Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Circulation ; 147(6): 498-511, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36484260

ABSTRACT

BACKGROUND: Myocardial infarction (MI) induces a repair response that ultimately generates a stable fibrotic scar. Although the scar prevents cardiac rupture, an excessive profibrotic response impairs optimal recovery by promoting the development of noncontractile fibrotic areas. The mechanisms that lead to cardiac fibrosis are diverse and incompletely characterized. We explored whether the expansion of cardiac fibroblasts after MI can be regulated through a paracrine action of cardiac stromal cells. METHODS: We performed a bioinformatic secretome analysis of cardiac stromal PW1+ cells isolated from normal and post-MI mouse hearts to identify novel secreted proteins. Functional assays were used to screen secreted proteins that promote fibroblast proliferation. The expressions of candidates were subsequently analyzed in mouse and human hearts and plasmas. The relationship between levels of circulating protein candidates and adverse post-MI cardiac remodeling was examined in a cohort of 80 patients with a first ST-segment-elevation MI and serial cardiac magnetic resonance imaging evaluations. RESULTS: Cardiac stromal PW1+ cells undergo a change in paracrine behavior after MI, and the conditioned media from these cells induced a significant increase in the proliferation of fibroblasts. We identified a total of 12 candidates as secreted proteins overexpressed by cardiac PW1+ cells after MI. Among these factors, GDF3 (growth differentiation factor 3), a member of the TGF-ß (transforming growth factor-ß) family, was markedly upregulated in the ischemic hearts. Conditioned media specifically enriched with GDF3 induced fibroblast proliferation at a high level by stimulation of activin-receptor-like kinases. In line with the secretory nature of this protein, we next found that GDF3 can be detected in mice and human plasma samples, with a significant increase in the days after MI. In humans, higher GDF3 circulating levels (measured in the plasma at day 4 after MI) were significantly associated with an increased risk of adverse remodeling 6 months after MI (adjusted odds ratio, 1.76 [1.03-3.00]; P=0.037), including lower left ventricular ejection fraction and a higher proportion of akinetic segments. CONCLUSIONS: Our findings define a mechanism for the profibrotic action of cardiac stromal cells through secreted cardiokines, such as GDF3, a candidate marker of adverse fibrotic remodeling after MI. REGISTRATION: URL: https://www. CLINICALTRIALS: gov; Unique identifier: NCT01113268.


Subject(s)
Myocardial Infarction , Myocardium , Animals , Humans , Mice , Cicatrix/pathology , Culture Media, Conditioned/pharmacology , Culture Media, Conditioned/metabolism , Disease Models, Animal , Fibrosis , Growth Differentiation Factor 3/metabolism , Myocardium/metabolism , Stroke Volume , Transforming Growth Factor beta/metabolism , Ventricular Function, Left , Ventricular Remodeling
2.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33830083

ABSTRACT

Macrophage-mediated inflammatory response has been implicated in the pathogenesis of obesity and insulin resistance. Brd4 has emerged as a key regulator in the innate immune response. However, the role of Brd4 in obesity-associated inflammation and insulin resistance remains uncharacterized. Here, we demonstrated that myeloid lineage-specific Brd4 knockout (Brd4-CKO) mice were protected from high-fat diet-induced (HFD-induced) obesity with less fat accumulation, higher energy expenditure, and increased lipolysis in adipose tissue. Brd4-CKO mice fed a HFD also displayed reduced local and systemic inflammation with improved insulin sensitivity. RNA-Seq of adipose tissue macrophages (ATMs) from HFD-fed WT and Brd4-CKO mice revealed that expression of antilipolytic factor Gdf3 was significantly decreased in ATMs of Brd4-CKO mice. We also found that Brd4 bound to the promoter and enhancers of Gdf3 to facilitate PPARγ-dependent Gdf3 expression in macrophages. Furthermore, Brd4-mediated expression of Gdf3 acted as a paracrine signal targeting adipocytes to suppress the expression of lipases and the associated lipolysis in cultured cells and mice. Controlling the expression of Gdf3 in ATMs could be one of the mechanisms by which Brd4 modulates lipid metabolism and diet-induced obesity. This study suggests that Brd4 could be a potential therapeutic target for obesity and insulin resistance.


Subject(s)
Adipose Tissue/cytology , Growth Differentiation Factor 3/genetics , Macrophages/metabolism , Nuclear Proteins/metabolism , Obesity/etiology , Transcription Factors/metabolism , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Gene Expression Regulation , Growth Differentiation Factor 3/metabolism , Insulin Resistance/genetics , Lipase/genetics , Lipase/metabolism , Lipid Metabolism/physiology , Lipolysis/genetics , Male , Mice, Knockout , Nuclear Proteins/genetics , PPAR gamma/metabolism , Promoter Regions, Genetic , Transcription Factors/genetics
3.
Cell Metab ; 32(4): 665-675.e6, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32941798

ABSTRACT

The thiazolidinediones (TZDs) are ligands of PPARγ that improve insulin sensitivity, but their use is limited by significant side effects. Recently, we demonstrated a mechanism wherein TZDs improve insulin sensitivity distinct from receptor agonism and adipogenesis: reversal of obesity-linked phosphorylation of PPARγ at serine 273. However, the role of this modification hasn't been tested genetically. Here we demonstrate that mice encoding an allele of PPARγ that cannot be phosphorylated at S273 are protected from insulin resistance, without exhibiting differences in body weight or TZD-associated side effects. Indeed, hyperinsulinemic-euglycemic clamp experiments confirm insulin sensitivity. RNA-seq in these mice reveals reduced expression of Gdf3, a BMP family member. Ectopic expression of Gdf3 is sufficient to induce insulin resistance in lean, healthy mice. We find Gdf3 inhibits BMP signaling and insulin signaling in vitro. Together, these results highlight the diabetogenic role of PPARγ S273 phosphorylation and focus attention on a putative target, Gdf3.


Subject(s)
Growth Differentiation Factor 3/metabolism , Obesity/drug therapy , PPAR gamma/metabolism , Thiazolidinediones/pharmacology , Alleles , Animals , Cells, Cultured , Growth Differentiation Factor 3/genetics , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , PPAR gamma/genetics , Phosphorylation/drug effects
4.
Cells ; 9(1)2020 01 03.
Article in English | MEDLINE | ID: mdl-31947892

ABSTRACT

Macrophages are critical for regulation of inflammatory response during endotoxemia and septic shock. However, the mediators underlying their regulatory function remain obscure. Growth differentiation factor 3 (GDF3), a member of transforming growth factor beta (TGF-ß) superfamily, has been implicated in inflammatory response. Nonetheless, the role of GDF3 in macrophage-regulated endotoxemia/sepsis is unknown. Here, we show that serum GDF3 levels in septic patients are elevated and strongly correlate with severity of sepsis and 28-day mortality. Interestingly, macrophages treated with recombinant GDF3 protein (rGDF3) exhibit greatly reduced production of pro-inflammatory cytokines, comparing to controls upon endotoxin challenge. Moreover, acute administration of rGDF3 to endotoxin-treated mice suppresses macrophage infiltration to the heart, attenuates systemic and cardiac inflammation with less pro-inflammatory macrophages (M1) and more anti-inflammatory macrophages (M2), as well as prolongs mouse survival. Mechanistically, GDF3 is able to activate Smad2/Smad3 phosphorylation, and consequently inhibits the expression of nod-like receptor protein-3 (NLRP3) in macrophages. Accordingly, blockade of Smad2/Smad3 phosphorylation with SB431542 significantly offsets rGDF3-mediated anti-inflammatory effects. Taken together, this study uncovers that GDF3, as a novel sepsis-associated factor, may have a dual role in the pathophysiology of sepsis. Acute administration of rGDF3 into endotoxic shock mice could increase survival outcome and improve cardiac function through anti-inflammatory response by suppression of M1 macrophage phenotype. However, constitutive high levels of GDF3 in human sepsis patients are associated with lethality, suggesting that GDF3 may promote macrophage polarization toward M2 phenotype which could lead to immunosuppression.


Subject(s)
Growth Differentiation Factor 3/metabolism , Heart/physiopathology , Inflammation/pathology , Macrophages/pathology , Sepsis/prevention & control , Sepsis/physiopathology , Adult , Animals , Case-Control Studies , Cell Polarity/drug effects , Cytokines/biosynthesis , Endotoxins , Growth Differentiation Factor 3/blood , Growth Differentiation Factor 3/genetics , Humans , Inflammation/blood , Mice, Inbred C57BL , Models, Biological , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Sepsis/blood , Smad Proteins/metabolism , Spleen/pathology , Survival Analysis , Treatment Outcome
5.
Mov Disord ; 33(7): 1108-1118, 2018 07.
Article in English | MEDLINE | ID: mdl-30153385

ABSTRACT

BACKGROUND: The most likely genetic cause of X-linked dystonia-parkinsonism, a neurodegenerative movement disorder endemic to the Philippines, is a 2672-bp-long retrotransposon insertion in intron 32 of the TAF1 gene. The objectives of this study were to investigate whether (1) TAF1 expression is altered in induced pluripotent stem cells and differentiated neuronal models and (2) excision of the retrotransposon insertion restores normal TAF1 expression. METHODS: Expression of TAF1 and its neuronal isoform were determined in induced pluripotent stem cells and in induced pluripotent stem cell-derived cortical neurons and spiny projection neurons using quantitative PCR. Genome editing-based excision of the retrotransposon insertion was performed on induced pluripotent stem cells from 3 X-linked dystonia-parkinsonism patients. Edited and unedited induced pluripotent stem cells from X-linked dystonia-parkinsonism patients and controls were differentiated into cortical neurons and spiny projection neurons, and TAF1 expression was compared across groups. RESULTS: TAF1 was reduced in patient-derived induced pluripotent stem cells (P < 0.05) and spiny projection neurons (P < 0.01). After genome editing, we observed higher TAF1 expression in edited compared with unedited induced pluripotent stem cells (P < 0.0001). In edited spiny projection neurons, TAF1 expression was also increased, but did not reach statistical significance. No expression differences were observed in cortical neurons. CONCLUSIONS: (1) TAF1 reduction in X-linked dystonia-parkinsonism is likely due to the retrotransposon insertion and is recapitulated in induced pluripotent stem cells and differentiated spiny projection neurons. (2) TAF1 reduction is a tractable molecular phenotype of X-linked dystonia-parkinsonism that can be driven by excision of the retrotransposon insertion. (3) Successful rescue of the molecular phenotype in an endogenous, genome-edited model serves as a proof of principle that may successfully be transferred to other inherited neurodegenerative diseases. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Dystonic Disorders/genetics , Dystonic Disorders/metabolism , Gene Editing/methods , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Histone Acetyltransferases/metabolism , Induced Pluripotent Stem Cells/physiology , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Adult , Cells, Cultured , Cerebral Cortex/cytology , Female , Growth Differentiation Factor 3/metabolism , Humans , Male , Middle Aged , Nanog Homeobox Protein/metabolism , Nerve Tissue Proteins/metabolism , Octamer Transcription Factor-3/metabolism , RNA, Messenger/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transfection , Tubulin/metabolism
6.
Diabetes ; 67(9): 1761-1772, 2018 09.
Article in English | MEDLINE | ID: mdl-29945891

ABSTRACT

Previous genetic studies in mice have shown that functional loss of activin receptor-like kinase 7 (ALK7), a type I transforming growth factor-ß receptor, increases lipolysis to resist fat accumulation in adipocytes. Although growth/differentiation factor 3 (GDF3) has been suggested to function as a ligand of ALK7 under nutrient-excess conditions, it is unknown how GDF3 production is regulated. Here, we show that a physiologically low level of insulin converts CD11c- adipose tissue macrophages (ATMs) into GDF3-producing CD11c+ macrophages ex vivo and directs ALK7-dependent accumulation of fat in vivo. Depletion of ATMs by clodronate upregulates adipose lipases and reduces fat mass in ALK7-intact obese mice, but not in their ALK7-deficient counterparts. Furthermore, depletion of ATMs or transplantation of GDF3-deficient bone marrow negates the in vivo effects of insulin on both lipolysis and fat accumulation in ALK7-intact mice. The GDF3-ALK7 axis between ATMs and adipocytes represents a previously unrecognized mechanism by which insulin regulates both fat metabolism and mass.


Subject(s)
Activin Receptors, Type I/metabolism , Adipose Tissue, White/drug effects , Growth Differentiation Factor 3/agonists , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Lipolysis/drug effects , Macrophages/drug effects , Activin Receptors, Type I/genetics , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity/drug effects , Animals , Bone Marrow Transplantation , CD11c Antigen/metabolism , Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , HEK293 Cells , Humans , Hypoglycemic Agents/therapeutic use , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Insulin/therapeutic use , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Congenic , Mice, Inbred Strains , Mice, Knockout , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Obesity/therapy , Weight Gain/drug effects
7.
Vitam Horm ; 107: 227-261, 2018.
Article in English | MEDLINE | ID: mdl-29544632

ABSTRACT

Primordial germ cells migrate to the fetal gonads and proliferate during gestation to generate a fixed complement of primordial follicles, the so-called ovarian reserve. Primordial follicles comprise an oocyte arrested at the diplotene stage of meiosis, surrounded by a layer of pregranulosa cells. Activation of primordial follicles to grow beyond this arrested stage is of particular interest because, once activated, they are subjected to regulatory mechanisms involved in growth, selection, maturation, and ultimately, ovulation or atresia. The vast majority of follicles succumb to atresia and are permanently lost from the quiescent or growing pool of follicles. The bone morphogenetic proteins (BMPs), together with other intraovarian growth factors, are intimately involved in regulation of follicle recruitment, dominant follicle selection, ovulation, and atresia. Activation of primordial follicles appears to be a continuous process, and the number of small antral follicles at the beginning of the menstrual cycle provides an indirect indication of ovarian reserve. Continued antral follicle development during the follicular phase of the menstrual cycle is driven by follicle stimulating hormone (FSH) and luteinizing hormone (LH) in conjunction with many intraovarian growth factors and inhibitors interrelated in a complex web of regulatory balance. The BMP signaling system has a major intraovarian role in many species, including the human, in the generation of transcription factors that influence proliferation, steroidogenesis, cell differentiation, and maturation prior to ovulation, as well as formation of corpora lutea after ovulation. At the anterior pituitary level, BMPs also contribute to the regulation of gonadotrophin production.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Follicular Phase/metabolism , Gene Expression Regulation, Developmental , Models, Biological , Oogenesis , Ovary/physiology , Animals , Anti-Mullerian Hormone/genetics , Anti-Mullerian Hormone/metabolism , Bone Morphogenetic Protein Receptors/agonists , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/genetics , Female , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , Growth Differentiation Factor 9/genetics , Growth Differentiation Factor 9/metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiology , Ligands , Ovary/cytology , Ovary/metabolism , Ovulation/metabolism , Signal Transduction
8.
Stem Cell Res Ther ; 8(1): 200, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28962587

ABSTRACT

BACKGROUND: Although tissue-resident mesenchymal stromal cells (MSCs) in the larynx have been described, their distinct characteristics and roles have not been thoroughly explored. Therefore, we investigated stem cell characteristics and regenerative potentials of single clonal populations isolated from rat epiglottic mucosa (EM), lamina propria (LP), and macula flava (MF) to determine whether they comprised laryngeal tissue-resident stem cells. METHODS: Single clonal laryngeal cells were isolated following microdissection of the EM, LP, and MF from the rat larynx. Several clonal populations from the three laryngeal subsites were selected and expanded in vitro. We compared the stem cell characteristics of self-renewal and differentiation potential, as well as the cell surface phenotypes and gene expression profiles, of laryngeal MSC-like cells to that of bone marrow MSCs (BM-MSCs). We also investigated the regenerative potential of the laryngeal cells in a radiation-induced laryngeal injury animal model. RESULTS: Self-renewing, clonal cell populations were obtained from rat EM, LP, and MF. EM-derived and LP-derived clonal cells had fibroblast-like features, while MF-resident clonal cells had stellate cell morphology and lipid droplets containing vitamin A. All laryngeal clonal cell populations had MSC-like cell surface marker expression (CD29, CD44, CD73, and CD90) and the potential to differentiate into bone and cartilage cell lineages; EM-derived and MF-derived cells, but not LP-derived cells, were also able to differentiate into adipocytes. Clonal cells isolated from the laryngeal subsites exhibited differential extracellular matrix-related gene expression. We found that the mesenchymal and stellate cell-related genes desmin and nestin were enriched in laryngeal MSC-like cells relative to BM-MSCs (P < 0.001). Growth differentiation factor 3 (GDF3) and glial fibrillary acidic protein (GFAP) transcript and protein levels were higher in MF-derived cells than in other laryngeal populations (P < 0.001). At 4 weeks after transplantation, laryngeal MF-derived and EM-derived cells contributed to laryngeal epithelial and/or glandular regeneration in response to radiation injury. CONCLUSIONS: These results suggest that cell populations with MSC characteristics reside in the EM, LP, and MF of the larynx. Laryngeal MSC-like cells contribute to regeneration of the larynx following injury; further investigation is needed to clarify the differential roles of the populations in laryngeal tissue regeneration, as well as the clinical implications for the treatment of laryngeal disease.


Subject(s)
Cell Differentiation , Larynx/cytology , Mesenchymal Stem Cells/cytology , Animals , Cell Lineage , Cell Proliferation , Desmin/genetics , Desmin/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , Larynx/injuries , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/classification , Mesenchymal Stem Cells/metabolism , Nestin/genetics , Nestin/metabolism , Radiation Injuries, Experimental/therapy , Rats , Rats, Sprague-Dawley
9.
Nature ; 550(7674): 119-123, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28953873

ABSTRACT

Catecholamine-induced lipolysis, the first step in the generation of energy substrates by the hydrolysis of triglycerides, declines with age. The defect in the mobilization of free fatty acids in the elderly is accompanied by increased visceral adiposity, lower exercise capacity, failure to maintain core body temperature during cold stress, and reduced ability to survive starvation. Although catecholamine signalling in adipocytes is normal in the elderly, how lipolysis is impaired in ageing remains unknown. Here we show that adipose tissue macrophages regulate the age-related reduction in adipocyte lipolysis in mice by lowering the bioavailability of noradrenaline. Unexpectedly, unbiased whole-transcriptome analyses of adipose macrophages revealed that ageing upregulates genes that control catecholamine degradation in an NLRP3 inflammasome-dependent manner. Deletion of NLRP3 in ageing restored catecholamine-induced lipolysis by downregulating growth differentiation factor-3 (GDF3) and monoamine oxidase A (MAOA) that is known to degrade noradrenaline. Consistent with this, deletion of GDF3 in inflammasome-activated macrophages improved lipolysis by decreasing levels of MAOA and caspase-1. Furthermore, inhibition of MAOA reversed the age-related reduction in noradrenaline concentration in adipose tissue, and restored lipolysis with increased levels of the key lipolytic enzymes adipose triglyceride lipase (ATGL) and hormone sensitive lipase (HSL). Our study reveals that targeting neuro-immunometabolic signalling between the sympathetic nervous system and macrophages may offer new approaches to mitigate chronic inflammation-induced metabolic impairment and functional decline.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Aging/metabolism , Catecholamines/metabolism , Inflammasomes/metabolism , Lipolysis , Macrophages/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Aging/drug effects , Aging/genetics , Animals , Caspase 1/metabolism , Catecholamines/pharmacology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Growth Differentiation Factor 3/deficiency , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , Lipase/metabolism , Lipolysis/drug effects , Lipolysis/genetics , Mice , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Norepinephrine/metabolism , Sterol Esterase/metabolism
11.
FEBS Lett ; 591(11): 1584-1600, 2017 06.
Article in English | MEDLINE | ID: mdl-28471520

ABSTRACT

The process of cell reprogramming has been characterized considerably since the successful generation of induced pluripotent stem cells. However, the importance of cell-cell communications for cellular reprogramming remains largely unknown. Secreted factors, which are expressed and secreted during reprogramming, may influence the reprogramming efficiency. Here, we have identified Sostdc1, Glb1l2, Fetub, Dpp4, Gdf3, Trh, and Tdgf1 as prominently upregulated secreted factors during reprogramming. Our detailed analysis reveals that these seven factors may be categorized into four groups based on their expression patterns in relation to the reprogramming stages. Remarkably, knockdown of Sostdc1, which is the most prominently upregulated factor and which is expressed earlier than the other six factors, results in reduced reprogramming efficiency, suggesting its involvement in the reprogramming process.


Subject(s)
Cellular Reprogramming/genetics , Gene Expression Regulation , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cells, Cultured , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Fetuin-B/genetics , Fetuin-B/metabolism , Fibroblasts/metabolism , Flow Cytometry , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , Immunoblotting , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Microarray Analysis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
Immunity ; 45(5): 1038-1051, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27836432

ABSTRACT

Tissue regeneration requires inflammatory and reparatory activity of macrophages. Macrophages detect and eliminate the damaged tissue and subsequently promote regeneration. This dichotomy requires the switch of effector functions of macrophages coordinated with other cell types inside the injured tissue. The gene regulatory events supporting the sensory and effector functions of macrophages involved in tissue repair are not well understood. Here we show that the lipid activated transcription factor, PPARγ, is required for proper skeletal muscle regeneration, acting in repair macrophages. PPARγ controls the expression of the transforming growth factor-ß (TGF-ß) family member, GDF3, which in turn regulates the restoration of skeletal muscle integrity by promoting muscle progenitor cell fusion. This work establishes PPARγ as a required metabolic sensor and transcriptional regulator of repair macrophages. Moreover, this work also establishes GDF3 as a secreted extrinsic effector protein acting on myoblasts and serving as an exclusively macrophage-derived regeneration factor in tissue repair.


Subject(s)
Growth Differentiation Factor 3/metabolism , Muscle, Skeletal/physiology , Myoblasts/metabolism , PPAR gamma/metabolism , Regeneration/physiology , Animals , Blotting, Western , Cell Separation , Chromatin Immunoprecipitation , Disease Models, Animal , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/injuries , Oligonucleotide Array Sequence Analysis , Wound Healing/physiology
13.
PLoS One ; 8(8): e70612, 2013.
Article in English | MEDLINE | ID: mdl-23950971

ABSTRACT

Misexpression of growth factors, particularly those related to stem cell-like phenotype, is often observed in several cancer types. It has been found to influence parameters of disease progression like cell proliferation, differentiation, maintenance of undifferentiated phenotype and modulation of the immune system. GDF3 is a TGFB family member associated with pluripotency and differentiation during embryonic development that has been previously reported to be re-expressed in a number of cancer types. However, its role in tumor development and progression has not been clarified yet. In this study we decipher the role of GDF3 in an in vitro model of cancer stem cells, NCCIT cells. By classical approach to study protein function combined with high-throughput technique for transcriptome analysis and differentiation assays we evaluated GDF3 as a potential therapeutic target. We observed that GDF3 robustly induces a panel of genes related to differentiation, including several potent tumor suppressors, without impacting the proliferative capacity. Moreover, we report for the first time the protective effect of GDF3 against retinoic acid-induced apoptosis in cells with stem cell-like properties. Our study implies that blocking of GDF3 combined with retinoic acid-treatment of solid cancers is a compelling direction for further investigations, which can lead to re-design of cancer differentiation therapies.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/genetics , Gene Expression Regulation , Growth Differentiation Factor 3/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Tretinoin/pharmacology , Activin Receptors, Type I/antagonists & inhibitors , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cluster Analysis , Gene Expression Profiling , Gene Knockdown Techniques , Growth Differentiation Factor 3/metabolism , Humans , Signal Transduction
14.
Ugeskr Laeger ; 174(37): 2138-42, 2012 Sep 10.
Article in Danish | MEDLINE | ID: mdl-22971292

ABSTRACT

Type 2 diabetes (T2D) is a heterogeneous disease with a multifactorial aetiology involving defects in the pancreatic beta cells, liver, muscles, adipose tissue, guts, brain, kidneys and heart. While genetics may only explain a minor proportion of T2D, the contribution of an adverse intrauterine environment may take centre stage in the global propagation of T2D. Impaired expandability of subcutaneous adipose tissue in persons with low birthweight may cause T2D due to lipotoxicity in non-adipose organs. Future implications include a stronger focus on individualized treatments in T2D patients and prevention of T2D in the next generations.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Adipose Tissue/metabolism , Causality , DNA Methylation , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Epigenesis, Genetic , Genetic Predisposition to Disease , Genotype , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , Heat-Shock Proteins/genetics , Humans , Infant, Low Birth Weight/metabolism , Infant, Newborn , Lipid Metabolism , MicroRNAs/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype , Risk Factors , Transcription Factors/genetics
15.
PLoS One ; 7(8): e42424, 2012.
Article in English | MEDLINE | ID: mdl-22879976

ABSTRACT

The CCCTC-binding factor CTCF is the only known vertebrate insulator protein and has been shown to regulate important developmental processes such as imprinting, X-chromosome inactivation and genomic architecture. In this study, we examined the role of CTCF in human embryonic stem cell (hESC) biology. We demonstrate that CTCF associates with several important pluripotency genes, including NANOG, SOX2, cMYC and LIN28 and is critical for hESC proliferation. CTCF depletion impacts expression of pluripotency genes and accelerates loss of pluripotency upon BMP4 induced differentiation, but does not result in spontaneous differentiation. We find that CTCF associates with the distal ends and internal sites of the co-regulated 160 kb NANOG-DPPA3-GDF3 locus. Each of these sites can function as a CTCF-dependent enhancer-blocking insulator in heterologous assays. In hESCs, CTCF exists in multisubunit protein complexes and can be poly(ADP)ribosylated. Known CTCF cofactors, such as Cohesin, differentially co-localize in the vicinity of specific CTCF binding sites within the NANOG locus. Importantly, the association of some cofactors and protein PARlation selectively changes upon differentiation although CTCF binding remains constant. Understanding how unique cofactors may impart specialized functions to CTCF at specific genomic locations will further illuminate its role in stem cell biology.


Subject(s)
Embryonic Stem Cells/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Binding Sites , Biomarkers/metabolism , CCCTC-Binding Factor , Cell Differentiation/genetics , Cell Line , Chromosomal Proteins, Non-Histone , Embryonic Stem Cells/cytology , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genetic Loci/genetics , Growth Differentiation Factor 3/metabolism , Homeodomain Proteins/metabolism , Humans , Models, Biological , Nanog Homeobox Protein , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Protein Binding/genetics , Proteins/metabolism
16.
J Cancer Res Clin Oncol ; 138(6): 1073-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22488170

ABSTRACT

PURPOSE: The aim of this study is to investigate whether GDF3 is related to the progression of human breast cancer and the effects of GDF3 on breast cancer cells. METHODS: The expression of GDF3 in 24 breast cancer specimens paired with corresponding neighboring nontumorous tissue was studied by Western blot. Breast cancer cells were treated with different concentrations of recombinant human GDF3 protein. Using lentivirus containing sh-RNA, we knocked down the expression of GDF3. Soft agar assay was performed to explore the effects of GDF3 on colony formation. Different anti-tumor drugs dealt with MCF-7 cells stably expressing GDF3. RESULTS: We found that GDF3 expression level was significantly down-regulated in breast cancer tissues compared to the surrounding nontumorous tissues. GDF3 proteins could inhibit the proliferation of MCF-7 and T47D cells. We also found that the knockdown of GDF3 resulted in the promotion of colony formation and enhanced the ability of anchorage-independent cell growth in soft agar. Furthermore, overexpression of GDF3 could promote the apoptosis induced by Taxol. CONCLUSIONS: Our data indicated that GDF3 expression is significantly decreased in human breast cancer tissues, and reconstitution of GDF3 in breast cancer may be a potential therapeutic approach to inhibit aggressive growth of breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Growth Differentiation Factor 3/biosynthesis , Paclitaxel/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Down-Regulation/drug effects , Female , Gene Knockdown Techniques/methods , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Stem Cell Assay/methods
17.
Fertil Steril ; 97(4): 979-83, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22305102

ABSTRACT

OBJECTIVE: To examine the relevance of growth differentiation factor 3 (GDF-3) and bone morphogenetic protein (BMP) cytokines in human ovary. DESIGN: Molecular studies. SETTING: Research laboratory. PATIENT(S): Eight women undergoing salpingo-oophorectomy and 30 women undergoing ovarian stimulation for in vitro fertilization. INTERVENTION(S): Localizing GDF-3 protein in human ovaries; granulosa cells (GC) cultured with GDF-3, BMP-6, or BMP-7 followed by RNA extraction. MAIN OUTCOME MEASURE(S): The localization of GDF-3 protein in normal human ovaries via immunohistochemical analysis, GDF-3 messenger RNA (mRNA) expression evaluation via quantitative real-time reverse transcription and polymerase chain reaction (RT-PCR), and evaluation of the effect of GDF-3 on leuteinizing hormone (LH) receptor mRNA expression via quantitative real-time RT-PCR. RESULT(S): In the ovary, BMP cytokines, of the transforming growth factor beta (TGF-ß) superfamily, are known as a luteinization inhibitor by suppressing LH receptor expression in GC. Growth differentiation factor 3, a TGF-ß superfamily cytokine, is recognized as an inhibitor of BMP cytokines in other cells. Immunohistochemical analysis showed that GDF-3 was strongly detected in the GC of antral follicles. An in vitro assay revealed that BMP-6 or BMP-7 induced GDF-3 mRNA in GC. Also, GDF-3 increased LH receptor mRNA expression and inhibited the effect of BMP-7, which suppressed the LH receptor mRNA expression in GC. CONCLUSION(S): GDF-3, induced with BMP-6 and BMP-7, might play a role in folliculogenesis by inhibiting the effect of BMP cytokines.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein 7/metabolism , Granulosa Cells/metabolism , Growth Differentiation Factor 3/metabolism , Luteinizing Hormone/genetics , RNA, Messenger/metabolism , Adult , Cells, Cultured , Female , Humans , Immunohistochemistry , Real-Time Polymerase Chain Reaction , Receptors, LH/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
18.
Cell Death Differ ; 19(6): 1003-12, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22223106

ABSTRACT

Nutrition during early mammalian development permanently influences health of the adult, including increasing the risk of type 2 diabetes and coronary heart disease. However, the molecular mechanisms underlying such programming are poorly defined. Here we demonstrate that programmed changes in miRNA expression link early-life nutrition to long-term health. Specifically, we show that miR-483-3p is upregulated in adipose tissue from low-birth-weight adult humans and prediabetic adult rats exposed to suboptimal nutrition in early life. We demonstrate that manipulation of miR-483-3p levels in vitro substantially modulates the capacity of adipocytes to differentiate and store lipids. We show that some of these effects are mediated by translational repression of growth/differentiation factor-3, a target of miR-483-3p. We propose that increased miR-483-3p expression in vivo, programmed by early-life nutrition, limits storage of lipids in adipose tissue, causing lipotoxicity and insulin resistance and thus increasing susceptibility to metabolic disease.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet , Growth Differentiation Factor 3/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Adult , Animals , Animals, Newborn , Base Sequence , Cell Differentiation , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Down-Regulation , Female , Growth Differentiation Factor 3/antagonists & inhibitors , Growth Differentiation Factor 3/genetics , HEK293 Cells , Humans , Lipid Metabolism , Male , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Wistar
19.
Mol Cell Biochem ; 359(1-2): 115-23, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21805089

ABSTRACT

Members of the transforming growth factor-ß (TGF-ß) superfamily have significant roles in the regulation of a wide variety of physiological processes. In our present work, phylogenetic tree analysis showed that human GDF3 (Growth and differentiation factor 3) and human GDF1 formed a subgroup of closely related molecules. Through quantitative real-time PCR analysis in different human tissues, GDF1 and GDF3 expression level had a big difference in brain. GDF3 could activate downstream signaling through associating with ALK7 (Activin receptor-like kinase 7) in a Cripto-dependent fashion. A CHO cell line stably transfected with the encoding sequence of GDF3, named CHO-GDF3, was established. Western blotting analysis demonstrated that GDF3 protein could be secreted into the medium from CHO cells and immunofluorescence experiment showed that GDF3 was mainly distributed in cytoplasm of the stable cell line, the primary hippocampal neurons, and brain tissues. Furthermore, the conditioned medium from CHO-GDF3 could reduce PC12 cell growth and induce cell differentiation. All these findings bring new insights into the functional study of GDF3.


Subject(s)
Brain/metabolism , Cell Differentiation/drug effects , Growth Differentiation Factor 3/pharmacology , Activin Receptors, Type I/metabolism , Animals , Brain/cytology , Brain Chemistry , CHO Cells , Cricetinae , Culture Media, Conditioned/pharmacology , Growth Differentiation Factor 1 , Growth Differentiation Factor 3/metabolism , Humans , PC12 Cells , Rats , Signal Transduction , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...