Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Small GTPases ; 12(4): 273-281, 2021 07.
Article in English | MEDLINE | ID: mdl-32043900

ABSTRACT

Malignant melanoma is characterized by mutations in a number of driver genes, most notably BRAF and NRAS. Recent genomic analyses revealed that 4-9% of sun-exposed melanomas bear activating mutations in RAC1, which encodes a small GTPase that is known to play key roles in cell proliferation, survival, and migration. The RAC1 protein activates several effector pathways, including Group A p21-activated kinases (PAKs), phosphoinositol-3-kinases (PI3Ks), in particular the beta isoform, and the serum-response factor/myocardin-related transcription factor (SRF/MRTF). Having previously shown that inhibition of Group A PAKs impedes oncogenic signalling from RAC1P29S, we here extend this analysis to examine the roles of PI3Ks and SRF/MRTF in melanocytes and/or in a zebrafish model. We demonstrate that a selective Group A PAK inhibitor (Frax-1036), a pan-PI3K (BKM120), and two PI3Kß inhibitors (TGX221, GSK2636771) impede the growth of melanoma cells driven by mutant RAC1 but not by mutant BRAF, while other PI3K selective inhibitors, including PI3Kα, δ and γ, are less effective. Using these compounds as well as an SRF/MRTF inhibitor (CCG-203,971), we observed similar results in vivo, using embryonic zebrafish development as a readout. These results suggest that targeting Group A PAKs, PI3Kß, and/or SRF/MRTF represent a promising approach to suppress RAC1 signalling in malignant melanoma.


Subject(s)
Embryo, Nonmammalian/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/drug therapy , Mutation , rac1 GTP-Binding Protein/genetics , Animals , Apoptosis , Cell Proliferation , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Phosphatidylinositol 3-Kinases/chemistry , Serum Response Factor/antagonists & inhibitors , Signal Transduction , Trans-Activators/antagonists & inhibitors , Tumor Cells, Cultured , Zebrafish , p21-Activated Kinases/antagonists & inhibitors
2.
Trends Cancer ; 6(6): 478-488, 2020 06.
Article in English | MEDLINE | ID: mdl-32460002

ABSTRACT

Small GTPases of the RAS and RHO families are related signaling proteins that, when activated by growth factors or by mutation, drive oncogenic processes. While activating mutations in KRAS, NRAS, and HRAS genes have long been recognized and occur in many types of cancer, similar mutations in RHO family genes, such as RAC1 and RHOA, have only recently been detected as the result of extensive cancer genome-sequencing efforts and are linked to a restricted set of malignancies. In this review, we focus on the role of RAC1 signaling in malignant melanoma, emphasizing recent advances that describe how this oncoprotein alters melanocyte proliferation and motility and how these findings might lead to new therapeutics in RAC1-mutant tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Skin Neoplasms/drug therapy , rac1 GTP-Binding Protein/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Gain of Function Mutation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/genetics , Melanoma/pathology , Phosphatidylinositol 3-Kinase , Protein Kinase Inhibitors/therapeutic use , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
3.
Cells ; 9(2)2020 02 15.
Article in English | MEDLINE | ID: mdl-32075310

ABSTRACT

Although thousands of long noncoding RNAs (lncRNAs) have been identified in porcine growth and development, the regulation mechanisms of functional lncRNAs have not been well explored. In this study, using 5'- and 3'-rapid amplification of cDNA ends (RACE) assays, we obtained two different variants of lncRNA maternally expressed gene 3 (MEG3), namely, MEG3 v1 and MEG3 v2, that were both highly expressed in porcine skeletal muscle and in the early stage of the differentiation of porcine satellite cells. Moreover, we identified the core transcript MEG3 v2. Functional analyses showed that MEG3 overexpression could effectively arrest myoblasts in the G1 phase, inhibit DNA replication, and promote myoblast differentiation, whereas MEG3 knockdown resulted in the opposite effects. Interestingly, the expression of serum response factor (SRF), a crucial transcription factor for myogenesis process, remarkably increased and decreased in mRNA and protein levels with the respective overexpression and knockdown of MEG3. Dual luciferase reporter assay showed that MEG3 could attenuate the decrease of luciferase activity of SRF induced by miR-423-5p in a dose-dependent manner. MEG3 overexpression could relieve the inhibitory effect on SRF and myoblast differentiation induced by miR-423-5p. In addition, results of RNA immunoprecipitation analysis suggested that MEG3 could act as a ceRNA for miR-423-5p. Our findings initially established a novel connection among MEG3, miR-423-5p, and SRF in porcine satellite cell differentiation. This novel role of MEG3 may shed new light on understanding of molecular regulation of lncRNA in porcine myogenesis.


Subject(s)
MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Serum Response Factor/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Male , MicroRNAs/genetics , Muscle Development , RNA, Long Noncoding/genetics , Satellite Cells, Skeletal Muscle/cytology , Serum Response Factor/antagonists & inhibitors , Swine , Transfection
4.
Cell Death Dis ; 10(6): 453, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186405

ABSTRACT

Hepatocellular carcinoma (HCC) has a high mortality rate due to the lack of effective treatments and drugs. Arsenic trioxide (ATO), which has been proved to successfully treat acute promyelocytic leukemia (APL), was recently reported to show therapeutic potential in solid tumors including HCC. However, its anticancer mechanisms in HCC still need further investigation. In this study, we demonstrated that ATO inhibits tumorigenesis and distant metastasis in mouse models, corresponding with a prolonged mice survival time. Also, ATO was found to significantly decrease the cancer stem cell (CSC)-associated traits. Minichromosome maintenance protein (MCM) 7 was further identified to be a potential target suppressed dramatically by ATO, of which protein expression is increased in patients and significantly correlated with tumor size, cellular differentiation, portal venous emboli, and poor patient survival. Moreover, MCM7 knockdown recapitulates the effects of ATO on CSCs and metastasis, while ectopic expression of MCM7 abolishes them. Mechanistically, our results suggested that ATO suppresses MCM7 transcription by targeting serum response factor (SRF)/MCM7 complex, which functions as an important transcriptional regulator modulating MCM7 expression. Taken together, our findings highlight the importance of ATO in the treatment of solid tumors. The identification of SRF/MCM7 complex as a target of ATO provides new insights into ATO's mechanism, which may benefit the appropriate use of this agent in the treatment of HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenic Trioxide/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Minichromosome Maintenance Complex Component 7/metabolism , Neoplastic Stem Cells/metabolism , Serum Response Factor/metabolism , Animals , Antineoplastic Agents/therapeutic use , Arsenic Trioxide/therapeutic use , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Disease Progression , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Ontology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/mortality , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Minichromosome Maintenance Complex Component 7/genetics , Neoplastic Stem Cells/drug effects , Prognosis , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/genetics , Transplantation, Heterologous
5.
J Med Chem ; 62(9): 4350-4369, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30951312

ABSTRACT

Through a phenotypic high-throughput screen using a serum response element luciferase promoter, we identified a novel 5-aryl-1,3,4-oxadiazol-2-ylthiopropionic acid lead inhibitor of Rho/myocardin-related transcription factor (MRTF)/serum response factor (SRF)-mediated gene transcription with good potency (IC50 = 180 nM). We were able to rapidly improve the cellular potency by 5 orders of magnitude guided by sharply defined and synergistic SAR. The remarkable potency and depth of the SAR, as well as the relatively low molecular weight of the series, suggests, but does not prove, that binding to the unknown molecular target may be occurring through a covalent mechanism. The series nevertheless has no observable cytotoxicity up to 100 µM. Ensuing pharmacokinetic optimization resulted in the development of two potent and orally bioavailable anti-fibrotic agents that were capable of dose-dependently reducing connective tissue growth factor gene expression in vitro as well as significantly reducing the development of bleomycin-induced dermal fibrosis in mice in vivo.


Subject(s)
Carboxylic Acids/therapeutic use , Enzyme Inhibitors/therapeutic use , Fibrosis/drug therapy , Oxadiazoles/therapeutic use , Serum Response Factor/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors , Animals , Carboxylic Acids/chemical synthesis , Carboxylic Acids/pharmacokinetics , Connective Tissue Growth Factor/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Female , Fibrosis/pathology , Mice, Inbred C57BL , Microsomes, Liver/drug effects , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacokinetics , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/pathology , Signal Transduction/drug effects , Skin/pathology , Structure-Activity Relationship , Transcription, Genetic/drug effects , rho GTP-Binding Proteins/antagonists & inhibitors
6.
JCI Insight ; 4(6)2019 03 21.
Article in English | MEDLINE | ID: mdl-30762586

ABSTRACT

About one-third of dilated cardiomyopathy (DCM) cases are caused by mutations in sarcomere or cytoskeletal proteins. However, treating the cytoskeleton directly is not possible because drugs that bind to actin are not well tolerated. Mutations in the actin binding protein CAP2 can cause DCM and KO mice, either whole body (CAP2-KO) or cardiomyocyte-specific KOs (CAP2-CKO) develop DCM with cardiac conduction disease. RNA sequencing analysis of CAP2-KO hearts and isolated cardiomyocytes revealed overactivation of fetal genes, including serum response factor-regulated (SRF-regulated) genes such as Myl9 and Acta2 prior to the emergence of cardiac disease. To test if we could treat CAP2-KO mice, we synthesized and tested the SRF inhibitor CCG-1423-8u. CCG-1423-8u reduced expression of the SRF targets Myl9 and Acta2, as well as the biomarker of heart failure, Nppa. The median survival of CAP2-CKO mice was 98 days, while CCG-1423-8u-treated CKO mice survived for 116 days and also maintained normal cardiac function longer. These results suggest that some forms of sudden cardiac death and cardiac conduction disease are under cytoskeletal stress and that inhibiting signaling through SRF may benefit DCM by reducing cytoskeletal stress.


Subject(s)
Anilides/administration & dosage , Benzamides/administration & dosage , Cardiomyopathy, Dilated/drug therapy , Carrier Proteins/genetics , Gene Expression Regulation, Developmental/drug effects , Serum Response Factor/antagonists & inhibitors , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/mortality , Cardiomyopathy, Dilated/pathology , Carrier Proteins/metabolism , Cytoskeleton/drug effects , Cytoskeleton/pathology , Disease Models, Animal , Female , Fetus , Heart/drug effects , Heart/embryology , Humans , Longevity/drug effects , Male , Mice , Myocardium/cytology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA-Seq , Serum Response Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , Transcription Factors/metabolism
7.
Cell Physiol Biochem ; 51(2): 763-777, 2018.
Article in English | MEDLINE | ID: mdl-30463073

ABSTRACT

BACKGROUND/AIMS: Vascular muscularity is a key event in vessel remodeling during pulmonary artery hypertension (PAH). Endothelial-mesenchymal transdifferentiation (EndMT) has been increasingly reported to play a role in disease occurrence. Galectin-3, a carbohydrate-binding protein regulates cell proliferation, differentiation, migration and neovascularization. However, whether galectin-3 controls endothelial cell transdifferentiation during the development of PAH is unknown. METHODS: Rats were exposed to normoxic or hypoxic conditions (fraction of inspired O2 0.10) for 21 d to establish PAH models. Hemodynamic changes were evaluated through surgery of the right jugular vein and ultrasound biomicroscopy inviVue. And vessel pathological alterations were detected by H&E staining. Galectin-3 (Gal-3)-induced pulmonary artery endothelium cell (PAEC) dynamic alterations were measured by MTT assays, Cell immunofluorescence, Flow cytometry, Real-time PCR and Western blot. RESULTS: Our study demonstrated that Gal-3 was expressed in hypoxic pulmonary vascular adventitia and intima. The increased Gal-3 expression was responsible for hypoxic vessel remodeling and PAH development in vivo. Gal-3 was found to inhibit cell proliferation and apoptosis in cultured endothelial cells. Meanwhile endothelial cell morphology was altered and exhibited smooth muscle-like cell features as demonstrated by the expression of α-SMA after Gal-3 treatment. Gal-3 activated Jagged1/Notch1 pathways and induced MyoD and SRF. When MyoD or SRF were silenced with siRNAs, Gal-3-initiated transdifferentiation in endothelial cells was blocked as indicated by a lack of α-SMA. CONCLUSION: These results suggest that Gal-3 induces PAECs to acquire an α-SMA phenotype via a transdifferentiation process which depends on the activation of Jagged1/Notch1 pathways that mediate MyoD and SRF expression.


Subject(s)
Cell Transdifferentiation , Galectin 3/metabolism , Vascular Remodeling , Animals , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , Cell Transdifferentiation/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Galectin 3/antagonists & inhibitors , Galectin 3/genetics , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Lung/metabolism , Male , MyoD Protein/antagonists & inhibitors , MyoD Protein/genetics , MyoD Protein/metabolism , Pulmonary Artery/cytology , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Receptor, Notch1/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/genetics , Serum Response Factor/metabolism , Vascular Remodeling/drug effects
8.
J Nanobiotechnology ; 16(1): 97, 2018 Nov 27.
Article in English | MEDLINE | ID: mdl-30482196

ABSTRACT

BACKGROUND: Sustained drug delivery is a large unmet clinical need in glaucoma. Here, we incorporated a Myocardin-Related Transcription Factor/Serum Response Factor inhibitor, CCG-222740, into slow release large unilamellar vesicles derived from the liposomes DOTMA (1,2-di-O-octadecenyl-3-trimethylammonium propane) and DOPC (1,2-dioleoyl-sn-glycero-3-phosphocholine), and tested their effects in vitro and in vivo. RESULTS: The vesicles were spherical particles of around 130 nm and were strongly cationic. A large amount of inhibitor could be incorporated into the vesicles. We showed that the nanocarrier CCG-222740 formulation gradually released the inhibitor over 14 days using high performance liquid chromatography. Nanocarrier CCG-222740 significantly decreased ACTA2 gene expression and was not cytotoxic in human conjunctival fibroblasts. In vivo, nanocarrier CCG-222740 doubled the bleb survival from 11.0 ± 0.6 days to 22.0 ± 1.3 days (p = 0.001), decreased conjunctival scarring and did not have any local or systemic adverse effects in a rabbit model of glaucoma filtration surgery. CONCLUSIONS: Our study demonstrates proof-of-concept that a nanocarrier-based formulation efficiently achieves a sustained release of a Myocardin-Related Transcription Factor/Serum Response Factor inhibitor and prevents conjunctival fibrosis in an established rabbit model of glaucoma filtration surgery.


Subject(s)
Delayed-Action Preparations/chemistry , Drug Delivery Systems , Serum Response Factor/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Conjunctival Diseases/drug therapy , Female , Fibroblasts/drug effects , Fibrosis/drug therapy , Humans , Liposomes/chemistry , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Rabbits , Tissue Distribution , Trans-Activators/antagonists & inhibitors , Trans-Activators/chemistry
9.
Prostate ; 78(5): 343-352, 2018 04.
Article in English | MEDLINE | ID: mdl-29341215

ABSTRACT

BACKGROUND: The ERK signaling pathway is frequently deregulated in tumorigenesis, mostly by classical mechanisms such as gene mutation of its components (eg, RAS and RAF). However, whether and how multiple key components of ERK pathway are regulated by microRNAs are not clear. METHODS: We firstly predicted post-transcriptional regulation of multiple key components of the ERK signaling pathway by miR181c through bioinformatics analysis, and then confirmed the post-transcriptional regulation by dual luciferase reporter gene assays and Western blot analysis. The biological effects of miR181c on prostate cancer cell proliferation, apoptosis, migration, and invasion were measured by CCK-8 assay, flow cytometry, wound scratch assay, transwell cell migration, and invasion assays. RESULTS: miR181c post-transcriptionally regulated multiple key members of the ERK signaling pathway, including extracellular signal-regulated kinase 2 (ERK2), ribosomal S6 kinase 2 (RSK2), serum response factor (SRF), and FBJ murine osteosarcoma viral oncogene homolog (c-Fos). Ectopic expression of miR181c mimics effectively suppressed prostate cancer cell proliferation, migration, and invasion, but promoted cell apoptosis. Furthermore, miR181c treatment combined with the multi-kinase inhibitor sorafenib significantly enhanced these anti-tumor effects. CONCLUSIONS: Downregulation of miR181c results in deregulated ERK signaling and promotes prostate cancer cell growth and metastasis.


Subject(s)
MAP Kinase Signaling System , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Down-Regulation , Humans , Male , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Neoplasm Invasiveness , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/metabolism , Sorafenib/pharmacology
10.
Cell Physiol Biochem ; 44(2): 701-715, 2017.
Article in English | MEDLINE | ID: mdl-29169155

ABSTRACT

BACKGROUND/AIMS: Our previous studies demonstrated that intrinsic aortic smooth muscle cell (VSMC) stiffening plays a pivotal role in aortic stiffening in aging and hypertension. However, the underlying molecular mechanisms remain largely unknown. We here hypothesized that Rho kinase (ROCK) acts as a novel mediator that regulates intrinsic VSMC mechanical properties through the serum response factor (SRF) /myocardin pathway and consequently regulates aortic stiffness and blood pressure in hypertension. METHODS: Four-month old male spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats were studied. Aortic stiffness was measured by echography. Intrinsic mechanical properties of VSMCs were measured by atomic force microscopy (AFM) in vitro. RESULTS: Compared to WKY rats, SHR showed a significant increase in aortic stiffness and blood pressure, which is accompanied by a remarkable cell stiffening and ROCK activation in thoracic aortic (TA) VSMCs. Theses alterations in SHR were abolished by Y-27632, a specific inhibitor of ROCK. Additionally, boosted filamentous/globular actin ratio was detected in TA VSMCs from SHR versus WKY rats, resulting in an up-regulation of SRF and myocardin expression and its downstream stiffness-associated genes including α-smooth muscle actin, SM22, smoothelin and myosin heavy chain 11. Reciprocally, these alterations in SHR TA VSMCs were also suppressed by Y-27632. Furthermore, a specific inhibitor of SRF/myocardin, CCG-100602, showed a similar effect to Y-27632 in SHR in both TA VSMCs stiffness in vitro and aorta wall stiffness in vivo. CONCLUSION: ROCK is a novel mediator modulating aortic VSMC stiffness through SRF/myocardin signaling which offers a therapeutic target to reduce aortic stiffening in hypertension.


Subject(s)
Actins/metabolism , Hypertension/physiopathology , Muscle, Smooth, Vascular/physiology , Nuclear Proteins/metabolism , Serum Response Factor/metabolism , Trans-Activators/metabolism , rho-Associated Kinases/metabolism , Amides/pharmacology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/physiology , Blood Pressure , Cytoskeletal Proteins/metabolism , Echocardiography , Hypertension/veterinary , Male , Microscopy, Atomic Force , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myosin Heavy Chains/metabolism , Nipecotic Acids/pharmacology , Nuclear Proteins/antagonists & inhibitors , Pyridines/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Serum Response Factor/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors , Ultrasonography , Up-Regulation , Vascular Stiffness/drug effects , rho-Associated Kinases/antagonists & inhibitors
11.
J Biol Chem ; 292(24): 10180-10196, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28465353

ABSTRACT

We have previously shown that dysbindin is a potent inducer of cardiomyocyte hypertrophy via activation of Rho-dependent serum-response factor (SRF) signaling. We have now performed a yeast two-hybrid screen using dysbindin as bait against a cardiac cDNA library to identify the cardiac dysbindin interactome. Among several putative binding proteins, we identified tripartite motif-containing protein 24 (TRIM24) and confirmed this interaction by co-immunoprecipitation and co-immunostaining. Another tripartite motif (TRIM) family protein, TRIM32, has been reported earlier as an E3 ubiquitin ligase for dysbindin in skeletal muscle. Consistently, we found that TRIM32 also degraded dysbindin in neonatal rat ventricular cardiomyocytes as well. Surprisingly, however, TRIM24 did not promote dysbindin decay but rather protected dysbindin against degradation by TRIM32. Correspondingly, TRIM32 attenuated the activation of SRF signaling and hypertrophy due to dysbindin, whereas TRIM24 promoted these effects in neonatal rat ventricular cardiomyocytes. This study also implies that TRIM32 is a key regulator of cell viability and apoptosis in cardiomyocytes via simultaneous activation of p53 and caspase-3/-7 and inhibition of X-linked inhibitor of apoptosis. In conclusion, we provide here a novel mechanism of post-translational regulation of dysbindin and hypertrophy via TRIM24 and TRIM32 and show the importance of TRIM32 in cardiomyocyte apoptosis in vitro.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Carrier Proteins/metabolism , Dystrophin-Associated Proteins/metabolism , Myocytes, Cardiac/metabolism , Serum Response Factor/metabolism , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Newborn , Apoptosis , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Hypertrophic/pathology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cells, Cultured , Dysbindin , Dystrophin-Associated Proteins/chemistry , Dystrophin-Associated Proteins/genetics , HEK293 Cells , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Stability , Proteolysis , RNA Interference , Rats , Rats, Wistar , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serum Response Factor/agonists , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
12.
J Biol Chem ; 292(28): 11777-11791, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28546428

ABSTRACT

Megakaryoblastic leukemia (MKL)/serum-response factor (SRF)-mediated gene transcription is a highly conserved mechanism that connects dynamic reorganization of the actin cytoskeleton to regulation of expression of a wide range of genes, including SRF itself and many important structural and regulatory components of the actin cytoskeleton. In this study, we examined the possible role of MKL/SRF in the context of regulation of profilin (Pfn), a major controller of actin dynamics and actin cytoskeletal remodeling in cells. We demonstrated that despite being located on different genomic loci, two major isoforms of Pfn (Pfn1 and Pfn2) are co-regulated by a common mechanism involving the action of MKL that is independent of its SRF-related activity. We found that MKL co-regulates the expression of Pfn isoforms indirectly by modulating signal transducer and activator of transcription 1 (STAT1) and utilizing its SAP-domain function. Unexpectedly, our studies revealed that cellular externalization, rather than transcription of Pfn1, is affected by the perturbations of MKL. We further demonstrated that MKL can influence cell migration by modulating Pfn1 expression, indicating a functional connection between MKL and Pfn1 in actin-dependent cellular processes. Finally, we provide initial evidence supporting the ability of Pfn to influence MKL and SRF expression. Collectively, these findings suggest that Pfn may play a role in a possible feedback loop of the actin/MKL/SRF signaling circuit.


Subject(s)
Gene Expression Regulation , Profilins/metabolism , STAT1 Transcription Factor/metabolism , Trans-Activators/metabolism , Cell Line, Tumor , Cell Movement , HEK293 Cells , Humans , Oligopeptides/genetics , Oligopeptides/metabolism , Profilins/genetics , Protein Interaction Domains and Motifs , Protein Transport , RNA Interference , RNA, Messenger/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/genetics , Serum Response Factor/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics
13.
Sci Rep ; 7(1): 518, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28364121

ABSTRACT

The myocardin-related transcription factor/serum response factor (MRTF/SRF) pathway represents a promising therapeutic target to prevent fibrosis. We have tested the effects of new pharmacological inhibitors of MRTF/SRF signalling in a preclinical model of fibrosis. CCG-222740, a novel MRTF/SRF inhibitor, markedly decreased SRF reporter gene activity and showed a greater inhibitory effect on MRTF/SRF target genes than the previously described MRTF-A inhibitor CCG-203971. CCG-222740 was also five times more potent, with an IC50 of 5 µM, in a fibroblast-mediated collagen contraction assay, was less cytotoxic, and a more potent inhibitor of alpha-smooth muscle actin protein expression than CCG-203971. Local delivery of CCG-222740 and CCG-203971 in a validated and clinically relevant rabbit model of scar tissue formation after glaucoma filtration surgery increased the long-term success of the surgery by 67% (P < 0.0005) and 33% (P < 0.01), respectively, and significantly decreased fibrosis and scarring histologically. Unlike mitomycin-C, neither CCG-222740 nor CCG-203971 caused any detectable epithelial toxicity or systemic side effects with very low drug levels measured in the aqueous, vitreous, and serum. We conclude that inhibitors of MRTF/SRF-regulated gene transcription such as CCG-222740, potentially represent a new therapeutic strategy to prevent scar tissue formation in the eye and other tissues.


Subject(s)
Cicatrix/metabolism , Cicatrix/pathology , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Animals , Cells, Cultured , Cicatrix/prevention & control , Collagen/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Extracellular Matrix , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Humans , Rabbits , Signal Transduction/drug effects
14.
J Mol Cell Cardiol ; 74: 127-38, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24839911

ABSTRACT

Valvular interstitial cells (VICs), the fibroblast-like cellular constituents of aortic heart valves, maintain structural integrity of valve tissue. Activation into contractile myofibroblasts occurs under pathological situations and under standard cell culture conditions of isolated VICs. Reversal of this phenotype switch would be of major importance in respect to fibrotic valve diseases. In this investigation, we found that exogenous polyunsaturated fatty acids (PUFAs) decreased contractility and expression of myofibroblastic markers like α-smooth muscle actin (αSMA) in cultured VICs from porcine aortic valves. The most active PUFAs, docosahexaenoic acid (DHA) and arachidonic acid (AA) reduced the level of active RhoA and increased the G/F-actin ratio. The G-actin-regulated nuclear translocation of myocardin-related transcription factors (MRTFs), co-activators of serum response factor, was also reduced by DHA and AA. The same effects were observed after blocking RhoA directly with C3 transferase. In addition, increased contractility after induction of actin polymerisation with jasplakinolide and concomitant expression of αSMA were ameliorated by active PUFAs. Furthermore, reduced αSMA expression under PUFA exposure was observed in valve tissue explants demonstrating physiological relevance. In conclusion, RhoA/G-actin/MRTF signalling is operative in VICs, and this pathway can be partially blocked by certain PUFAs whereby the activation into the myofibroblastic phenotype is reversed.


Subject(s)
Aortic Valve/drug effects , Arachidonic Acid/pharmacology , Docosahexaenoic Acids/pharmacology , Myofibroblasts/drug effects , ADP Ribose Transferases/pharmacology , Actins/genetics , Actins/metabolism , Animals , Aortic Valve/cytology , Aortic Valve/metabolism , Botulinum Toxins/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Depsipeptides/pharmacology , Gene Expression Regulation , Myofibroblasts/cytology , Myofibroblasts/metabolism , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/genetics , Serum Response Factor/metabolism , Signal Transduction , Swine , Tissue Culture Techniques , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
15.
Inflamm Bowel Dis ; 20(1): 154-65, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24280883

ABSTRACT

BACKGROUND: Ras homolog gene family, member A (RhoA)/Rho-associated coiled-coil forming protein kinase signaling is a key pathway in multiple types of solid organ fibrosis, including intestinal fibrosis. However, the pleiotropic effects of RhoA/Rho-associated coiled-coil forming protein kinase signaling have frustrated targeted drug discovery efforts. Recent recognition of the role of Rho-regulated gene transcription by serum response factor (SRF) and its transcriptional cofactor myocardin-related transcription factor A (MRTF-A) suggest a novel locus for pharmacological intervention. METHODS: Because RhoA signaling is mediated by both physical and biochemical stimuli, we examined whether pharmacological inhibition of RhoA or the downstream transcription pathway of MRTF-A/SRF could block intestinal fibrogenesis in 2 in vitro models. RESULTS: In this study, we demonstrate that inhibition of RhoA signaling blocks both matrix-stiffness and transforming growth factor beta-induced fibrogenesis in human colonic myofibroblasts. Repression of alpha-smooth muscle actin and collagen expression was associated with the inhibition of MRTF-A nuclear localization. CCG-1423, a first-generation Rho/MRTF/SRF pathway inhibitor, repressed fibrogenesis in both models, yet has unacceptable cytotoxicity. Novel second-generation inhibitors (CCG-100602 and CCG-203971) repressed both matrix-stiffness and transforming growth factor beta-mediated fibrogenesis as determined by protein and gene expression in a dose-dependent manner. CONCLUSIONS: Targeting the Rho/MRTF/SRF mechanism with second-generation Rho/MRTF/SRF inhibitors may represent a novel approach to antifibrotic therapeutics.


Subject(s)
Anilides/pharmacology , Benzamides/pharmacology , Colon/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Fibrosis/prevention & control , Myofibroblasts/drug effects , Oncogene Proteins, Fusion/antagonists & inhibitors , Serum Response Factor/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors , Blotting, Western , Cell Adhesion/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Colon/metabolism , Colon/pathology , DNA-Binding Proteins/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Humans , Myofibroblasts/metabolism , Myofibroblasts/pathology , Oncogene Proteins, Fusion/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Serum Response Factor/metabolism , Signal Transduction/drug effects , Trans-Activators , Transforming Growth Factor beta/genetics , rho GTP-Binding Proteins/metabolism
16.
J Basic Clin Physiol Pharmacol ; 24(4): 287-97, 2013.
Article in English | MEDLINE | ID: mdl-23893683

ABSTRACT

BACKGROUND: Modulation of inflammatory signaling has been elucidated in several disease models. Acrolein, an environmental pollutant, has been linked to diseases such as atherosclerosis and to the inflammatory process involving nuclear factor κB (NFκB). Serum response factor (SRF), a transcription factor, regulates cell development, differentiation and proliferation through signaling molecules such as extracellular signal-regulated kinase 1/2 (ERK1/2) and CD36. We hypothesized that acrolein toxicity involves SRF in the process of activating NFκB and may involve CD36/ERK1/2. METHODS: Vascular smooth muscle cells (VSMCs) were exposed to acrolein (0.5 µg/mL) in the presence or absence of 10 nM QNZ (NFκB inhibitor), 300 nM CCG1423 (SRF inhibitor) and 50 µM PD98059 (ERK1/2 inhibitor). Protein and RNA were isolated. Changes in expression were determined by Western blot and polymerase chain reaction (PCR) array. RESULTS: Subtoxic doses of acrolein increased ERK1/2, SRF and NFκB protein expression, whereas CD36 expression was unchanged. Increase in NFκB expression was accompanied by an increase in activity. ERK1/2 inhibition only blunted SRF expression. SRF inhibition blunted NFκB expression but not that of ERK1/2. CD36 expression was unchanged in the presence of either inhibitor. PCR array analysis indicated up-regulation of nine genes (>4- to 50-fold) and down-regulation of six genes (>4- to 12-fold) involved in inflammatory signaling. CONCLUSIONS: We propose that SRF is required in acrolein activation of NFκB and is ERK1/2 dependent.


Subject(s)
Acrolein/toxicity , Environmental Pollutants/toxicity , Inflammation/chemically induced , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Serum Response Factor/immunology , Animals , Cell Survival/drug effects , Cells, Cultured , Inflammation/genetics , Inflammation/immunology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Muscle, Smooth, Vascular/immunology , Myocytes, Smooth Muscle/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Rats , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/genetics
17.
Bioorg Med Chem Lett ; 23(13): 3826-32, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23707258

ABSTRACT

CCG-1423 (1) is a novel inhibitor of Rho/MKL1/SRF-mediated gene transcription that inhibits invasion of PC-3 prostate cancer cells in a Matrigel model of metastasis. We recently reported the design and synthesis of conformationally restricted analogs (e.g., 2) with improved selectivity for inhibiting invasion versus acute cytotoxicity. In this study we conducted a survey of aromatic substitution with the goal of improving physicochemical parameters (e.g., ClogP, MW) for future efficacy studies in vivo. Two new compounds were identified that attenuated cytotoxicity even further, and were fourfold more potent than 2 at inhibiting PC-3 cell migration in a scratch wound assay. One of these (8a, CCG-203971, IC50=4.2 µM) was well tolerated in mice for 5 days at 100mg/kg/day i.p., and was able to achieve plasma levels exceeding the migration IC50 for up to 3 h.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Nipecotic Acids/pharmacology , Oncogene Proteins, Fusion/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Serum Response Factor/antagonists & inhibitors , rhoA GTP-Binding Protein/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Anilides/chemical synthesis , Anilides/chemistry , Anilides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzamides/chemical synthesis , Benzamides/chemistry , Benzamides/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Molecular Structure , Neoplasm Metastasis/drug therapy , Nipecotic Acids/chemical synthesis , Nipecotic Acids/chemistry , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Serum Response Factor/genetics , Serum Response Factor/metabolism , Structure-Activity Relationship , Trans-Activators , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
18.
Nucleic Acid Ther ; 23(1): 62-70, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23308381

ABSTRACT

To treat urethral strictures of the lower urinary tract, urethrotomy is the method of choice. But this minimally invasive method suffers from poor outcome rates and leads often to restenosis of the urinary tract because of hyper-proliferating fibroblasts. Our aim is to minimize the proliferation of excessive tissue due to a new minimal invasive therapeutic approach. As an appropriate model, we isolated fibroblasts from different benign prostatic hyperplasia patients and transfected them with small interfering RNA (siRNA) against the transcription factor serum response factor (SRF), a key factor for cell cycle regulation and apoptosis. The resulting knockdown of SRF was examined on the messenger RNA level by quantitative real-time polymerase chain reaction and on the protein level by western blot. The correlation of SRF silencing and impact on cell proliferation was examined by xCELLigence, 5-bromo-2'-deoxiuridine proliferation assay, total cell counts, and senescence assay. The transfection of primary prostatic fibroblasts with SRFsiRNA revealed specific and significant knockdown of SRF, leading to significant inhibition of proliferation after the second transfection, which was revealed by proliferation assay and total cell number. The results of this study indicate a substantial role of SRF in prostatic fibroblasts and we suggest that SRF silencing might be used for the treatment of urethral strictures to achieve a durably patent urethra.


Subject(s)
Prostatic Hyperplasia/therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Serum Response Factor/antagonists & inhibitors , Cell Proliferation , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serum Response Factor/genetics , Transfection
19.
Cancer Biother Radiopharm ; 28(2): 146-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23134219

ABSTRACT

Serum response factor (SRF) is a transcription factor of the MADS box family. To date, DNA binding sites for SRF [serum response elements (SREs)] have been found in the promoters of approximately 50 different genes known to be involved in the regulation cell proliferation, differentiation, and apoptosis. Recent studies have indicated that SRF plays a role in the development of some tumors, including hepatocellular, thyroid, esophageal, and lung carcinomas. However, expression of SRF and its roles in gastric carcinoma are unclear. We found SRF to be highly expressed in human gastric carcinoma as well as ectopic or reduced expression for E-cadherin and ß-catenin. Blockage of SRF expression was found to inhibit proliferation, invasion, and migration. We also found that an inhibitor (Y-27632) of Rho-associated coiled kinase (ROCK1), a regulator of actin cytoskeleton that regulates cell adhesion, migration, and motility, suppressed SRF expression as well. These results demonstrate that SRF is involved in the aggressive behavior of gastric carcinoma cells. We also found that the inhibition of ROCK1 by Y-27632 can inhibit the invasion and migration of gastric cells done at least, in part, by attenuating SRF expression.


Subject(s)
Apoptosis , Cell Movement , Serum Response Factor/metabolism , Stomach Neoplasms/pathology , Blotting, Western , Cadherins/metabolism , Cell Adhesion , Cell Differentiation , Cell Proliferation , Female , Flow Cytometry , Gastric Mucosa/metabolism , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Retrospective Studies , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/genetics , Stomach/pathology , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Wound Healing , beta Catenin/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
20.
Nat Neurosci ; 13(9): 1082-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20694003

ABSTRACT

It has been suggested that gene expression and protein synthesis are required for both long-term memory consolidation and late phases of long-term potentiation and long-term depression (LTD). The necessary genes and the specific transcription factor binding sites in their promoters remain unknown. We found that inhibition of the transcription factor SRF or its cofactor MAL blocked the late phase of LTD in mouse cultured cerebellar Purkinje cells, as did deletion of the immediate early gene Arc. Using neuronal bacterial artificial chromosome (BAC) transfection, we found that, in Arc-/- cells transfected with a wild-type Arc BAC, late-phase LTD was rescued. However, mutation of one SRF-binding site in the Arc promoter (SRE 6.9) blocked this rescue. Co-transfection of wild-type Arc and SRF engineered to bind mutated SRE 6.9 restored late-phase LTD in Arc-/-, SRE 6.9 mutant BAC cells. Thus, SRF binding to SRE 6.9 in the Arc promoter is required for the late phase of cerebellar LTD.


Subject(s)
Cytoskeletal Proteins/metabolism , Long-Term Synaptic Depression/physiology , Nerve Tissue Proteins/metabolism , Purkinje Cells/physiology , Serum Response Factor/metabolism , Animals , Cells, Cultured , Cytoskeletal Proteins/genetics , Long-Term Synaptic Depression/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Serum Response Factor/antagonists & inhibitors , Time Factors , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...