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











Publication year range
1.
Front Endocrinol (Lausanne) ; 15: 1395750, 2024.
Article in English | MEDLINE | ID: mdl-38859907

ABSTRACT

Background: The beneficial effect of thermogenic adipocytes in maintaining body weight and protecting against metabolic disorders has raised interest in understanding the regulatory mechanisms defining white and beige adipocyte identity. Although alternative splicing has been shown to propagate adipose browning signals in mice, this has yet to be thoroughly investigated in human adipocytes. Methods: We performed parallel white and beige adipogenic differentiation using primary adipose stem cells from 6 unrelated healthy subjects and assessed differential gene and isoform expression in mature adipocytes by RNA sequencing. Results: We find 777 exon junctions with robust differential usage between white and beige adipocytes in all 6 subjects, mapping to 562 genes. Importantly, only 10% of these differentially spliced genes are also differentially expressed, indicating that alternative splicing constitutes an additional layer of gene expression regulation during beige adipocyte differentiation. Functional classification of alternative isoforms points to a gain of function for key thermogenic transcription factors such as PPARG and CITED1, and enzymes such as PEMT, or LPIN1. We find that a large majority of the splice variants arise from differential TSS usage, with beige-specific TSSs being enriched for PPARγ and MED1 binding compared to white-specific TSSs. Finally, we validate beige specific isoform expression at the protein level for two thermogenic regulators, PPARγ and PEMT. Discussion: These results suggest that differential isoform expression through alternative TSS usage is an important regulatory mechanism for human adipocyte thermogenic specification.


Subject(s)
Adipocytes, Beige , Alternative Splicing , Protein Isoforms , Thermogenesis , Humans , Adipocytes, Beige/metabolism , Thermogenesis/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Cell Differentiation , Adipogenesis/genetics , Male , Female , Adult , Cells, Cultured , Gene Expression Regulation , PPAR gamma/genetics , PPAR gamma/metabolism
2.
Commun Biol ; 7(1): 458, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622242

ABSTRACT

Differentiation of adipose progenitor cells into mature adipocytes entails a dramatic reorganization of the cellular architecture to accommodate lipid storage into cytoplasmic lipid droplets. Lipid droplets occupy most of the adipocyte volume, compressing the nucleus beneath the plasma membrane. How this cellular remodeling affects sub-nuclear structure, including size and number of nucleoli, remains unclear. We describe the morphological remodeling of the nucleus and the nucleolus during in vitro adipogenic differentiation of primary human adipose stem cells. We find that cell cycle arrest elicits a remodeling of nucleolar structure which correlates with a decrease in protein synthesis. Strikingly, triggering cytoskeletal rearrangements mimics the nucleolar remodeling observed during adipogenesis. Our results point to nucleolar remodeling as an active, mechano-regulated mechanism during adipogenic differentiation and demonstrate a key role of the actin cytoskeleton in defining nuclear and nucleolar architecture in differentiating human adipose stem cells.


Subject(s)
Adipogenesis , Cytoskeleton , Humans , Cells, Cultured , Cytoskeleton/metabolism , Adipocytes/metabolism , Lipid Droplets/metabolism
3.
BMC Genomics ; 23(1): 658, 2022 Sep 17.
Article in English | MEDLINE | ID: mdl-36115964

ABSTRACT

BACKGROUND: LncRNAs are tissue-specific and emerge as important regulators of various biological processes and as disease biomarkers. HOTAIR is a well-established pro-oncogenic lncRNA which has been attributed a variety of functions in cancer and native contexts. However, a lack of an exhaustive, cell type-specific annotation questions whether HOTAIR functions are supported by the expression of multiple isoforms. RESULTS: Using a capture long-read sequencing approach, we characterize HOTAIR isoforms expressed in human primary adipose stem cells. We find HOTAIR isoforms population displays varied splicing patterns, frequently leading to the exclusion or truncation of canonical LSD1 and PRC2 binding domains. We identify a highly cell type-specific HOTAIR isoform pool regulated by distinct promoter usage, and uncover a shift in the HOTAIR TSS usage that modulates the balance of HOTAIR isoforms at differentiation onset. CONCLUSION: Our results highlight the complexity and cell type-specificity of HOTAIR isoforms and open perspectives on functional implications of these variants and their balance to key cellular processes.


Subject(s)
Cell Differentiation , Protein Isoforms , RNA, Long Noncoding , Biomarkers , Cell Differentiation/genetics , Histone Demethylases , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
4.
Sci Rep ; 12(1): 10157, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35710716

ABSTRACT

The long non-coding RNA HOTAIR is the most differentially expressed gene between upper- and lower-body adipose tissue, yet its functional significance in adipogenesis is unclear. We report that HOTAIR expression is transiently induced during early adipogenic differentiation of gluteofemoral adipose progenitors and repressed in mature adipocytes. Upon adipogenic commitment, HOTAIR regulates protein synthesis pathways and cytoskeleton remodeling with a later impact on mature adipocyte lipid storage capacity. Our results support novel and important functions of HOTAIR in the physiological context of adipogenesis.


Subject(s)
Adipogenesis , RNA, Long Noncoding , Adipocytes/metabolism , Adipogenesis/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cytoskeleton/genetics , Lipids , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
5.
Genome Biol ; 21(1): 85, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32241294

ABSTRACT

At the nuclear periphery, associations of chromatin with the nuclear lamina through lamina-associated domains (LADs) aid functional organization of the genome. We review the organization of LADs and provide evidence of LAD heterogeneity from cell ensemble and single-cell data. LADs are typically repressive environments in the genome; nonetheless, we discuss findings of lamin interactions with regulatory elements of active genes, and the role lamins may play in genome regulation. We address the relationship between LADs and other genome organizers, and the involvement of LADs in laminopathies. The current data lay the basis for future studies on the significance of lamin-chromatin interactions in health and disease.


Subject(s)
Nuclear Lamina/physiology , Chromatin/metabolism , Euchromatin/metabolism , Genome , Heterochromatin/metabolism , Humans , Intermediate Filaments/metabolism , Laminopathies/genetics , Lamins/metabolism , Nuclear Envelope/metabolism , Nuclear Lamina/chemistry , Nuclear Lamina/metabolism , Single-Cell Analysis
6.
Front Cell Dev Biol ; 6: 73, 2018.
Article in English | MEDLINE | ID: mdl-30057899

ABSTRACT

At the nuclear periphery, the genome is anchored to A- and B-type nuclear lamins in the form of heterochromatic lamina-associated domains. A-type lamins also associate with chromatin in the nuclear interior, away from the peripheral nuclear lamina. This nucleoplasmic lamin A environment tends to be euchromatic, suggesting distinct roles of lamin A in the regulation of gene expression in peripheral and more central regions of the nucleus. The hot-spot lamin A R482W mutation causing familial partial lipodystrophy of Dunnigan-type (FPLD2), affects lamin A association with chromatin at the nuclear periphery and in the nuclear interior, and is associated with 3-dimensional (3D) rearrangements of chromatin. Here, we highlight features of nuclear lamin association with the genome at the nuclear periphery and in the nuclear interior. We address recent data showing a rewiring of such interactions in cells from FPLD2 patients, and in adipose progenitor and induced pluripotent stem cell models of FPLD2. We discuss associated epigenetic and genome conformation changes elicited by the lamin A R482W mutation at the gene level. The findings argue that the mutation adversely impacts both global and local genome architecture throughout the nucleus space. The results, together with emerging new computational modeling tools, mark the start of a new era in our understanding of the 3D genomics of laminopathies.

7.
Nucleus ; 9(1): 216-226, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29517398

ABSTRACT

The nuclear lamina contributes to the regulation of gene expression and to chromatin organization. Mutations in A-type nuclear lamins cause laminopathies, some of which are associated with a loss of heterochromatin at the nuclear periphery. Until recently however, little if any information has been provided on where and how lamin A interacts with the genome and on how disease-causing lamin A mutations may rearrange genome conformation. Here, we review aspects of nuclear lamin association with the genome. We highlight recent evidence of reorganization of lamin A-chromatin interactions in cellular models of laminopathies, and implications on the 3-dimensional rearrangement of chromatin in these models, including patient cells. We discuss how a hot-spot lipodystrophic lamin A mutation alters chromatin conformation and epigenetic patterns at an anti-adipogenic locus, and conclude with remarks on links between lamin A, Polycomb and the pathophysiology of laminopathies. The recent findings presented here collectively argue towards a deregulation of large-scale and local spatial genome organization by a subset of lamin A mutations causing laminopathies.


Subject(s)
Chromatin/chemistry , Chromatin/genetics , Lamin Type A/genetics , Musculoskeletal Diseases/genetics , Mutation , Nuclear Lamina/chemistry , Nuclear Lamina/genetics , Chromatin/metabolism , Humans , Lamin Type A/metabolism , Musculoskeletal Diseases/metabolism , Nuclear Lamina/metabolism
8.
Nucleus ; 9(1): 235-248, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29578370

ABSTRACT

Mutations in LMNA, encoding A-type lamins, are responsible for laminopathies including muscular dystrophies, lipodystrophies, and premature ageing syndromes. LMNA mutations have been shown to alter nuclear structure and stiffness, binding to partners at the nuclear envelope or within the nucleoplasm, gene expression and/or prelamin A maturation. LMNA-associated lipodystrophic features, combining generalized or partial fat atrophy and metabolic alterations associated with insulin resistance, could result from altered adipocyte differentiation or from altered fat structure. Recent studies shed some light on how pathogenic A-type lamin variants could trigger lipodystrophy, metabolic complications, and precocious cardiovascular events. Alterations in adipose tissue extracellular matrix and TGF-beta signaling could initiate metabolic inflexibility. Premature senescence of vascular cells could contribute to cardiovascular complications. In affected families, metabolic alterations occur at an earlier age across generations, which could result from epigenetic deregulation induced by LMNA mutations. Novel cellular models recapitulating adipogenic developmental pathways provide scalable tools for disease modeling and therapeutic screening.


Subject(s)
Lamin Type A/genetics , Lipodystrophy , Mutation , Humans , Lamin Type A/metabolism , Lipodystrophy/drug therapy , Lipodystrophy/genetics , Lipodystrophy/metabolism
9.
Hum Mol Genet ; 27(8): 1447-1459, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29438482

ABSTRACT

The p.R482W hotspot mutation in A-type nuclear lamins causes familial partial lipodystrophy of Dunnigan-type (FPLD2), a lipodystrophic syndrome complicated by early onset atherosclerosis. Molecular mechanisms underlying endothelial cell dysfunction conferred by the lamin A mutation remain elusive. However, lamin A regulates epigenetic developmental pathways and mutations could perturb these functions. Here, we demonstrate that lamin A R482W elicits endothelial differentiation defects in a developmental model of FPLD2. Genome modeling in fibroblasts from patients with FPLD2 caused by the lamin A R482W mutation reveals repositioning of the mesodermal regulator T/Brachyury locus towards the nuclear center relative to normal fibroblasts, suggesting enhanced activation propensity of the locus in a developmental model of FPLD2. Addressing this issue, we report phenotypic and transcriptional alterations in mesodermal and endothelial differentiation of induced pluripotent stem cells we generated from a patient with R482W-associated FPLD2. Correction of the LMNA mutation ameliorates R482W-associated phenotypes and gene expression. Transcriptomics links endothelial differentiation defects to decreased Polycomb-mediated repression of the T/Brachyury locus and over-activation of T target genes. Binding of the Polycomb repressor complex 2 to T/Brachyury is impaired by the mutated lamin A network, which is unable to properly associate with the locus. This leads to a deregulation of vascular gene expression over time. By connecting a lipodystrophic hotspot lamin A mutation to a disruption of early mesodermal gene expression and defective endothelial differentiation, we propose that the mutation rewires the fate of several lineages, resulting in multi-tissue pathogenic phenotypes.


Subject(s)
Endothelial Cells/metabolism , Fetal Proteins/genetics , Gene Expression Regulation, Developmental , Lamin Type A/genetics , Lipodystrophy, Familial Partial/genetics , Polycomb-Group Proteins/genetics , T-Box Domain Proteins/genetics , Adolescent , Adult , Case-Control Studies , Cell Differentiation/genetics , Cell Lineage/genetics , Endothelial Cells/pathology , Female , Fetal Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Regulatory Networks , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Lamin Type A/metabolism , Lipodystrophy, Familial Partial/metabolism , Lipodystrophy, Familial Partial/pathology , Male , Mesoderm/metabolism , Mesoderm/pathology , Middle Aged , Mutation , Polycomb-Group Proteins/metabolism , Primary Cell Culture , Protein Binding , Signal Transduction , T-Box Domain Proteins/metabolism
10.
J Cell Biol ; 216(9): 2731-2743, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28751304

ABSTRACT

Mutations in the Lamin A/C (LMNA) gene-encoding nuclear LMNA cause laminopathies, which include partial lipodystrophies associated with metabolic syndromes. The lipodystrophy-associated LMNA p.R482W mutation is known to impair adipogenic differentiation, but the mechanisms involved are unclear. We show in this study that the lamin A p.R482W hot spot mutation prevents adipogenic gene expression by epigenetically deregulating long-range enhancers of the anti-adipogenic MIR335 microRNA gene in human adipocyte progenitor cells. The R482W mutation results in a loss of function of differentiation-dependent lamin A binding to the MIR335 locus. This impairs H3K27 methylation and instead favors H3K27 acetylation on MIR335 enhancers. The lamin A mutation further promotes spatial clustering of MIR335 enhancer and promoter elements along with overexpression of the MIR355 gene after adipogenic induction. Our results link a laminopathy-causing lamin A mutation to an unsuspected deregulation of chromatin states and spatial conformation of an miRNA locus critical for adipose progenitor cell fate.


Subject(s)
Adipocytes , Adipogenesis/genetics , Epigenesis, Genetic , Fibroblasts , Lamin Type A/genetics , Lipodystrophy, Familial Partial/genetics , MicroRNAs/genetics , Mutation , Stem Cells , Acetylation , Adipocytes/metabolism , Adipocytes/pathology , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Fibroblasts/metabolism , Fibroblasts/pathology , Genetic Predisposition to Disease , Histones/metabolism , Humans , Lamin Type A/metabolism , Lipodystrophy, Familial Partial/metabolism , Lipodystrophy, Familial Partial/pathology , Lipodystrophy, Familial Partial/physiopathology , Methylation , MicroRNAs/chemistry , MicroRNAs/metabolism , Nucleic Acid Conformation , Phenotype , Promoter Regions, Genetic , Stem Cells/metabolism , Stem Cells/pathology , Structure-Activity Relationship , Up-Regulation
11.
Diabetes ; 66(6): 1470-1478, 2017 06.
Article in English | MEDLINE | ID: mdl-28270520

ABSTRACT

Activation of thermogenic beige adipocytes has recently emerged as a promising therapeutic target in obesity and diabetes. Relevant human models for beige adipocyte differentiation are essential to implement such therapeutic strategies. We report a straightforward and efficient protocol to generate functional human beige adipocytes from human induced pluripotent stem cells (hiPSCs). Without overexpression of exogenous adipogenic genes, our method recapitulates an adipogenic developmental pathway through successive mesodermal and adipogenic progenitor stages. hiPSC-derived adipocytes are insulin sensitive and display beige-specific markers and functional properties, including upregulation of thermogenic genes, increased mitochondrial content, and increased oxygen consumption upon activation with cAMP analogs. Engraftment of hiPSC-derived adipocytes in mice produces well-organized and vascularized adipose tissue, capable of ß-adrenergic-responsive glucose uptake. Our model of human beige adipocyte development provides a new and scalable tool for disease modeling and therapeutic screening.


Subject(s)
Adipocytes, Beige/metabolism , Adipose Tissue/metabolism , Cellular Reprogramming Techniques/methods , Glucose/metabolism , Induced Pluripotent Stem Cells/cytology , Insulin Resistance , Obesity , Thermogenesis/genetics , Adipocytes, Beige/cytology , Adipocytes, Beige/drug effects , Adipocytes, Beige/transplantation , Adipogenesis , Adipose Tissue/drug effects , Adrenergic beta-Agonists/pharmacology , Animals , Cell Transplantation , Fluorodeoxyglucose F18 , Gene Expression Profiling , Humans , Isoproterenol/pharmacology , Mice , Mitochondria/metabolism , Oxygen Consumption , RNA, Messenger/metabolism , Radiopharmaceuticals , Real-Time Polymerase Chain Reaction , Up-Regulation
12.
Genome Biol ; 18(1): 21, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28137286

ABSTRACT

Current three-dimensional (3D) genome modeling platforms are limited by their inability to account for radial placement of loci in the nucleus. We present Chrom3D, a user-friendly whole-genome 3D computational modeling framework that simulates positions of topologically-associated domains (TADs) relative to each other and to the nuclear periphery. Chrom3D integrates chromosome conformation capture (Hi-C) and lamin-associated domain (LAD) datasets to generate structure ensembles that recapitulate radial distributions of TADs detected in single cells. Chrom3D reveals unexpected spatial features of LAD regulation in cells from patients with a laminopathy-causing lamin mutation. Chrom3D is freely available on github.


Subject(s)
Chromatin/genetics , Computational Biology/methods , Nuclear Lamina/genetics , Adult , Female , Genome , HeLa Cells , Humans , Male , Models, Genetic , Young Adult
13.
Virologie (Montrouge) ; 20(1): 47-60, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-33065852

ABSTRACT

Hemagglutinin (HA) and neuraminidase (NA) are major glycoproteins expressed on the surface of influenza virus. They have complementary and antagonistic functions that facilitate in the life cycle of the virus. The functional equilibrium generated between HA and NA can impact the evolution and adaptation of influenza virus strains within the human reservoir. This functional equilibrium is referred to as the "HA-NA balance". An imbalanced HA-NA can restrict the multiplication and transmission capacity of influenza viruses. Moreover, this equilibrium is likely a limiting factor against species crossover for the virus. In light of such considerations, the HA-NA balance should be precisely studied to gain a better understanding of the emergence of pandemic and seasonal influenza virus strains. This review describes the concept of the HA-NA balance, the methods used to study it, plus a discussion of the HA-NA balance in the evolution of the pandemic influenza A H1N1 strains that plagued the world in 1918 and 2009.

14.
Adipocyte ; 4(2): 158-60, 2015.
Article in English | MEDLINE | ID: mdl-26167412

ABSTRACT

Recent data indicate that cell size fluctuation, a key property in adipocyte pathophysiology primarily dependent on lipid storage, is linked to a novel function of lipid droplet organelles acting as mechano-active organelles to regulate cell membrane remodeling and caveolae dynamics.

15.
Hum Mol Genet ; 24(7): 2096-109, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25524705

ABSTRACT

Nuclear lamins are involved in many cellular functions due to their ability to bind numerous partners including chromatin and transcription factors, and affect their properties. Dunnigan type familial partial lipodystrophy (FPLD2; OMIM#151660) is caused in most cases by the A-type lamin R482W mutation. We report here that the R482W mutation affects the regulatory activity of sterol response element binding protein 1 (SREBP1), a transcription factor that regulates hundreds of genes involved in lipid metabolism and adipocyte differentiation. Using in situ proximity ligation assays (PLA), reporter assays and biochemical and transcriptomic approaches, we show that interactions of SREBP1 with lamin A and lamin C occur at the nuclear periphery and in the nucleoplasm. These interactions involve the Ig-fold of A-type lamins and are favored upon SREBP1 binding to its DNA target sequences. We show that SREBP1, LMNA and sterol response DNA elements form ternary complexes in vitro. In addition, overexpression of A-type lamins reduces transcriptional activity of SREBP1. In contrast, both overexpression of LMNA R482W in primary human preadipocytes and endogenous expression of A-type lamins R482W in FPLD2 patient fibroblasts, reduce A-type lamins-SREBP1 in situ interactions and upregulate a large number of SREBP1 target genes. As this LMNA mutant was previously shown to inhibit adipogenic differentiation, we propose that deregulation of SREBP1 by mutated A-type lamins constitutes one underlying mechanism of the physiopathology of FPLD2. Our data suggest that SREBP1 targeting molecules could be considered in a therapeutic context.


Subject(s)
Amino Acid Substitution , Lamin Type A/genetics , Lipodystrophy, Familial Partial/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Adult , Female , Humans , Lamin Type A/metabolism , Lipodystrophy, Familial Partial/genetics , Male , Middle Aged , Mutation, Missense , Protein Binding , Sterol Regulatory Element Binding Protein 1/genetics , Young Adult
16.
Diabetes ; 63(12): 4032-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24969108

ABSTRACT

Adipocytes specialized in the storage of energy as fat are among the most caveolae-enriched cell types. Loss of caveolae produces lipodystrophic diabetes in humans, which cannot be reversed by endothelial rescue of caveolin expression in mice, indicating major importance of adipocyte caveolae. However, how caveolae participate in fat cell functions is poorly understood. We investigated dynamic conditions of lipid store fluctuations and demonstrate reciprocal regulation of caveolae density and fat cell lipid droplet storage. We identified caveolin-1 expression as a crucial step in adipose cell lines and in mice to raise the density of caveolae, to increase adipocyte ability to accommodate larger lipid droplets, and to promote cell expansion by increased glucose utilization. In human subjects enrolled in a trial of 8 weeks of overfeeding to promote fattening, adipocyte expansion response correlated with initial caveolin-1 expression. Conversely, lipid mobilization in cultured adipocytes to induce lipid droplet shrinkage led to biphasic response of cavin-1 with ultimate loss of expression of cavin-1 and -3 and EHD2 by protein degradation, coincident with caveolae disassembly. We have identified the key steps in cavin/caveolin interplay regulating adipocyte caveolae dynamics. Our data establish that caveolae participate in a unique cell response connected to lipid store fluctuation, suggesting lipid-induced mechanotension in adipocytes.


Subject(s)
Adipocytes/metabolism , Caveolae/metabolism , Caveolin 1/genetics , Lipid Metabolism , Membrane Proteins/genetics , RNA, Messenger/analysis , RNA-Binding Proteins/genetics , 3T3-L1 Cells , Adult , Animals , Caveolin 1/metabolism , Female , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Nude , RNA-Binding Proteins/metabolism , Young Adult
17.
Curr Diab Rep ; 13(6): 757-67, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24026869

ABSTRACT

Genetic lipodystrophic syndromes are rare diseases characterized by generalized or partial fat atrophy (lipoatrophy) associated with severe metabolic complications such as insulin resistance (IR), diabetes, dyslipidemia, nonalcoholic fatty liver disease, and ovarian hyperandrogenism. During the last 15 years, mutations in several genes have been shown to be responsible for monogenic forms of lipodystrophic syndromes, of autosomal dominant or recessive transmission. Although the molecular basis of lipodystrophies is heterogeneous, most mutated genes lead to impaired adipogenesis, adipocyte lipid storage, and/or formation or maintenance of the adipocyte lipid droplet (LD), showing that primary alterations of adipose tissue (AT) can result in severe systemic metabolic and endocrine consequences. The reduced expandability of AT alters its ability to buffer excess caloric intake, leading to ectopic lipid storage that impairs insulin signaling and other cellular functions ("lipotoxicity"). Genetic studies have also pointed out the close relationships between ageing, inflammatory processes, lipodystrophy, and IR.


Subject(s)
Insulin Resistance/physiology , Lipodystrophy/genetics , Aging/physiology , Animals , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Humans , Inflammation/genetics , Inflammation/physiopathology , Insulin Resistance/genetics , Lipodystrophy/physiopathology
18.
Diabetes ; 60(2): 448-53, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21270257

ABSTRACT

OBJECTIVE: Defective caveolin-1 expression is now recognized as a cause of lipoatrophic diabetes in patients, due to primary caveolin gene mutations or secondary caveolin deficiency caused by PTRF/cavin gene defects. The goal of this study was to establish the relative contribution of endothelial cells and adipocytes, both highly expressing caveolin-1 to the lipoatrophic phenotype of mice with global caveolin-1 gene invalidation (Cav1-KO). RESEARCH DESIGN AND METHODS: We compared adipose tissue development and metabolic phenotype of wild-type (WT), lipoatrophic Cav1-KO, and a murine model with specific rescue of caveolin-1 expression in endothelial cells (caveolin-1-reconstituted [Cav1-RC]). RESULTS: Defective adipose tissue development, reduced adipocyte size, and global alteration in adipose tissue gene expression that characterize lipoatrophic caveolin-1 null mice were still observed in Cav1-RC, indicating a prominent role of adipocyte-derived caveolin in lipoatrophy. We also observed that Cav1-KO adipose tissue contained an increased proportion of infiltrated macrophages compared with control mice, mostly with an alternate activation M2 phenotype. In contrast with defective lipid storage and lipoatrophy, macrophage infiltration was normalized in Cav1-RC mice, pointing to caveolin-1-dependent endothelium permeability as the causing factor for adipose tissue macrophage infiltration in this model. CONCLUSIONS: This is the first report of a specific role for adipocyte caveolin expression in lipid storage. Our study also shows that endothelium caveolin critically participates in the control of macrophage extravasation from the blood into adipose tissue, therefore establishing distinct roles depending on topology of caveolin expression in different cell types of adipose tissue.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Caveolin 1/metabolism , Endothelial Cells/metabolism , Macrophages/metabolism , Analysis of Variance , Animals , Blotting, Western , Caveolin 1/genetics , Immunohistochemistry , Mice , Mice, Knockout , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
19.
Biochimie ; 93(1): 71-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20363285

ABSTRACT

Caveolae are specialized lipid microdomains, forming small invaginations in the plasma membrane, known to be implicated in multiple functions including lipid storage, cell signaling and endocytosis. Formation of these wide flask-shaped invaginations is dependent on the expression of a caveolar coat protein, namely caveolin. Until now, the accepted paradigm was that caveolin was the sole and only structural protein of caveolae since its expression was necessary and sufficient to drive caveolae biogenesis. The recent characterizations of PTRF/cavin-1 and subsequently other cavin family members in caveolae formation have highlighted additional levels of complexity in the biogenesis of these plasma membrane invaginations. In this review, recent advances on the role of the different cavin family members in the regulation of caveolae structures as well as potential new functions will be discussed.


Subject(s)
Caveolae , Caveolins/metabolism , Membrane Proteins , Animals , Caveolae/metabolism , Caveolae/ultrastructure , Cell Membrane/metabolism , Endocytosis , Humans , Lipodystrophy/metabolism , Membrane Proteins/metabolism , Mice , RNA-Binding Proteins , Signal Transduction/physiology
20.
Autophagy ; 6(6): 754-63, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20574167

ABSTRACT

Adipose tissue lipoatrophy caused by caveolin gene deletion in mice is not linked to defective adipocyte differentiation. We show that adipose tissue development cannot be rescued by endothelial specific caveolin-1 re-expression, indicating primordial role of caveolin in mature adipocytes. Partial or total caveolin deficiency in adipocytes induced broad protein expression defects, including but not limited to previously described downregulation of insulin receptor. Global alterations in protein turnover, and accelerated degradation of long-lived proteins were found in caveolin-deficient adipocytes. Lipidation of endogenous LC3 autophagy marker and distribution of GFP-LC3 into aggregates demonstrated activated autophagy in the absence of caveolin-1 in adipocytes. Furthermore, electron microscopy revealed autophagic vacuoles in caveolin-1 deficient but not control adipocytes. Surprisingly, significant levels of lipidated LC3-II were found around lipid droplets of normal adipocytes, maintained in nutrient-rich conditions or isolated from fed mice, which do not display autophagy. Altogether, these data indicate that caveolin deficiency induce autophagy in adipocytes, a feature that is not a physiological response to fasting in normal fat cells. This likely resulted from defective insulin and lipolytic responses that converge in chronic nutrient shortage in adipocytes lacking caveolin-1. This is the first report of a pathological situation with autophagy as an adaptative response to adipocyte failure.


Subject(s)
Adipocytes/cytology , Autophagy , Caveolin 1/deficiency , Cell Differentiation , Lipid Metabolism , Adipocytes/ultrastructure , Animals , Caveolin 1/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Gene Silencing , Green Fluorescent Proteins/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Models, Animal , Models, Biological , Phagosomes/metabolism , Phagosomes/ultrastructure , Protein Processing, Post-Translational , Protein Transport , Recombinant Fusion Proteins/metabolism , Stromal Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL