , Thiazoles/pharmacology
11.
EMBO Mol Med ; 6(1): 80-98, 2014 01.
Article in English | MEDLINE | ID: mdl-24203162

ABSTRACT

Although non-melanoma skin cancer (NMSC) is the most common human cancer and its incidence continues to rise worldwide, the mechanisms underlying its development remain incompletely understood. Here, we unveil a cascade of events involving peroxisome proliferator-activated receptor (PPAR) ß/δ and the oncogene Src, which promotes the development of ultraviolet (UV)-induced skin cancer in mice. UV-induced PPARß/δ activity, which directly stimulated Src expression, increased Src kinase activity and enhanced the EGFR/Erk1/2 signalling pathway, resulting in increased epithelial-to-mesenchymal transition (EMT) marker expression. Consistent with these observations, PPARß/δ-null mice developed fewer and smaller skin tumours, and a PPARß/δ antagonist prevented UV-dependent Src stimulation. Furthermore, the expression of PPARß/δ positively correlated with the expression of SRC and EMT markers in human skin squamous cell carcinoma (SCC), and critically, linear models applied to several human epithelial cancers revealed an interaction between PPARß/δ and SRC and TGFß1 transcriptional levels. Taken together, these observations motivate the future evaluation of PPARß/δ modulators to attenuate the development of several epithelial cancers.


Subject(s)
Carcinoma, Squamous Cell/pathology , PPAR delta/metabolism , PPAR-beta/metabolism , Skin Neoplasms/pathology , Skin/radiation effects , Ultraviolet Rays , src-Family Kinases/metabolism , Animals , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/metabolism , Enzyme Activation , Epithelial-Mesenchymal Transition/radiation effects , Female , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Mice , Mice, Hairless , Mice, Knockout , PPAR delta/antagonists & inhibitors , PPAR delta/genetics , PPAR-beta/antagonists & inhibitors , PPAR-beta/genetics , RNA, Messenger/metabolism , Signal Transduction/radiation effects , Skin/metabolism , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , src-Family Kinases/genetics
12.
Biochim Biophys Acta ; 1832(8): 1241-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23507144

ABSTRACT

The role of peroxisome proliferator activator receptor (PPAR)ß/δ in the pathogenesis of Alzheimer's disease has only recently been explored through the use of PPARß/δ agonists. Here we evaluated the effects of PPARß/δ deficiency on the amyloidogenic pathway and tau hyperphosphorylation. PPARß/δ-null mice showed cognitive impairment in the object recognition task, accompanied by enhanced DNA-binding activity of NF-κB in the cortex and increased expression of IL-6. In addition, two NF-κB-target genes involved in ß-amyloid (Aß) synthesis and deposition, the ß site APP cleaving enzyme 1 (Bace1) and the receptor for advanced glycation endproducts (Rage), respectively, increased in PPARß/δ-null mice compared to wild type animals. The protein levels of glial fibrillary acidic protein (GFAP) increased in the cortex of PPARß/δ-null mice, which would suggest the presence of astrogliosis. Finally, tau hyperphosphorylation at Ser199 and enhanced levels of PHF-tau were associated with increased levels of the tau kinases CDK5 and phospho-ERK1/2 in the cortex of PPARß/δ(-/-) mice. Collectively, our findings indicate that PPARß/δ deficiency results in cognitive impairment associated with enhanced inflammation, astrogliosis and tau hyperphosphorylation in the cortex.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Cerebral Cortex/metabolism , PPAR-beta/deficiency , Receptors, Immunologic/metabolism , tau Proteins/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Cognition/physiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glial Fibrillary Acidic Protein , Inflammation , Interleukin-6/genetics , Interleukin-6/metabolism , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PPAR-beta/genetics , PPAR-beta/metabolism , Phosphorylation , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , tau Proteins/genetics
13.
Clín. investig. arterioscler. (Ed. impr.) ; 24(6): 275-283, nov.-dic. 2012. ilus
Article in Spanish | IBECS | ID: ibc-106335

ABSTRACT

Introducción: La resistencia a la insulina precede y predice la presencia de diabetes mellitustipo 2, condición que supone un notable incremento del riesgo cardiovascular. La interleucina-6es uno de los mediadores que relacionan la inflamación crónica observada en estados de obesidad con la resistencia a la insulina a través de la activación de STAT3 (signal transducer and activator of transcription 3), con el consiguiente aumento de SOCS3 (suppressor of cytokinesignaling 3) en el hígado. El objetivo de este estudio ha sido evaluar si un agonista del receptor activado por proliferadores peroxisómicos (PPAR) / , GW501516, es capaz de evitar la activación de la vía de senalización IL-6/STAT3/SOCS3 y la resistencia a la insulina en células hepáticas. Material y métodos: Células HepG2 humanas se estimularon con IL-6 (20 ng/ml) en presenciao en ausencia de GW501516 (10 M). También analizamos el hígado de ratones salvajes y con deficiencia PPAR / . Los niveles de ARNm y proteínas se analizaron mediante las técnicas deRT-PCR y Western-Blot, respectivamente. Resultados: GW501516 evitó la fosforilación en Tyr705 y en Ser727 de STAT3 y el aumento deSOCS3 inducidas por la IL-6. Asimismo, el tratamiento con este fármaco evitó la activación por la IL-6 de la ERK1/2, una serina-treonina cinasa implicada en la fosforilación de STAT3 enSer727. Cabe destacar que el hígado de ratones deficientes en PPAR / mostró un aumento (..) (AU)


Introduction: Insulin resistance precedes and predicts the development of type 2 diabetes mellitus, a disease which increases the risk of cardiovascular events. Interleukin (IL)-6 is one of the mediators linking obesity-derived chronic inflammation with insulin resistance through activation of STAT3, with subsequent up regulation of suppressor of cytokine signaling 3 (SOCS3)in liver. The aim of this study was to evaluate whether peroxisome proliferator-activated receptor (PPAR) / agonist GW501516 prevented activation of the IL-6/STAT3/SOCS3 pathway and insulin resistance in hepatic cells. Material and methods: Human HepG2 cells were stimulated for 10 min with IL-6 (20 ng/mL)in the presence or in the absence of 10 M GW501516, then mRNA and protein levels were analyzed by RT-PCR or Western-Blot, respectively. In addition, we also analyzed protein levels from PPAR / null mice and wild-type mice livers. Results: GW501516 prevented IL-6-induced STAT3 phosphorylation on Tyr705 and Ser727 and avoided the increase in SOCS3 caused by this cytokine. In addition, this drug also preventedIL-6-dependent ERK1/2 phosphorylation, a serine-threonine protein kinase involved in STAT3phosphorylation on Ser727. Interestingly, livers from PPAR / null mice showed increased phosphorylations on Tyr 705 and Ser727 of STAT3 as well as phosphorylated ERK1/2 levels. Finally, all (..) (AU)


Subject(s)
Humans , Peroxisome Proliferators/pharmacokinetics , Insulin Resistance , Hepatocytes/metabolism , Biotransformation/physiology , Interleukin-6/pharmacokinetics , Oxidative Phosphorylation , Cytokines/pharmacokinetics
14.
Trends Endocrinol Metab ; 23(7): 351-63, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22704720

ABSTRACT

Nuclear receptors (NRs) are ligand-dependent transcription factors whose activation affects genes controlling vital processes. Among them, the peroxisome proliferator-activated receptors (PPARs) have emerged as links between lipids, metabolic diseases, and innate immunity. PPARs are activated by fatty acids and their derivatives, many of which also signal through membrane receptors, thereby creating a lipid signaling network between the cell surface and the nucleus. Tissues that play a role in whole-body metabolic homeostasis, such as adipose tissue, liver, skeletal muscle, intestines, and blood vessel walls, are prone to inflammation when metabolism is disturbed, a complication that promotes type 2 diabetes and cardiovascular disease. This review discusses the protective roles of PPARs in inflammatory conditions and the therapeutic anti-inflammatory potential of PPAR ligands.


Subject(s)
Inflammation/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Humans , Inflammation/genetics , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Peroxisome Proliferator-Activated Receptors/genetics
15.
Clín. investig. arterioscler. (Ed. impr.) ; 24(3): 131-140, mayo-jun. 2012. ilus, tab
Article in Spanish | IBECS | ID: ibc-105086

ABSTRACT

Introducción El consumo de dietas ricas en grasas se relaciona con alteraciones cardíacas caracterizadas por un proceso inflamatorio de baja intensidad mediado por NF-κB. PPARbeta/delta ha sido propuesto como potencial diana terapéutica para paliar el proceso inflamatorio asociado a alteraciones cardiovasculares. Sin embargo, se desconoce la implicación de este receptor en la respuesta inflamatoria inducida por lípidos en el corazón (..) (AU)


Introduction High-fat diet intake is associated with cardiac disorders characterised by a low-grade inflammatory process which involves NF-κB activation. PPARbeta/delta has been proposed as a potential therapeutic target to mitigate the inflammatory process related to cardiovascular disorders. However, the involvement of this receptor in lipid-induced inflammatory response in the heart is not yet known (..) (AU)


Subject(s)
Animals , Mice , PPAR-beta/pharmacokinetics , Inflammation/physiopathology , Inflammation Mediators/analysis , Lipids/adverse effects , NF-kappa B/physiology , Cytokines , Chemokines , Palmitates/pharmacokinetics , Mice, Knockout
16.
Article in English | MEDLINE | ID: mdl-21778043

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are fatty acid-activated transcription factors belonging to the nuclear hormone receptor family. While PPARs are best known as regulators of energy homeostasis, evidence also has accumulated recently for their involvement in basic cellular functions. We review novel insights into PPAR functions in skin wound healing and liver, with emphasis on PPARß/δ and PPARα, respectively. Activation of PPARß/δ expression in response to injury promotes keratinocyte survival, directional sensing, and migration over the wound bed. In addition, interleukin (IL)-1 produced by the keratinocytes activates PPARß/δ expression in the underlying fibroblasts, which hinders the mitotic activity of keratinocytes via inhibition of IL-1 signaling. Initially, roles were identified for PPARα in fatty acid catabolism. However, PPARα is also involved in downregulating many genes in female mammals. We have elucidated the mechanism of this repression, which requires sumoylation of PPARα. Physiologically, this control confers protection against estrogen-induced intrahepatic cholestasis.


Subject(s)
Energy Metabolism , Fatty Acids/metabolism , Liver/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Skin/metabolism , Wound Healing , Animals , Cholestasis, Intrahepatic/etiology , Cholestasis, Intrahepatic/metabolism , Cholestasis, Intrahepatic/pathology , Estrogens/metabolism , Female , Humans , Interleukin-1/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Sex Characteristics , Signal Transduction , Skin/pathology
17.
Diabetes ; 60(7): 1990-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21617181

ABSTRACT

OBJECTIVE: It has been suggested that interleukin (IL)-6 is one of the mediators linking obesity-derived chronic inflammation with insulin resistance through activation of STAT3, with subsequent upregulation of suppressor of cytokine signaling 3 (SOCS3). We evaluated whether peroxisome proliferator-activated receptor (PPAR)-ß/-δ prevented activation of the IL-6-STAT3-SOCS3 pathway and insulin resistance in adipocytes. RESEARCH DESIGN AND METHODS: Adipocytes and white adipose tissue from wild-type and PPAR-ß/-δ-null mice were used to evaluate the effect of PPAR-ß/-δ on the IL-6-STAT3-SOCS3 pathway. RESULTS: First, we observed that the PPAR-ß/-δ agonist GW501516 prevented both IL-6-dependent reduction in insulin-stimulated Akt phosphorylation and glucose uptake in adipocytes. In addition, this drug treatment abolished IL-6-induced SOCS3 expression in differentiated 3T3-L1 adipocytes. This effect was associated with the capacity of the drug to prevent IL-6-induced STAT3 phosphorylation on Tyr(705) and Ser(727) residues in vitro and in vivo. Moreover, GW501516 prevented IL-6-dependent induction of extracellular signal-related kinase (ERK)1/2, a serine-threonine-protein kinase involved in serine STAT3 phosphorylation. Furthermore, in white adipose tissue from PPAR-ß/-δ-null mice, STAT3 phosphorylation (Tyr(705) and Ser(727)), STAT3 DNA-binding activity, and SOCS3 protein levels were higher than in wild-type mice. Several steps in STAT3 activation require its association with heat shock protein 90 (Hsp90), which was prevented by GW501516 as revealed in immunoprecipitation studies. Consistent with this finding, the STAT3-Hsp90 association was enhanced in white adipose tissue from PPAR-ß/-δ-null mice compared with wild-type mice. CONCLUSIONS: Collectively, our findings indicate that PPAR-ß/-δ activation prevents IL-6-induced STAT3 activation by inhibiting ERK1/2 and preventing the STAT3-Hsp90 association, an effect that may contribute to the prevention of cytokine-induced insulin resistance in adipocytes.


Subject(s)
Adipocytes/metabolism , Insulin/physiology , PPAR delta/metabolism , PPAR-beta/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipose Tissue, White/metabolism , Animals , Glucose/metabolism , HSP90 Heat-Shock Proteins/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/pharmacology , Male , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , PPAR delta/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Rats , Thiazoles/pharmacology
18.
PLoS One ; 6(2): e14665, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21326871

ABSTRACT

Reactive oxygen species, ROS, are regulators of endothelial cell migration, proliferation and survival, events critically involved in angiogenesis. Different isoforms of ROS-generating NOX enzymes are expressed in the vasculature and provide distinct signaling cues through differential localization and activation. We show that mice deficient in NOX1, but not NOX2 or NOX4, have impaired angiogenesis. NOX1 expression and activity is increased in primary mouse and human endothelial cells upon angiogenic stimulation. NOX1 silencing decreases endothelial cell migration and tube-like structure formation, through the inhibition of PPARα, a regulator of NF-κB. Administration of a novel NOX-specific inhibitor reduced angiogenesis and tumor growth in vivo in a PPARα dependent manner. In conclusion, vascular NOX1 is a critical mediator of angiogenesis and an attractive target for anti-angiogenic therapies.


Subject(s)
Endothelial Cells/metabolism , NADH, NADPH Oxidoreductases/genetics , Neoplasms/blood supply , Neovascularization, Pathologic/genetics , PPAR alpha/physiology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Cells, Cultured , Endothelial Cells/drug effects , Female , Gene Knockdown Techniques , Gene Targeting , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Targeted Therapy , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/physiology , NADPH Oxidase 1 , Neoplasms/drug therapy , Neoplasms/genetics , Neovascularization, Pathologic/drug therapy , PPAR alpha/genetics , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
19.
Biochim Biophys Acta ; 1811(2): 59-67, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21070867

ABSTRACT

Owing to its high fat content, the classical Western diet has a range of adverse effects on the heart, including enhanced inflammation, hypertrophy, and contractile dysfunction. Proinflammatory factors secreted by cardiac cells, which are under the transcriptional control of nuclear factor-κB (NF-κB), may contribute to heart failure and dilated cardiomyopathy. The underlying mechanisms are complex, since they are linked to systemic metabolic abnormalities and changes in cardiomyocyte phenotype. Peroxisome proliferator-activated receptors (PPARs) are transcription factors that regulate metabolism and are capable of limiting myocardial inflammation and hypertrophy via inhibition of NF-κB. Since PPARß/δ is the most prevalent PPAR isoform in the heart, we analyzed the effects of the PPARß/δ agonist GW501516 on inflammatory parameters. A high-fat diet induced the expression of tumor necrosis factor-α, monocyte chemoattractant protein-1, and interleukin-6, and enhanced the activity of NF-κB in the heart of mice. GW501516 abrogated this enhanced proinflammatory profile. Similar results were obtained when human cardiac AC16 cells exposed to palmitate were coincubated with GW501516. PPARß/δ activation by GW501516 enhanced the physical interaction between PPARß/δ and p65, which suggests that this mechanism may also interfere NF-κB transactivation capacity in the heart. GW501516-induced PPARß/δ activation can attenuate the inflammatory response induced in human cardiac AC16 cells exposed to the saturated fatty acid palmitate and in mice fed a high-fat diet. This is relevant, especially taking into account that PPARß/δ has been postulated as a potential target in the treatment of obesity and the insulin resistance state.


Subject(s)
Heart/drug effects , Lipids/pharmacology , PPAR delta/metabolism , PPAR-beta/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Cells, Cultured , Dietary Fats/adverse effects , Dietary Fats/metabolism , Humans , Inflammation/immunology , Mice , Mice, Knockout , Myocardium/immunology , PPAR delta/agonists , PPAR-beta/agonists , Thiazoles/metabolism , Transcription Factor RelA/metabolism
20.
Mol Cell Biol ; 29(22): 6006-17, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19752192

ABSTRACT

Transforming growth factor beta (TGF-beta) and platelet-derived growth factor A (PDGFAlpha) play a central role in tissue morphogenesis and repair, but their interplay remain poorly understood. The nuclear factor I C (NFI-C) transcription factor has been implicated in TGF-beta signaling, extracellular matrix deposition, and skin appendage pathologies, but a potential role in skin morphogenesis or healing had not been assessed. To evaluate this possibility, we performed a global gene expression analysis in NFI-C(-/-) and wild-type embryonic primary murine fibroblasts. This indicated that NFI-C acts mostly to repress gene expression in response to TGF-beta1. Misregulated genes were prominently overrepresented by regulators of connective tissue inflammation and repair. In vivo skin healing revealed a faster inflammatory stage and wound closure in NFI-C(-/-) mice. Expression of PDGFA and PDGF-receptor alpha were increased in wounds of NFI-C(-/-) mice, explaining the early recruitment of macrophages and fibroblasts. Differentiation of fibroblasts to contractile myofibroblasts was also elevated, providing a rationale for faster wound closure. Taken together with the role of TGF-beta in myofibroblast differentiation, our results imply a central role of NFI-C in the interplay of the two signaling pathways and in regulation of the progression of tissue regeneration.


Subject(s)
NFI Transcription Factors/metabolism , Platelet-Derived Growth Factor/metabolism , Signal Transduction , Skin/pathology , Transforming Growth Factor beta1/metabolism , Wound Healing , Animals , Cell Differentiation , Cell Movement , Cells, Cultured , Embryo, Mammalian/cytology , Extracellular Matrix/genetics , Extracellular Matrix Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Deletion , Gene Expression Regulation , Macrophages/cytology , Mice , Mice, Knockout , NFI Transcription Factors/deficiency , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL