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.
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
3.
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
4.
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
5.
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
6.
Curr Opin Cell Biol ; 17(2): 216-22, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15780600

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors involved in lipid and glucose homeostasis, inflammation and wound healing. In addition to ligand binding, phosphorylation can also regulate PPARs; the biological effects of phosphorylation depend on the stimulus, the kinase, the PPAR isotype, the residue modified, the cell type and the promoter investigated. The study of this dual regulation mode, which allows PPARs to integrate signals conveyed by lipophilic ligands with those coming from the plasma membrane, may ultimately offer new therapeutic strategies.


Subject(s)
Cell Nucleus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Phosphotransferases/metabolism , Signal Transduction/physiology , Animals , Energy Metabolism/physiology , Humans , Phosphorylation , Transcriptional Activation/physiology
7.
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
8.
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
9.
Mol Cell Biol ; 27(20): 7161-75, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17682064

ABSTRACT

After an injury, keratinocytes acquire the plasticity necessary for the reepithelialization of the wound. Here, we identify a novel pathway by which a nuclear hormone receptor, until now better known for its metabolic functions, potentiates cell migration. We show that peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) enhances two phosphatidylinositol 3-kinase-dependent pathways, namely, the Akt and the Rho-GTPase pathways. This PPARbeta/delta activity amplifies the response of keratinocytes to a chemotactic signal, promotes integrin recycling and remodeling of the actin cytoskeleton, and thereby favors cell migration. Using three-dimensional wound reconstructions, we demonstrate that these defects have a strong impact on in vivo skin healing, since PPARbeta/delta-/- mice show an unexpected and rare epithelialization phenotype. Our findings demonstrate that nuclear hormone receptors not only regulate intercellular communication at the organism level but also participate in cell responses to a chemotactic signal. The implications of our findings may be far-reaching, considering that the mechanisms described here are important in many physiological and pathological situations.


Subject(s)
Cell Movement/physiology , Cell Polarity , Chemotaxis/physiology , PPAR delta/metabolism , PPAR-beta/metabolism , Signal Transduction/physiology , Actins/metabolism , Animals , Cells, Cultured , Enzyme Activation , Epidermal Growth Factor/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Mice , Mice, Knockout , PPAR delta/genetics , PPAR-beta/genetics , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stress Fibers/metabolism , Wound Healing , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
10.
J Clin Invest ; 116(3): 598-606, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16511592

ABSTRACT

Tissue damage resulting from chemical, mechanical, and biological injury, or from interrupted blood flow and reperfusion, is often life threatening. The subsequent tissue response involves an intricate series of events including inflammation, oxidative stress, immune cell recruitment, and cell survival, proliferation, migration, and differentiation. In addition, fibrotic repair characterized by myofibroblast transdifferentiation and the deposition of ECM proteins is activated. Failure to initiate, maintain, or stop this repair program has dramatic consequences, such as cell death and associated tissue necrosis or carcinogenesis. In this sense, inflammation and oxidative stress, which are beneficial defense processes, can become harmful if they do not resolve in time. This repair program is largely based on rapid and specific changes in gene expression controlled by transcription factors that sense injury. PPARs are such factors and are activated by lipid mediators produced after wounding. Here we highlight advances in our understanding of PPAR action during tissue repair and discuss the potential for these nuclear receptors as therapeutic targets for tissue injury.


Subject(s)
Ischemia/metabolism , Peroxisome Proliferator-Activated Receptors/physiology , Wound Healing/physiology , Animals , Humans , Ischemia/physiopathology
11.
Biochim Biophys Acta ; 1771(8): 991-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17400022

ABSTRACT

Peroxisome proliferator-activated receptors, PPARalpha, PPARbeta/delta and PPARgamma, are fatty acid activated transcription factors that belong to the nuclear hormone receptor family. While they are best known as transcriptional regulators of lipid and glucose metabolism, evidence has also accumulated for their importance in skin homeostasis. The three PPAR isotypes are expressed in rodent and human skin. Various cell culture and in vivo approaches suggest that PPARalpha contributes to fetal skin development, to epidermal barrier maturation and to sebocyte activity. PPARbeta/delta regulates sebocyte differentiation, promotes hair follicle growth and has pro-differentiating effects in keratinocytes in normal and inflammatory conditions. In contrast, the role of PPARgamma appears to be rather minor in keratinocytes, whereas its activity is required for sebaceous gland differentiation. Importantly, PPARalpha and beta/delta are instrumental in skin repair after an injury, each of them playing specific roles. Due to their collective diverse functions in skin biology, PPARs represent a major research target for the understanding and treatment of many skin diseases, such as benign epidermal tumors, papillomas, acne vulgaris and psoriasis.


Subject(s)
Peroxisome Proliferator-Activated Receptors/physiology , Skin Diseases/physiopathology , Skin Physiological Phenomena , Epidermis/physiology , Fetal Development , Hair Follicle/physiology , Humans , Melanocytes/physiology , PPAR alpha/physiology , PPAR delta/physiology , PPAR gamma/physiology , PPAR-beta/physiology , Skin/cytology , Skin/pathology , Skin/physiopathology , Skin Neoplasms/physiopathology
12.
Mol Cell Biol ; 25(5): 1696-712, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15713628

ABSTRACT

Hair follicle morphogenesis depends on a delicate balance between cell proliferation and apoptosis, which involves epithelium-mesenchyme interactions. We show that peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) and Akt1 are highly expressed in follicular keratinocytes throughout hair follicle development. Interestingly, PPARbeta/delta- and Akt1-deficient mice exhibit similar retardation of postnatal hair follicle morphogenesis, particularly at the hair peg stage, revealing a new important function for both factors in the growth of early hair follicles. We demonstrate that a time-regulated activation of the PPARbeta/delta protein in follicular keratinocytes involves the up-regulation of the cyclooxygenase 2 enzyme by a mesenchymal paracrine factor, the hepatocyte growth factor. Subsequent PPARbeta/delta-mediated temporal activation of the antiapoptotic Akt1 pathway in vivo protects keratinocytes from hair pegs against apoptosis, which is required for normal hair follicle development. Together, these results demonstrate that epithelium-mesenchyme interactions in the skin regulate the activity of PPARbeta/delta during hair follicle development via the control of ligand production and provide important new insights into the molecular biology of hair growth.


Subject(s)
Hair Follicle/growth & development , Mesoderm/physiology , PPAR delta/metabolism , PPAR-beta/metabolism , Paracrine Communication/physiology , Animals , Apoptosis/physiology , Cyclooxygenase 2 , Epithelium/chemistry , Epithelium/growth & development , Epithelium/physiology , Hair Follicle/chemistry , Hair Follicle/cytology , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/physiology , Intercellular Signaling Peptides and Proteins/physiology , Keratinocytes/chemistry , Keratinocytes/cytology , Keratinocytes/physiology , Mice , Mice, Knockout , Models, Biological , Morphogenesis/genetics , Morphogenesis/physiology , PPAR delta/genetics , PPAR delta/physiology , PPAR-beta/genetics , PPAR-beta/physiology , Phosphorylation , Prostaglandin-Endoperoxide Synthases/physiology , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt
13.
Prog Lipid Res ; 45(2): 120-59, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16476485

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) compose a family of three nuclear receptors which act as lipid sensors to modulate gene expression. As such, PPARs are implicated in major metabolic and inflammatory regulations with far-reaching medical consequences, as well as in important processes controlling cellular fate. Throughout this review, we focus on the cellular functions of these receptors. The molecular mechanisms through which PPARs regulate transcription are thoroughly addressed with particular emphasis on the latest results on corepressor and coactivator action. Their implication in cellular metabolism and in the control of the balance between cell proliferation, differentiation and survival is then reviewed. Finally, we discuss how the integration of various intra-cellular signaling pathways allows PPARs to participate to whole-body homeostasis by mediating regulatory crosstalks between organs.


Subject(s)
Cell Physiological Phenomena , Peroxisome Proliferator-Activated Receptors/physiology , Animals , Gene Expression Regulation/physiology , Homeostasis/physiology , Humans , Lipid Metabolism/physiology , Signal Transduction/physiology , Transcription, Genetic/physiology
15.
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
16.
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
17.
Mol Endocrinol ; 20(8): 1715-27, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16556735

ABSTRACT

Recent evidence has emerged that peroxisome proliferator-activated receptor alpha (PPARalpha), which is largely involved in lipid metabolism, can play an important role in connecting circadian biology and metabolism. In the present study, we investigated the mechanisms by which PPARalpha influences the pacemakers acting in the central clock located in the suprachiasmatic nucleus and in the peripheral oscillator of the liver. We demonstrate that PPARalpha plays a specific role in the peripheral circadian control because it is required to maintain the circadian rhythm of the master clock gene brain and muscle Arnt-like protein 1 (bmal1) in vivo. This regulation occurs via a direct binding of PPARalpha on a potential PPARalpha response element located in the bmal1 promoter. Reversely, BMAL1 is an upstream regulator of PPARalpha gene expression. We further demonstrate that fenofibrate induces circadian rhythm of clock gene expression in cell culture and up-regulates hepatic bmal1 in vivo. Together, these results provide evidence for an additional regulatory feedback loop involving BMAL1 and PPARalpha in peripheral clocks.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Brain/metabolism , Circadian Rhythm , Liver/physiology , Muscles/metabolism , PPAR alpha/metabolism , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , CLOCK Proteins , Cells, Cultured , Down-Regulation , Eating/physiology , Feedback, Physiological , Female , Gene Expression Regulation , Male , Mice , Mice, Transgenic , Motor Activity , RNA, Messenger/metabolism , Rats , Suprachiasmatic Nucleus/metabolism , Trans-Activators/genetics
18.
Cardiovasc Res ; 65(4): 832-41, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15721863

ABSTRACT

OBJECTIVE: Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) is the predominant PPAR subtype in cardiac cells and plays a prominent role in the regulation of cardiac lipid metabolism. However, the role of PPARbeta/delta activators in cardiac hypertrophy is not yet known. METHODS AND RESULTS: In cultured neonatal rat cardiomyocytes, the selective PPARbeta/delta activator L-165041 (10 micromol/L) inhibited phenylephrine (PE)-induced protein synthesis ([(3)H]leucine uptake), induction of the fetal-type gene atrial natriuretic factor (ANF) and cardiac myocyte size. Induction of cardiac hypertrophy by PE stimulation also led to a reduction in the transcript levels of both muscle-type carnitine palmitoyltransferase (50%, P<0.05) and pyruvatedehydrogenase kinase 4 (30%, P<0.05), and these changes were reversed in the presence of the PPARbeta/delta agonist L-165041. Stimulation of neonatal rat cardiomyocytes with PE and embryonic rat heart-derived H9c2 cells with lipopolysaccharide (LPS) enhanced the expression of the nuclear factor (NF)-kappaB-target gene monocyte chemoattractant protein 1 (MCP-1). The induction of MCP-1 was reduced in the presence of L-165041, suggesting that this compound prevented NF-kappaB activation. Electrophoretic mobility shift assay (EMSA) revealed that L-165041 significantly decreased LPS-stimulated NF-kappaB binding activity in H9c2 myotubes. Finally, coimmunoprecipitation studies showed that L-165041 strongly enhanced the physical interaction between PPARbeta/delta and the p65 subunit of NF-kappaB, suggesting that increased association between these two proteins is the mechanism responsible for antagonizing NF-kappaB activation by PPARbeta/delta activators. CONCLUSION: These results suggest that PPARbeta/delta activation inhibits PE-induced cardiac hypertrophy and LPS-induced NF-kappaB activation.


Subject(s)
Cardiomegaly/pathology , Myocytes, Cardiac/pathology , PPAR delta/physiology , PPAR-beta/physiology , Acetates/pharmacology , Animals , Animals, Newborn , Cardiomegaly/metabolism , Cells, Cultured , Chemokine CCL2/metabolism , Gene Expression Regulation/drug effects , Ligands , Lipid Metabolism , Lipopolysaccharides/pharmacology , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , PPAR delta/agonists , PPAR-beta/agonists , Phenols/pharmacology , Phenoxyacetates , Phenylephrine/pharmacology , Rats , Rats, Sprague-Dawley
19.
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
20.
Endocrinology ; 146(1): 375-82, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15459119

ABSTRACT

The cellular response to fasting and starvation in tissues such as heart, skeletal muscle, and liver requires peroxisome proliferator-activated receptor-alpha (PPARalpha)-dependent up-regulation of energy metabolism toward fatty acid oxidation (FAO). PPARalpha null (PPARalphaKO) mice develop hyperinsulinemic hypoglycemia in the fasting state, and we previously showed that PPARalpha expression is increased in islets at low glucose. On this basis, we hypothesized that enhanced PPARalpha expression and FAO, via depletion of lipid-signaling molecule(s) for insulin exocytosis, are also involved in the normal adaptive response of the islet to fasting. Fasted PPARalphaKO mice compared with wild-type mice had supranormal ip glucose tolerance due to increased plasma insulin levels. Isolated islets from the PPARalpha null mice had a 44% reduction in FAO, normal glucose use and oxidation, and enhanced glucose-induced insulin secretion. In normal rats, fasting for 24 h increased islet PPARalpha, carnitine palmitoyltransferase 1, and uncoupling protein-2 mRNA expression by 60%, 62%, and 82%, respectively. The data are consistent with the view that PPARalpha, via transcriptionally up-regulating islet FAO, can reduce insulin secretion, and that this mechanism is involved in the normal physiological response of the pancreatic islet to fasting such that hypoglycemia is avoided.


Subject(s)
Adaptation, Physiological , Fasting/physiology , Fatty Acids/metabolism , Islets of Langerhans/physiology , PPAR alpha/physiology , Transcription, Genetic , Up-Regulation , Animals , Gene Expression/physiology , Glucose/metabolism , Glucose/physiology , Glucose Tolerance Test , Hormones/metabolism , Insulin/blood , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Oxidation-Reduction , PPAR alpha/deficiency , PPAR alpha/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL