Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 191
Filter
Add more filters

Publication year range
1.
Circ Res ; 125(9): 834-846, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31495264

ABSTRACT

RATIONALE: Pathogenic variations in the lamin gene (LMNA) cause familial dilated cardiomyopathy (DCM). LMNA insufficiency caused by LMNA pathogenic variants is believed to be the basic mechanism underpinning LMNA-related DCM. OBJECTIVE: To assess whether silencing of cardiac Lmna causes DCM and investigate the role of Yin Yang 1 (Yy1) in suppressing Lmna DCM. METHODS AND RESULTS: We developed a Lmna DCM mouse model induced by cardiac-specific Lmna short hairpin RNA. Silencing of cardiac Lmna induced DCM with associated cardiac fibrosis and inflammation. We demonstrated that upregulation of Yy1 suppressed Lmna DCM and cardiac fibrosis by inducing Bmp7 expression and preventing upregulation of Ctgf. Knockdown of upregulated Bmp7 attenuated the suppressive effect of Yy1 on DCM and cardiac fibrosis. However, upregulation of Bmp7 alone was not sufficient to suppress DCM and cardiac fibrosis. Importantly, upregulation of Bmp7 together with Ctgf silencing significantly suppressed DCM and cardiac fibrosis. Mechanistically, upregulation of Yy1 regulated Bmp7 and Ctgf reporter activities and modulated Bmp7 and Ctgf gene expression in cardiomyocytes. Downregulation of Ctgf inhibited TGF-ß (transforming growth factor-ß)/Smad signaling in DCM hearts. Regulation of both Bmp7 and Ctgf further suppressed TGFß/Smad signaling. In addition, co-modulation of Bmp7 and Ctgf reduced CD3+ T cell numbers in DCM hearts. CONCLUSIONS: Our findings demonstrate that upregulation of Yy1 or co-modulation of Bmp7 and Ctgf offer novel therapeutic strategies for the treatment of DCM caused by LMNA insufficiency.


Subject(s)
Bone Morphogenetic Protein 7/biosynthesis , Cardiomyopathies/metabolism , Cardiomyopathies/prevention & control , Connective Tissue Growth Factor/biosynthesis , YY1 Transcription Factor/biosynthesis , Animals , Bone Morphogenetic Protein 7/genetics , Cardiomyopathies/genetics , Connective Tissue Growth Factor/genetics , Endothelium, Vascular/metabolism , Fibrosis/genetics , Fibrosis/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , YY1 Transcription Factor/genetics
2.
Cell Tissue Res ; 379(3): 549-559, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31773302

ABSTRACT

Diabetic nephropathy (DN) is one of the most devastating complications of diabetes. Connective tissue growth factor (CTGF) levels are up-regulated in patients with DN and in renal tubular epithelial cells (RTECs) exposed to high glucose (HG). The underlying epigenetic mechanism remains to be elucidated. In the present study, we investigate the role of myocardin-related transcription factor A (MRTF-A) in HG-induced CTGF transcription in RTECs. We report that in two different animal models of DN, one induced by streptozotocin (STZ) injection and the other induced by high-fat diet (HFD) feeding, MRTF-A deficiency attenuated CTGF induction in the kidneys. In cultured RTECs, MRTF-A knockdown similarly ameliorated CTGF induction by HG treatment. Upon CTGF induction, there was an increase in acetylated histone H3 (AcH3) and trimethylated H3K4 (H3K4Me3) on the CTGF promoter region accompanying a decrease in dimethylated H3K9 (H3K9Me2). MRTF-A ablation in vivo or depletion in vitro comparably dampened the accumulation of AcH3 and H3K4Me3 but restored H3K9Me2 on the CTGF promoter. Further analyses revealed that MRTF-A interacted with and recruited histone demethylase KDM3A to the CTGF promoter to activate transcription. KDM3A silencing equivalently weakened HG-induced CTGF induction in RTECs. In conclusion, MRTF-A contributes to HG-induced CTGF transcription via an epigenetic mechanism.


Subject(s)
Connective Tissue Growth Factor/genetics , Diabetic Nephropathies/metabolism , Trans-Activators/metabolism , Animals , Connective Tissue Growth Factor/biosynthesis , Diabetic Nephropathies/genetics , Disease Models, Animal , Epigenomics/methods , Epithelial Cells/metabolism , Glucose/administration & dosage , Glucose/metabolism , HEK293 Cells , Humans , Kidney Tubules/cytology , Kidney Tubules/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription, Genetic , Up-Regulation
3.
FASEB J ; 33(2): 2047-2057, 2019 02.
Article in English | MEDLINE | ID: mdl-30216109

ABSTRACT

In skeletal muscle, extracellular matrix (ECM) remodeling can either support the complete regeneration of injured muscle or facilitate pathologic fibrosis and muscle degeneration. Muscular dystrophy (MD) is a group of genetic disorders that results in a progressive decline in muscle function and is characterized by the abundant deposition of fibrotic tissue. Unlike acute injury, where ECM remodeling is acute and transient, in MD, remodeling persists until fibrosis obstructs the regenerative efforts of diseased muscles. Thus, understanding how ECM is deposited and organized is critical in the context of muscle repair. Connective tissue growth factor (CTGF or CCN2) is a matricellular protein expressed by multiple cell types in response to tissue injury. Although used as a general marker of fibrosis, the cell type-dependent role of CTGF in dystrophic muscle has not been elucidated. To address this question, a conditional Ctgf myofiber and fibroblast-knockout mouse lines were generated and crossed to a dystrophic background. Only myofiber-selective inhibition of CTGF protected δ-sarcoglycan-null ( Sgcd-/-) mice from the dystrophic phenotype, and it did so by affecting collagen organization in a way that allowed for improvements in dystrophic muscle regeneration and function. To confirm that muscle-specific CTGF functions to mediate collagen organization, we generated mice with transgenic muscle-specific overexpression of CTGF. Again, genetic modulation of CTGF in muscle was not sufficient to drive fibrosis, but altered collagen content and organization after injury. Our results show that the myofibers are critical mediators of the deleterious effects associated with CTGF in MD and acutely injured skeletal muscle.-Petrosino, J. M., Leask, A., Accornero, F. Genetic manipulation of CCN2/CTGF unveils cell-specific ECM-remodeling effects in injured skeletal muscle.


Subject(s)
Connective Tissue Growth Factor , Extracellular Matrix , Gene Expression Regulation , Muscle Fibers, Skeletal , Muscular Dystrophy, Animal , Animals , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Sarcoglycans/deficiency
4.
FASEB J ; 33(11): 12554-12564, 2019 11.
Article in English | MEDLINE | ID: mdl-31451010

ABSTRACT

Mycobacterium tuberculosis (M.tb) infection in lung causes pulmonary fibrosis, which leads to the irreversible reduction of pulmonary function. Fibrotic protein connective tissue growth factor (CTGF) expression has been confirmed to play a crucial role in lung fibrosis. However, the underlying signal pathway and effect of M.tb on CTGF expression in human lung fibroblasts are unclear. Our results revaled that M.tb caused time- and concentration-dependent increases in CTGF expression in human lung fibroblasts. A mechanistic investigation revealed that M.tb induced CTGF expression through TLR2 but not TLR4. The promoter activity assay indicated that M.tb-induced CTGF activity was mainly controlled by the promoter region at -747 to -184 bp, which contained signal transducer and activator of transcription 3 and activator protein 1 (AP-1) binding sites. Moreover, curcumin (AP-1 inhibitor) restrained M.tb-induced CTGF expression. M.tb also induced increases in AP-1 luciferase activity and DNA binding activity of c-Jun and c-Fos on the CTGF promoter. Furthermore, the knockdown of c-Jun by small interfering RNA attenuated M.tb-induced CTGF expression and AP-1 luciferase activity. A JNK inhibitor (SP600125) and a JNK dominant-negative mutant suppressed M.tb-induced CTGF expression. We also discovered that M.tb could induce the phosphorylation of JNK and c-Jun. Furthermore, SP600125 inhibited M.tb-induced c-Jun phosphorylation and AP-1- luciferase activity. M.tb-induced fibronectin expression was inhibited by anti-CTGF antibody. These results demonstrate that M.tb is activated through TLR2 to induce JNK activation, further increasing the DNA binding activity of c-Jun and c-Fos and finally inducing CTGF expression and extracellular matrix production.-Lee, H.-S., Hua, H.-S., Wang, C.-H., Yu, M.-C., Chen, B.-C., Lin, C.-H. Mycobacterium tuberculosis induces connective tissue growth factor expression through the TLR2-JNK-AP-1 pathway in human lung fibroblasts.


Subject(s)
Connective Tissue Growth Factor/biosynthesis , Fibroblasts/metabolism , Lung/metabolism , MAP Kinase Kinase 4/metabolism , Mycobacterium tuberculosis/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Transcription Factor AP-1/metabolism , Tuberculosis, Pulmonary/metabolism , Anthracenes/pharmacology , Cell Line , Connective Tissue Growth Factor/genetics , Fibroblasts/pathology , Gene Expression Regulation/drug effects , Humans , Lung/pathology , MAP Kinase Kinase 4/antagonists & inhibitors , Response Elements , Tuberculosis, Pulmonary/pathology
5.
Appl Microbiol Biotechnol ; 104(23): 9979-9990, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33074415

ABSTRACT

Human connective tissue growth factor (CTGF) is a secreted cysteine-rich peptide that stimulates cell proliferation, migration, and extracellular matrix production during tissue development, differentiation, angiogenesis, implantation, wound healing, and fibrosis processes, with broad application in the medical and cosmetic medical fields. However, the production of CTGF is currently limited by its low yield and purity in current bioreactors. In this study, two genetically modified silkworm strains were generated harboring artificially designed CTGF-8ht and pepCTGF-8ht genes, respectively, that contain an enhanced His-tag with eight histidine residues with or without a transdermal peptide (pep). Both recombinant CTGF-8ht and pepCTGF-8ht proteins were successfully expressed in the silkworm silk gland and cocoon, and could be easily extracted and purified from the cocoon by single-affinity immunoprecipitation column chromatography, achieving a purity of more than 95%. Moreover, compared with CTGF-8ht protein, pepCTGF-8ht protein exhibited better cell proliferation activity by activating the extracellular signal-regulated kinase (ERK) pathway and enhanced hyaluronic acid synthesis activity by upregulating hyaluronan synthase 3 expression; moreover, the addition of pep significantly improved the transmembrane ability of CTGF-8ht protein. These results should help to promote the application prospects of CTGF and further guide the design and development of protein drugs from silkworm and other bioreactor systems. KEY POINTS : A silkworm bioreactor was optimized to produce connective tissue growth factor (CTGF) The transgene contained an enhanced 8-His-tag and transmembrane peptide (pep) Recombinant CTGF was easily purified with maintained or higher biological activity.


Subject(s)
Bioreactors , Bombyx , Connective Tissue Growth Factor/biosynthesis , Animals , Cell Proliferation , Connective Tissue Growth Factor/genetics , Humans , Hyaluronic Acid , Recombinant Fusion Proteins/biosynthesis , Silk
6.
Am J Physiol Renal Physiol ; 317(4): F839-F851, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31364374

ABSTRACT

The small GTPase Rho and its effector Rho kinase (ROCK) are involved in the pathogenesis of diabetic kidney disease. Rho kinase has two isoforms: ROCK1 and ROCK2. However, it remains unclear which is mainly involved in the progression of diabetic glomerulosclerosis and the regulation of profibrotic mediators. Glomeruli isolated from type 2 diabetic db/db mice demonstrated increased gene expression of transforming growth factor (TGF)-ß and its downstream profibrotic mediators. Chemical inhibition of ROCK suppressed the expression of profibrotic mediators in both isolated glomeruli and cultured mesangial cells. An investigation of mechanisms underlying this observation revealed activated ROCK functions through the phosphorylation of JNK and Erk and the nuclear translocation of NF-κB via actin dynamics. Knockdown by siRNA against ROCK1 and ROCK2 showed that ROCK2 but not ROCK1 controls this fibrotic machinery. Further in vivo experiments showed that ROCK2 activity in the renal cortex of db/db mice was elevated compared with control db/m mice. Importantly, oral administration of ROCK2 inhibitor attenuated renal ROCK2 activity, albuminuria, and glomerular fibrosis in db/db mice. These observations indicate that ROCK2 is a key player in the development of diabetic renal injury. Glomerular ROCK2 may be a potential therapeutic target for the treatment of diabetic kidney disease.


Subject(s)
Connective Tissue Growth Factor/biosynthesis , Cytoskeleton/metabolism , Fibrosis/genetics , Glomerular Mesangium/metabolism , NF-kappa B/biosynthesis , Transforming Growth Factor beta/pharmacology , rho-Associated Kinases/metabolism , Actins/metabolism , Animals , Diabetic Nephropathies/metabolism , Enzyme Activation , Glomerular Mesangium/cytology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred NOD , rho-Associated Kinases/antagonists & inhibitors
7.
Biochem Biophys Res Commun ; 519(2): 309-315, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31506177

ABSTRACT

Jiadifenolide has been reported to have neurotrophin-like activity in primary rat cortical neurons, and also possesses neurotrophic effects in neuronal precursor cells derived from human induced pluripotent stem cells (hiPSCs), as we have previously reported. However, the molecular mechanisms by which jiadifenolide exerts its neurotrophic effects in rat and human neurons are unknown. Thus, we aimed to investigate the molecular mechanisms and pathways by which jiadifenolide promotes neurotrophic effects. Here, we found that jiadifenolide activated cellular communication network factor (CCN) signaling pathways by up-regulating mRNA level expression of CCN genes in human neuronal cells. We also found that CCN2 (also known as connective tissue growth factor, CTGF) protein promotes neurotrophic effects through activation of the p44/42 mitogen-activated protein kinase signaling pathway. This is the first discovery which links neurotrophic activity with CCN signaling.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Connective Tissue Growth Factor/biosynthesis , Induced Pluripotent Stem Cells/drug effects , Sesquiterpenes/pharmacology , Cells, Cultured , Connective Tissue Growth Factor/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Sesquiterpenes/chemical synthesis , Sesquiterpenes/chemistry
8.
Circ Res ; 120(2): 354-365, 2017 Jan 20.
Article in English | MEDLINE | ID: mdl-27923814

ABSTRACT

RATIONALE: Endothelial-mesenchymal transition (EndoMT) is implicated in myofibroblast-like cell-mediated damage to the coronary arterial wall in acute Kawasaki disease (KD) patients, as evidenced by positive staining for connective tissue growth factor (CTGF) and EndoMT markers in KD autopsy tissues. However, little is known about the molecular basis of EndoMT involved in KD. OBJECTIVE: We investigated the microRNA (miRNA) regulation of CTGF and the consequent EndoMT in KD pathogenesis. As well, the modulation of this process by statin therapy was studied. METHODS AND RESULTS: Sera from healthy children and KD subjects were incubated with human umbilical vein endothelial cells. Cardiovascular disease-related miRNAs, CTGF, and EndoMT markers were quantified using reverse transcriptase quantitative polymerase chain reaction, ELISA, and Western blotting. Compared with healthy controls, human umbilical vein endothelial cell incubated with sera from acute KD patients had decreased miR-483, increased CTGF, and increased EndoMT markers. Bioinformatics analysis followed by functional validation demonstrated that Krüppel-like factor 4 (KLF4) transactivates miR-483, which in turn targets the 3' untranslated region of CTGF mRNA. Overexpression of KLF4 or pre-miR-483 suppressed, whereas knockdown of KLF4 or anti-miR-483 enhanced, CTGF expression in endothelial cells in vitro and in vivo. Furthermore, atorvastatin, currently being tested in a phase I/IIa clinical trial in KD children, induced KLF4-miR-483, which suppressed CTGF and EndoMT in endothelial cells. CONCLUSIONS: KD sera suppress the KLF4-miR-483 axis in endothelial cells, leading to increased expression of CTGF and induction of EndoMT. This detrimental process in the endothelium may contribute to coronary artery abnormalities in KD patients. Statin therapy may benefit acute KD patients, in part, through the restoration of KLF4-miR-483 expression. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01431105.


Subject(s)
Atorvastatin/administration & dosage , Connective Tissue Growth Factor/biosynthesis , Epithelial-Mesenchymal Transition/physiology , Gene Targeting/methods , MicroRNAs/biosynthesis , Mucocutaneous Lymph Node Syndrome/blood , Mucocutaneous Lymph Node Syndrome/therapy , Animals , Cattle , Child, Preschool , Connective Tissue Growth Factor/genetics , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Infant , Kruppel-Like Factor 4 , Male , Mice , Mice, Transgenic , MicroRNAs/administration & dosage , MicroRNAs/genetics , Mucocutaneous Lymph Node Syndrome/genetics
9.
Retina ; 39(2): 265-273, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29190236

ABSTRACT

PURPOSE: We sought to characterize the angiofibrotic and apoptotic effects of vascular endothelial growth factor (VEGF)-inhibition on fibrovascular epiretinal membranes in eyes with traction retinal detachment because of proliferative diabetic retinopathy. METHODS: Membranes were excised from 20 eyes of 19 patients (10 randomized to intravitreal bevacizumab, 10 controls) at vitrectomy. Membranes were stained with antibodies targeting connective tissue growth factor (CTGF) or VEGF and colabeled with antibodies directed against endothelial cells (CD31), myofibroblasts, or retinal pigment epithelium markers. Quantitative and colocalization analyses of antibody labeling were obtained through immunofluorescence confocal microscopy. Masson trichrome staining, cell counting of hematoxylin and eosin sections, and terminal dUTP nick-end labeling staining were performed. RESULTS: High levels of fibrosis were observed in both groups. Cell apoptosis was higher (P = 0.05) in bevacizumab-treated membranes compared with controls. The bevacizumab group had a nonsignificant reduction in colocalization in CD31-CTGF and cytokeratin-VEGF studies compared with controls. Vascular endothelial growth factor in extracted membranes was positively correlated with vitreous levels of VEGF; CTGF in extracted membranes was negatively correlated with vitreous levels of CTGF. CONCLUSION: Bevacizumab suppresses vitreous VEGF levels, but does not significantly alter VEGF or CTGF in diabetic membranes that may be explained by high baseline levels of fibrosis. Bevacizumab may cause apoptosis within fibrovascular membranes.


Subject(s)
Apoptosis , Bevacizumab/administration & dosage , Diabetic Retinopathy/pathology , Epiretinal Membrane/surgery , Retina/pathology , Vitrectomy/methods , Actins/biosynthesis , Angiogenesis Inhibitors/administration & dosage , Cell Proliferation , Connective Tissue Growth Factor/biosynthesis , Diabetic Retinopathy/complications , Diabetic Retinopathy/drug therapy , Epiretinal Membrane/complications , Epiretinal Membrane/pathology , Fibrosis/pathology , Humans , Intravitreal Injections , Keratins/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Prospective Studies , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Retina/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
10.
J Biol Chem ; 292(18): 7542-7553, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28320863

ABSTRACT

The aim of the study was to characterize the role of Rac1 GTPase for the mineralocorticoid receptor (MR)-mediated pro-fibrotic remodeling. Transgenic mice with cardiac overexpression of constitutively active Rac1 (RacET) develop an age-dependent phenotype with atrial dilatation, fibrosis, and atrial fibrillation. Expression of MR was similar in RacET and WT mice. The expression of 11ß hydroxysteroid dehydrogenase type 2 (11ß-HSD2) was age-dependently up-regulated in the atria and the left ventricles of RacET mice on mRNA and protein levels. Statin treatment inhibiting Rac1 geranylgeranylation reduced 11ß-HSD2 up-regulation. Samples of human left atrial myocardium showed a positive correlation between Rac1 activity and 11ß-HSD2 expression (r = 0.7169). Immunoprecipitation showed enhanced Rac1-bound 11ß-HSD2 relative to Rac1 expression in RacET mice that was diminished with statin treatment. Both basal and phorbol 12-myristate 13-acetate (PMA)-induced NADPH oxidase activity were increased in RacET and correlated positively with 11ß-HSD2 expression (r = 0.788 and r = 0.843, respectively). In cultured H9c2 cardiomyocytes, Rac1 activation with l-buthionine sulfoximine increased; Rac1 inhibition with NSC23766 decreased 11ß-HSD2 mRNA and protein expression. Connective tissue growth factor (CTGF) up-regulation induced by aldosterone was prevented with NSC23766. Cardiomyocyte transfection with 11ß-HSD2 siRNA abolished the aldosterone-induced CTGF up-regulation. Aldosterone-stimulated MR nuclear translocation was blocked by the 11ß-HSD2 inhibitor carbenoxolone. In cardiac fibroblasts, nuclear MR translocation induced by aldosterone was inhibited with NSC23766 and spironolactone. NSC23766 prevented the aldosterone-induced proliferation and migration of cardiac fibroblasts and the up-regulation of CTGF and fibronectin. In conclusion, Rac1 GTPase regulates 11ß-HSD2 expression, MR activation, and MR-mediated pro-fibrotic signaling.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Endomyocardial Fibrosis/enzymology , Fibroblasts/enzymology , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Neuropeptides/biosynthesis , Signal Transduction , rac1 GTP-Binding Protein/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Aldosterone/pharmacology , Animals , Cell Line , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Endomyocardial Fibrosis/pathology , Fibroblasts/pathology , Fibronectins/biosynthesis , Fibronectins/genetics , Gene Expression Regulation/drug effects , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mice, Mutant Strains , Myocardium/pathology , Myocytes, Cardiac/pathology , Neuropeptides/genetics , Rats , Rats, Sprague-Dawley , Sulfoxides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , rac1 GTP-Binding Protein/genetics
11.
J Cell Physiol ; 233(3): 2067-2074, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28294324

ABSTRACT

Fibrosis of the subsynovial connective tissue (SSCT) in carpal tunnel syndrome (CTS) patients is increasingly recognized as an important aspect of CTS pathophysiology. In this study, we evaluated the effect of blocking profibrotic pathways in fibroblasts from the SSCT in CTS patients. Fibroblasts were stimulated with transforming growth factor ß1 (TGF-ß1), and then treated either with a specific fibrosis pathway inhibitor targeting TGF-ß receptor type 1 (TßRI), platelet-derived growth factor receptor (PDGFR), epidermal growth factor receptor (EGFR), or vascular endothelial growth factor receptor (VEGFR). Fibrosis array and quantitative real-time polymerase chain reaction of fibrotic genes were evaluated. Array gene expression analysis revealed significant down-regulation of multiple fibrotic genes after treatment with TßRI, PDGFR, and VEGFR inhibitors. No array fibrotic genes were significantly down-regulated with EGFR inhibition. Further gene expression analysis of known CTS fibrosis markers collagen type I A2 (Col1), collagen type III A1 (Col3), connective tissue growth factor (CTGF), and SERPINE1 showed significantly down-regulation after TßRI inhibition. In contrast, VEGFR inhibition significantly down-regulated CTGF and SERPINE1, whereas, PDGFR and EGFR inhibition significantly down-regulated Col3. Taken together the inhibition of TßRI appears to be the primary mediator of fibrotic gene expression in fibroblasts from CTS patients. TGF-ß/Smad activity was further evaluated, and as expected inhibition of Smad activity was significantly down-regulated after inhibition of TßRI, but not with PDGFR, VEGFR, or EGFR inhibition. These results indicate that local therapies specifically targeting TGF-ß signaling alone or in combination offer the potential of a novel local antifibrosis therapy for patients with CTS.


Subject(s)
Carpal Tunnel Syndrome/drug therapy , ErbB Receptors/antagonists & inhibitors , Fibrosis/pathology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Synovial Membrane/pathology , Transforming Growth Factor beta/metabolism , Carpal Tunnel Syndrome/pathology , Cells, Cultured , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type III/biosynthesis , Collagen Type III/genetics , Connective Tissue/pathology , Connective Tissue Cells/cytology , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Fibroblasts/metabolism , Fibrosis/drug therapy , Humans , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Synovial Membrane/cytology
12.
Rheumatology (Oxford) ; 57(9): 1675-1684, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29905853

ABSTRACT

Objectives: To investigate the mechanism of 2-methoxyestradiol (2-ME) in inhibiting hypoxia-induced collagen synthesis of fibroblasts in SSc. Methods: The expressions of hypoxia-inducible factor 1 alpha (HIF-1α) and connective tissue growth factor (CTGF) in skin specimens derived from SSc patients and healthy volunteers were examined by immunohistochemistry. HIF-1α was knocked down by lentiviral transduction, and SSc dermal fibroblasts cultured under normoxic (21% O2) or hypoxic (1% O2) condition were treated with PI3K inhibitor LY294002, rapamycin or 2-ME (25 µM). The protein levels of HIF-1α, CTGF, collagen I, p-Akt and p-mTOR were examined by western blotting or immunofluorescence. Apoptosis and cell cycle of fibroblasts were assessed by flow cytometry and by measuring caspase 3 activity, and cell proliferation was evaluated by Cell Counting Kit-8. Results: The expressions of HIF-1α and CTGF were increased in skins of SSc patients compared with healthy controls. Hypoxia up-regulated the protein levels of HIF-1α, CTGF and collagen I in SSc fibroblasts. In contrast, 2-ME inhibited PI3K/Akt/mTOR pathway and down-regulated protein levels of HIF-1α, CTGF and collagen I. Knockdown of HIF-1α reduced expressions of CTGF and collagen I, which were further down-regulated by 2-ME intervention. Moreover, 2-ME promoted the apoptosis and inhibited the proliferation of SSc fibroblasts by arresting the cell cycle at the G2/M phase. Conclusion: 2-ME reduced the production of CTGF and collagen I in SSc fibroblasts induced by hypoxia through PI3K/Akt/mTOR/HIF-1α signalling and inhibited the proliferation of fibroblasts. These findings suggested that 2-ME could be employed as a promising antifibrotic therapy for SSc.


Subject(s)
Collagen Type I/biosynthesis , Estradiol/analogs & derivatives , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Scleroderma, Systemic/genetics , TOR Serine-Threonine Kinases/genetics , 2-Methoxyestradiol , Cell Proliferation , Cells, Cultured , Collagen Type I/drug effects , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/drug effects , Estradiol/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/biosynthesis , RNA/genetics , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/metabolism , Signal Transduction , Skin/metabolism , Skin/pathology , TOR Serine-Threonine Kinases/biosynthesis , Tubulin Modulators/pharmacology
13.
Medicina (Kaunas) ; 54(5)2018 Nov 19.
Article in English | MEDLINE | ID: mdl-30463213

ABSTRACT

Background and objectives: Energy drinks are popular non-alcoholic beverages. They are consumed in large amounts, mainly by active, young people. Although they are easily accessible and marketed as safe, numerous cases of adverse effects have been published, including cardiac arrest, arrythmias, acute hepatitis, and renal failure. The aim of the current study is the assessment of energy drink influence on the histological structure of adrenal cortex in rats. Material and Methods: 15 male young Wistar rats were equally divided into three groups: control (C), experimental (E) and reversibility control (RC). C group received water and standard rodent food ad libitum while both E and RC groups had additionally unlimited access to energy drinks. C and E groups were decapitated after 8 weeks and RC was given another 8 weeks without energy drinks. Adrenal glands were embedded in paraffin blocks and 5 µm slides were prepared and stained according to standard H&E and Masson's trichrome protocols. Additionally, immunohistochemical stainings against Ki-67, p53, CTGF and caspase-3 were prepared. Results: Decreased vacuolization and numerous pyknotic nuclei were noted in E and RC groups. Overexpression of caspase-3 was noted both subcapsular in zona glomerulosa and along sinusoids in zona fasciculata. Increased collagen deposition in zona glomerulosa and zona fasciculata of E and RC was observed. Insular and irregular overexpression of CTGF was noted. The overall picture of CTGF expression matched the Masson's trichrome. No significant difference was observed in Ki-67 expression. Conclusions: The results of the current study suggest that the stimulation is so intense that it causes significant damage to adrenal cortical cells, resulting in their apoptosis. It seems, however, that the observed effects are at least partially reversible.


Subject(s)
Caffeine/adverse effects , Energy Drinks/adverse effects , Lipid Droplets , Taurine/adverse effects , Zona Fasciculata/metabolism , Zona Fasciculata/pathology , Zona Glomerulosa/metabolism , Zona Glomerulosa/pathology , Animals , Apoptosis , Caspase 3/biosynthesis , Collagen/biosynthesis , Connective Tissue Growth Factor/biosynthesis , Ki-67 Antigen/biosynthesis , Male , Rats , Rats, Wistar , Zona Fasciculata/cytology , Zona Glomerulosa/cytology
14.
Biochim Biophys Acta ; 1863(8): 2027-36, 2016 08.
Article in English | MEDLINE | ID: mdl-27155083

ABSTRACT

Pharmacological inhibition of oxygen sensing prolyl hydroxylase domain enzymes (PHDs) has been shown to preserve renal structure and function in various models of kidney disease. Since transforming growth factor ß-1 (TGFß-1) is one of the major mediators of kidney injury, we investigated if inhibition of PHDs with subsequent stabilization of hypoxia inducible transcription factors (HIF) might interfere with TGFß-1 signaling with special emphasis on its target gene connective tissue growth factor (CTGF). Overnight incubation of human renal tubular cells, primary cells and cell lines, with the PDH inhibitor DMOG increased Smad3 expression, but barely affected Smad2. Both Smads were translocated into the nucleus upon activation of the cells with TGFß-1. Interestingly, Smad3 nuclear localization was enhanced upon pretreatment of the cells with DMOG for several hours, whereas nuclear Smad2 was reduced. This differential localization was independent of Smad2/3 phosphorylation. Reduced nuclear Smad2 correlated with impaired CTGF secretion in DMOG-treated cells and transient downregulation of Smad2 interfered with TGFß-1-induced CTGF synthesis. Furthermore, YAP was confirmed as indispensable transcription factor involved in CTGF synthesis. Nuclear localization of YAP and TAZ was reduced in DMOG-treated cells. Our data thus provide evidence for DMOG-mediated reduction of CTGF expression by regulating the nuclear localization of the transcription factors Smad2, YAP and TAZ. Prolonged inhibition of PHDs was necessary to achieve alterations in cellular localization suggesting an indirect HIF-mediated effect. This mechanism might be extended to other transcription factors and target genes, and may thus represent a novel mechanism of negative regulation of gene expression by PHD inhibition.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Connective Tissue Growth Factor/biosynthesis , Kidney Tubules/metabolism , Phosphoproteins/metabolism , Prolyl Hydroxylases/physiology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Amino Acids, Dicarboxylic/pharmacology , Cell Hypoxia/genetics , Cells, Cultured , Connective Tissue Growth Factor/antagonists & inhibitors , Connective Tissue Growth Factor/physiology , Gene Expression Regulation/drug effects , Humans , Kidney Tubules/cytology , Oxygen/metabolism , Phosphoproteins/genetics , Primary Cell Culture , RNA Interference , RNA, Small Interfering/genetics , Smad2 Protein/genetics , Transforming Growth Factor beta1/physiology , YAP-Signaling Proteins
15.
Connect Tissue Res ; 58(5): 407-413, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27600173

ABSTRACT

PURPOSES: The adult human anterior cruciate ligament (ACL) has poor functional healing response. Hypoxia plays an important role in regulating the microenvironment of the joint cavity after ACL injury, however, its role in mechanical injury is yet to be examined fully in ACL fibroblasts. In this study, we used CoCl2 to induce Hypoxia-inducible factor-1α (HIF-1α) in our experimental model to study its affect on matrix metalloproteinase-2 (MMP-2), vascular endothelial growth factor (VEGF), and connective tissue growth factor (CTGF) expression in ACL fibroblasts after mechanical stretch. MATERIALS AND METHODS: Cell treatments were performed in the stretch chamber in all experimental groups. Quantitative real-time PCR was used to check mRNA expression levels of MMP-2, CTGF, VEGF, and HIF-1α. Western blot was used to detect the HIF-1α production. Enzyme-Linked immunosorbent assay was performed to check the VEGF and CTGF protein contents in supernatant. MMP-2 activity was assayed by gelatin zymography. RESULTS: The real-time PCR results show that mechanical stretch or CoCl2 treatment increases the expression of MMP-2, VEGF, CTGF, and HIF-1α; however, the combined effects of mechanical stretch and CoCl2-induced HIF-1α increased MMP-2 production but decreased the VEGF and CTGF expression, compared to the CoCl2 treatment group alone. Western blot analysis and ELISA also confirmed these results. CONCLUSIONS: Our results demonstrated that mechanical stretch and CoCl2-induced HIF-1α together increased the level of MMP-2 and decreased the levels of VEGF and CTGF in cultured ACL fibroblasts. The differential expression and production of HIF-1α, VEGF, MMP-2, and CTGF might help to explain the poor healing ability of ACL.


Subject(s)
Anterior Cruciate Ligament/metabolism , Connective Tissue Growth Factor/biosynthesis , Fibroblasts/metabolism , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Stress, Mechanical , Vascular Endothelial Growth Factor A/biosynthesis , Adult , Anterior Cruciate Ligament/cytology , Cells, Cultured , Cobalt/pharmacology , Female , Fibroblasts/cytology , Humans , Male , Matrix Metalloproteinase 2/biosynthesis , Middle Aged
16.
Proc Natl Acad Sci U S A ; 111(46): 16496-501, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25369932

ABSTRACT

High temperature requirement protein A1 (HtrA1) is a primarily secreted serine protease involved in a variety of cellular processes including transforming growth factor ß (TGF-ß) signaling. Loss of its activity causes cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL), an inherited form of cerebral small vessel disease leading to early-onset stroke and premature dementia. Dysregulated TGF-ß signaling is considered to promote CARASIL pathogenesis, but the underlying molecular mechanisms are incompletely understood. Here we present evidence from mouse brain tissue and embryonic fibroblasts as well as patient skin fibroblasts for a facilitating role of HtrA1 in TGF-ß pathway activation. We identify latent TGF-ß binding protein 1 (LTBP-1), an extracellular matrix protein and key regulator of TGF-ß bioavailability, as a novel HtrA1 target. Cleavage occurs at physiological protease concentrations, is prevented under HtrA1-deficient conditions as well as by CARASIL mutations and disrupts both LTBP-1 binding to fibronectin and its incorporation into the extracellular matrix. Hence, our data suggest an attenuation of TGF-ß signaling caused by a lack of HtrA1-mediated LTBP-1 processing as mechanism underlying CARASIL pathogenesis.


Subject(s)
Alopecia/genetics , Cerebral Infarction/genetics , Latent TGF-beta Binding Proteins/physiology , Leukoencephalopathies/genetics , Serine Endopeptidases/physiology , Spinal Diseases/genetics , Transforming Growth Factor beta1/physiology , Alopecia/metabolism , Animals , Brain/metabolism , Cells, Cultured , Cerebral Infarction/metabolism , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Fibroblasts/metabolism , Fibronectins/metabolism , Gene Expression Regulation , HEK293 Cells , High-Temperature Requirement A Serine Peptidase 1 , Humans , Latent TGF-beta Binding Proteins/genetics , Leukoencephalopathies/metabolism , Mice , Mice, Knockout , Mutation, Missense , Point Mutation , Protein Binding , Protein Interaction Mapping , Protein Processing, Post-Translational , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Serpin E2/biosynthesis , Serpin E2/genetics , Signal Transduction , Skin , Spinal Diseases/metabolism , Transfection
17.
Mol Cell Biochem ; 416(1-2): 99-108, 2016 May.
Article in English | MEDLINE | ID: mdl-27075467

ABSTRACT

Post-traumatic hypertrophic scar (HS) is a fibrotic disease with excessive extracellular matrix (ECM) production, which is a response to tissue injury by fibroblasts. Although emerging evidence has indicated that miRNA contributes to hypertrophic scarring, the role of miRNA in HS formation remains unclear. In this study, we found that miR-143-3p was markedly downregulated in HS tissues and fibroblasts (HSFs) using qRT-PCR. The expression of connective tissue growth factor (CTGF/CCN2) was upregulated both in HS tissues and HSFs, which is proposed to play a key role in ECM deposition in HS. The protein expression of collagen I (Col I), collagen III (Col III), and α-smooth muscle actin (α-SMA) was obviously inhibited after treatment with miR-143-3p in HSFs. The CCK-8 assay showed that miR-143-3p transfection reduced the proliferation ability of HSFs, and flow cytometry showed that either early or late apoptosis of HSFs was upregulated by miR-143-3p. In addition, the activity of caspase 3 and caspase 9 was increased after miR-143-3p transfection. On the contrary, the miR-143-3p inhibitor was demonstrated to increase cell proliferation and inhibit apoptosis of HSFs. Moreover, miR-143-3p targeted the 3'-UTR of CTGF and caused a significant decrease of CTGF. Western blot demonstrated that Akt/mTOR phosphorylation and the expression of CTGF, Col I, Col III, and α-SMA were inhibited by miR-143-3p, but increased by CTGF overexpression. In conclusion, we found that miR-143-3p inhibits hypertrophic scarring by regulating the proliferation and apoptosis of human HSFs, inhibiting ECM production-associated protein expression by targeting CTGF, and restraining the Akt/mTOR pathway.


Subject(s)
Apoptosis , Cicatrix, Hypertrophic/metabolism , Connective Tissue Growth Factor/biosynthesis , MicroRNAs/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Cells, Cultured , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/pathology , Connective Tissue Growth Factor/genetics , Female , Humans , Male , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics
18.
J Cardiovasc Pharmacol ; 67(2): 110-20, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26371948

ABSTRACT

Connective tissue growth factor (CTGF) is a fibrogenic cytokine that promotes fibrosis in various organs. In the heart, both cardiomyocytes (CM) and cardiac fibroblasts have been reported as a source of CTGF expression, aiding cardiac fibrosis. Although the mammalian target of rapamycin (mTOR) forms 2 distinct complexes, mTORC1 and mTORC2, and plays a central role in integrating biochemical signals for protein synthesis and cellular homeostasis, we explored its role in CTGF expression in adult feline CM. CM were stimulated with 10 µM phenylephrine (PE), 200 nM angiotensin (Ang), or 100 nM insulin for 24 hours. PE and Ang, but not insulin, caused an increase in CTGF mRNA expression with the highest expression observed with PE. Inhibition of mTOR with torin1 but not rapamycin significantly enhanced PE-stimulated CTGF expression. Furthermore, silencing of raptor and rictor using shRNA adenoviral vectors to suppress mTORC1 and mTORC2, respectively, or blocking phosphatidylinositol 3-kinase (PI3K) signaling with LY294002 (LY) or Akt signaling by dominant-negative Akt expression caused a substantial increase in PE-stimulated CTGF expression as measured by both mRNA and secreted protein levels. However, studies with dominant-negative delta isoform of protein kinase C demonstrate that delta isoform of protein kinase C is required for both agonist-induced CTGF expression and mTORC2/Akt-mediated CTGF suppression. Finally, PE-stimulated CTGF expression was accompanied with a corresponding increase in Smad3 phosphorylation and pretreatment of cells with SIS3, a Smad3 specific inhibitor, partially blocked the PE-stimulated CTGF expression. Therefore, a PI3K/mTOR/Akt axis plays a suppressive role on agonist-stimulated CTGF expression where the loss of this mechanism could be a contributing factor for the onset of cardiac fibrosis in the hypertrophying myocardium.


Subject(s)
Connective Tissue Growth Factor/agonists , Connective Tissue Growth Factor/biosynthesis , Myocytes, Cardiac/metabolism , TOR Serine-Threonine Kinases/biosynthesis , Angiotensins/pharmacology , Animals , Cats , Cells, Cultured , Myocytes, Cardiac/drug effects , Phenylephrine/pharmacology
19.
J Cell Mol Med ; 19(3): 651-63, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25712415

ABSTRACT

The large tumour suppressor 1 (LATS1) signalling network has been proved to be an essential regulator within the cell, participating in multiple cellular phenotypes. However, it is unclear concerning the clinical significance of LATS1 and the regulatory mechanisms of 17-Allylamino-17- demethoxygeldanamycin (17-AAG) in lung adenocarcinoma (LAC). The aim of the present study was to investigate the correlation of LATS1 and yes-associated protein (YAP) expression with clinicopathological characteristics in LAC patients, and the effects of 17-AAG on biological behaviours of LAC cells. Subcutaneous LAC tumour models were further established to observe the tumour growth in nude mice. The results showed that the positive expression of LATS1 was significantly lowered (26.7% versus 68.0%, P < 0.001), while that of YAP was elevated (76.0% versus 56.0%, P = 0.03) in LAC tissues compared to the adjacent non-cancerous tissues; LAST1 expression was negatively correlated with YAP expression (r = 0.432, P < 0.001) and lymphatic invasion of the tumour (P = 0.015). In addition, 17-AAG inhibited proliferation and invasion, and induced cell apoptosis and cycle arrest in LAC cells together with increased expression of E-cadherin and p-LATS1, and decreased expression of YAP and connective tissue growth factor. Tumour volumes and weight were much smaller in 17-AAG-treated groups than those in untreated group (P < 0.01). Taken together, our findings indicate that decreased expression of LATS1 is associated with lymphatic invasion of LAC, and 17-AAG suppresses growth and invasion of LAC cells via regulation of the LATS1/YAP pathway in vitro and in vivo, suggesting that we may provide a promising therapeutic strategy for the treatment of human LAC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/drug therapy , Apoptosis/drug effects , Benzoquinones/pharmacology , Lactams, Macrocyclic/pharmacology , Lung Neoplasms/drug therapy , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Adenocarcinoma of Lung , Animals , Antineoplastic Agents/pharmacology , Cadherins/biosynthesis , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Connective Tissue Growth Factor/biosynthesis , Female , Gene Expression Regulation, Neoplastic , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness/pathology , Phosphoproteins/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Signal Transduction/drug effects , Transcription Factors , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
20.
J Cell Physiol ; 230(11): 2634-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25899830

ABSTRACT

Connective Tissue Growth Factor (CCN2/CTGF) and Nephroblastoma Overexpressed (CCN3/NOV) execute key functions within the hematopoietic compartment. Both are abundant in the bone marrow stroma, which is a niche for hematopoiesis and supports marrow function. Roles for 1,25-dihydroxyvitamin D3 (calcitriol) and all-trans retinoic acid in the bone marrow have also been elucidated. Interestingly, some of the annotated roles of these vitamins overlap with established functions of CCN2 and CCN3. Yet, no factor has been identified that unifies these observations. In this study, we report the regulation of the CTGF and NOV genes by Myeloid Zinc Finger-1 (MZF-1), a hematopoietic transcription factor. We show the interaction of MZF-1 with the CTGF and NOV promoters in several cell types. Up-regulation of MZF-1 via calcitriol and vitamin A induces expression of CTGF and NOV, implicating a role for these vitamins in the functions of these two genes. Lastly, knockdown of MZF1 reduces levels of CTGF and NOV. Collectively, our results argue that MZF-1 regulates the CTGF and NOV genes in the hematopoietic compartment, and may be involved in their respective functions in the stroma.


Subject(s)
Connective Tissue Growth Factor/biosynthesis , Hematopoiesis/genetics , Kruppel-Like Transcription Factors/genetics , Nephroblastoma Overexpressed Protein/biosynthesis , Bone Marrow/metabolism , Calcitriol/metabolism , Connective Tissue Growth Factor/genetics , Gene Expression Regulation , Humans , Kruppel-Like Transcription Factors/metabolism , Nephroblastoma Overexpressed Protein/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tretinoin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL