Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Commun Biol ; 6(1): 214, 2023 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-36823181

RESUMO

Spermatogenesis is an extremely complex process, and any obstruction can cause male infertility. RhoGDIα has been identified as a risk of male sterility. In this study, we generate RhoGDIα knockout mice, and find that the males have severely low fertility. The testes from RhoGDIα-/- mice are smaller than that in WT mice. The numbers of spermatogonia and spermatocytes are decreased in RhoGDIα-/- testis. Spermatogenesis is compromised, and spermatocyte meiosis is arrested at zygotene stage in RhoGDIα-/- mice. Acrosome dysplasia is also observed in sperms of the mutant mice. At the molecular level, RhoGDIα deficiency activate the LIMK/cofilin signaling pathway, inhibiting F-actin depolymerization, impairing testis and inducing low fertility in mouse. In addition, the treatment of RhoGDIα-/- mice with Rac1 inhibitor NSC23766 alleviate testis injury and improve sperm quality by inhibiting the LIMK/cofilin/F-actin pathway during spermatogenesis. Together, these findings reveal a previously unrecognized RhoGDIα/Rac1/F-actin-dependent mechanism involved in spermatogenesis and male fertility.


Assuntos
Actinas , Infertilidade Masculina , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho , Animais , Masculino , Camundongos , Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Infertilidade Masculina/genética , Camundongos Knockout , Proteínas rac1 de Ligação ao GTP/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Sêmen/metabolismo , Transdução de Sinais/fisiologia , Espermatogênese
2.
J Cell Sci ; 135(20)2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36111497

RESUMO

Attainment of proper cell shape and the regulation of cell migration are essential processes in the development of an organism. The mixed lineage leukemia (MLL or KMT2A) protein, a histone 3 lysine 4 (H3K4) methyltransferase, plays a critical role in cell-fate decisions during skeletal development and haematopoiesis in higher vertebrates. Rho GTPases - RhoA, Rac1 and CDC42 - are small G proteins that regulate various key cellular processes, such as actin cytoskeleton formation, the maintenance of cell shape and cell migration. Here, we report that MLL regulates the homeostasis of these small Rho GTPases. Loss of MLL resulted in an abnormal cell shape and a disrupted actin cytoskeleton, which lead to diminished cell spreading and migration. MLL depletion affected the stability and activity of Rho GTPases in a SET domain-dependent manner, but these Rho GTPases were not direct transcriptional targets of MLL. Instead, MLL regulated the transcript levels of their chaperone protein RhoGDI1 (also known as ARHGDIA). Using MDA-MB-231, a triple-negative breast cancer cell line with high RhoGDI1 expression, we show that MLL depletion or inhibition by small molecules reduces tumour progression in nude mice. Our studies highlight the central regulatory role of MLL in Rho/Rac/CDC42 signalling pathways. This article has an associated First Person interview with the first author of the paper.


Assuntos
Proteínas rho de Ligação ao GTP , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho , Camundongos , Animais , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Camundongos Nus , Histonas/metabolismo , Lisina , Transdução de Sinais/fisiologia , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Movimento Celular/fisiologia , Citoesqueleto de Actina/metabolismo , Metiltransferases/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Actinas/metabolismo
3.
Hum Cell ; 35(5): 1440-1452, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35841528

RESUMO

Preeclampsia (PE) is a pregnancy-associated disease, which is the major cause of mortality on maternity and perinatal infants. It is hypothesized that PE is a consequence of the dysfunction of the trophoblast cells. Pleckstrin homology-like domain, family A, member 2 (PHLDA2) was shown to inhibit the proliferation, migration, and invasion of trophoblast cells in our previous studies. However, the mechanism by which PHLDA2 affects trophoblast cell function has not been clarified. In the current study, co-immunoprecipitation (Co-IP) with mass spectroscopy analysis was used to explore the proteins that interacted with PHLDA2. A total of 291 candidate proteins were found to be associated with PHLDA2. The interaction between PHLDA2 and Rho guanine nucleotide dissociation inhibitor (RhoGDI) 1 was identified by Co-IP and immunofluorescence staining. Western blot analysis indicated that overexpression of PHLDA2 resulted in upregulation of the RhoGDI1 protein levels, which were stabilized in the presence of cycloheximide. Similarly, overexpression of RhoGDI1 promoted PHLDA2 expression and its stability. Furthermore, pull-down and Co-IP results indicated that PHLDA2 repressed the activity of Rho guanosine triphosphate hydrolase family proteins by regulating RhoGDI1 expression. In addition, RhoGDI1 expression was upregulated in the placental tissues of patients with PE. The effects of the suppression of PHLDA2 expression on proliferation, migration, and invasion of trophoblast cells were partly abrogated following knockdown of RhoGDI1. Taken together, the data indicated that RhoGDI1 mediated regulation of PHLDA2 on the biological behavior of trophoblast cells and may participate in the pathophysiology of PE.


Assuntos
Pré-Eclâmpsia , Trofoblastos , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Humanos , Proteínas Nucleares , Placenta , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/patologia , Gravidez , Trofoblastos/patologia , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
5.
Neurosci Lett ; 772: 136469, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35066091

RESUMO

OBJECTIVES: To further investigate the effects of miR-497 on the biological behavior of human medulloblastoma cell line in vitro. METHODS: Human medulloblastoma cell lines, Daoy and D341, were used in this study, and the miR-497 expression in the cells was measured by Quantitative PCR with fluorescence. The Daoy cells were divided into the mimics group (Daoy cells treated with mimics), inhibitor group (Daoy cells treated with inhibitor), normal Daoy cells, ARHGDIA siRNA group (Daoy cells transfected with ARHGDIA siRNA), ARHGDIA control group (Daoy cells did not receive any treatment), and negative control group (normal cells transfected with ARHGDIA siRNA). The expression of miR-497 and ARHGDIA mRNA was measured by Quantitative PCR with fluorescence, while the level of ARHGDIA protein was measured by Western blot. The binding capability of ARHGDIA and miR-497 was assessed by luciferase assay, the migration of cells was assessed by wound healing assay, and the invasion of cells was assessed by Transwell assay. RESULTS: Compared to D341 cells, the miR-497 level was significantly higher in the Daoy cells (P < 0.01). The dual-luciferase reporter assay showed that miR-497 targets ARHGDIA. Transfecting the normal Daoy cells with miR-497 mimics significantly reduced the expression of ARHGDIA protein (P < 0.05), while transfecting normal Daoy cells with miR-497 inhibitor significantly increased the expression of ARHGDIA protein (P < 0.05). Consequently, the migration and invasion capability of cells increased significantly after transfection with miR-497 mimic (P < 0.05), and decreased significantly after transfection with miR-497 inhibitor (P < 0.05). In addition, the migration and invasion capabilities of the cells also increased significantly after transfection with ARHGDIA siRNA (P < 0.05). CONCLUSIONS: miR-497/ARHGDIA axis participates in the in vitro migration and invasion of human medulloblastoma cell lines.


Assuntos
Neoplasias Encefálicas/metabolismo , Meduloblastoma/metabolismo , MicroRNAs/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Movimento Celular , Humanos , Meduloblastoma/genética , MicroRNAs/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
6.
Cells ; 10(7)2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34359999

RESUMO

Much progress has been made toward deciphering RHO GTPase functions, and many studies have convincingly demonstrated that altered signal transduction through RHO GTPases is a recurring theme in the progression of human malignancies. It seems that 20 canonical RHO GTPases are likely regulated by three GDIs, 85 GEFs, and 66 GAPs, and eventually interact with >70 downstream effectors. A recurring theme is the challenge in understanding the molecular determinants of the specificity of these four classes of interacting proteins that, irrespective of their functions, bind to common sites on the surface of RHO GTPases. Identified and structurally verified hotspots as functional determinants specific to RHO GTPase regulation by GDIs, GEFs, and GAPs as well as signaling through effectors are presented, and challenges and future perspectives are discussed.


Assuntos
Proteínas Ativadoras de GTPase/genética , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Proteína cdc42 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Proteína rhoA de Ligação ao GTP/genética , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Transtornos Cognitivos/genética , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Doenças Transmissíveis/genética , Doenças Transmissíveis/metabolismo , Doenças Transmissíveis/patologia , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica , Humanos , Família Multigênica , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
7.
Mol Med Rep ; 24(1)2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34080651

RESUMO

Transforming growth factor­ß1 (TGF­ß1)­induced epithelial­mesenchymal transition (EMT) serves a significant role in pulmonary fibrosis (PF). Increasing evidence indicates that microRNAs (miRNAs or miRs) contribute to PF pathogenesis via EMT regulation. However, the role of miR­483­5p in PF remains unclear. Therefore, the present study investigated the potential effect of miR­483­5p on TGF­ß1­induced EMT in PF. It was found that the expression of miR­483­5p was upregulated in both PF tissue and A549 cells treated with TGF­ß1, whereas expression of Rho GDP dissociation inhibitor 1 (RhoGDI1) was downregulated. miR­483­5p mimic transfection promoted TGF­ß1­induced EMT; by contrast, miR­483­5p inhibitor inhibited TGF­ß1­induced EMT. Also, miR­483­5p mimic decreased RhoGDI1 expression, whereas miR­483­5p inhibitor increased RhoGDI1 expression. Furthermore, dual­luciferase reporter gene assay indicated that miR­483­5p directly regulated RhoGDI1. Moreover, RhoGDI1 knockdown eliminated the inhibitory effect of the miR­483­5p inhibitor on TGF­ß1­induced EMT via the Rac family small GTPase (Rac)1/PI3K/AKT pathway. In conclusion, these data indicated that miR­483­5p inhibition ameliorated TGF­ß1­induced EMT by targeting RhoGDI1 via the Rac1/PI3K/Akt signaling pathway in PF, suggesting a potential role of miR­483­5p in the prevention and treatment of PF.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , MicroRNAs/genética , MicroRNAs/metabolismo , Fibrose Pulmonar/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Células A549 , Proliferação de Células/efeitos dos fármacos , Transição Epitelial-Mesenquimal/fisiologia , Humanos , Fibrose Pulmonar/patologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
8.
Nat Genet ; 53(3): 342-353, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33558758

RESUMO

Technological and computational advances in genomics and interactomics have made it possible to identify how disease mutations perturb protein-protein interaction (PPI) networks within human cells. Here, we show that disease-associated germline variants are significantly enriched in sequences encoding PPI interfaces compared to variants identified in healthy participants from the projects 1000 Genomes and ExAC. Somatic missense mutations are also significantly enriched in PPI interfaces compared to noninterfaces in 10,861 tumor exomes. We computationally identified 470 putative oncoPPIs in a pan-cancer analysis and demonstrate that oncoPPIs are highly correlated with patient survival and drug resistance/sensitivity. We experimentally validate the network effects of 13 oncoPPIs using a systematic binary interaction assay, and also demonstrate the functional consequences of two of these on tumor cell growth. In summary, this human interactome network framework provides a powerful tool for prioritization of alleles with PPI-perturbing mutations to inform pathobiological mechanism- and genotype-based therapeutic discovery.


Assuntos
Biologia Computacional/métodos , Mutação , Neoplasias/genética , Mapas de Interação de Proteínas/genética , Araquidonato 5-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/metabolismo , Arginina/genética , Arginina/metabolismo , Doença/genética , Genoma Humano , Histonas/genética , Histonas/metabolismo , Humanos , Testes Farmacogenômicos , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Reprodutibilidade dos Testes , Serina/genética , Serina/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
9.
J Neurochem ; 157(3): 494-507, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33320336

RESUMO

Phospholipid Phosphatase-Related Protein Type 1 (PLPPR1) is a member of a family of lipid phosphatase related proteins, integral membrane proteins characterized by six transmembrane domains. This family of proteins is enriched in the brain and recent data indicate potential pleiotropic functions in several different contexts. An inherent ability of this family of proteins is to induce morphological changes, and we have previously reported that members of this family interact with each other and may function co-operatively. However, the function of PLPPR1 is not yet understood. Here we show that the expression of PLPPR1 reduces the inhibition of neurite outgrowth of cultured mouse hippocampal neurons by chondroitin sulfate proteoglycans and the retraction of neurites of Neuro-2a cells by lysophosphatidic acid (LPA). Further, we show that PLPPR1 reduces the activation of Ras homolog family member A (RhoA) by LPA in Neuro-2a cells, and that this is because of an association of PLPPR1with the Rho-specific guanine nucleotide dissociation inhibitor (RhoGDI1). These results establish a novel signaling pathway for the PLPPR1 protein.


Assuntos
Axônios/fisiologia , Proteínas de Membrana/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Proteoglicanas de Sulfatos de Condroitina/farmacologia , Hipocampo/citologia , Imuno-Histoquímica , Lisofosfolipídeos/farmacologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Neuritos , Proteômica , Transfecção , Proteínas ras/fisiologia , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética
10.
J Microbiol Biotechnol ; 30(10): 1488-1494, 2020 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-32876072

RESUMO

Rho guanine nucleotide dissociation inhibitor 1 (RhoGDI1) plays important roles in numerous cellular processes, including cell motility, adhesion, and proliferation, by regulating the activity of Rho GTPases. Its expression is altered in various human cancers and is associated with malignant progression. Here, we show that RhoGDI1 interacts with Cullin 3 (CUL3), a scaffold protein for E3 ubiquitin ligase complexes. Ectopic expression of CUL3 increases the ubiquitination of RhoGDI1. Furthermore, potassium channel tetramerization domain containing 5 (KCTD5) also binds to RhoGDI1 and increases its interaction with CUL3. Ectopic expression of KCTD5 increases the ubiquitination of RhoGDI1, whereas its knockdown by RNA interference has the opposite effect. Depletion of KCTD5 or expression of dominant-negative CUL3 (DN-CUL3) enhances the stability of RhoGDI1. Our findings reveal a previously unknown mechanism for controlling RhoGDI1 degradation that involves a CUL3/KCTD5 ubiquitin ligase complex.


Assuntos
Proteínas Culina/genética , Canais de Potássio/genética , Regiões Promotoras Genéticas , Ubiquitinação , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Movimento Celular , Proteínas Culina/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Canais de Potássio/metabolismo , Interferência de RNA , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
11.
Atherosclerosis ; 288: 124-136, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31362179

RESUMO

BACKGROUND AND AIMS: The physiological role of Rho-specific guanine nucleotide dissociation inhibitor (RhoGDI) in vascular remodeling remains unknown. We investigated the function of RhoGDI in angiotensin II (Ang II)-induced vascular remodeling in cultured human aortic vascular smooth muscle cells (HA-VSMCs) and in an Ang II-infusion vascular remodeling mouse model. METHODS: For in vitro assays of HA-VSMCs, proliferation was assessed by BrdU and EdU assays and immunofluorescence analysis of ki-67 expression. RhoGDI1 and RhoGDI2 function and expression were assessed by RNAi, Western blotting and real-time RT-PCR. RhoGDI ubiquitination and SUMOylation levels were evaluated by co-immunoprecipitation and Western blotting. The functions of proteosomal-mediated degradation, ubiquitination, SUMOylation and Ang II receptors were assessed using specific inhibitors. To evaluate the in vivo effects of Ang II and RhoGDI, H & E staining, Masson's trichrome staining, and immunostaining were employed. RESULTS: Ang II treatment of HA-VSMCs for 6 or 48 h promoted RhoGDI1 and RhoGDI2 protein degradation and reduced cell proliferation, which was reversed by proteosome inhibition. In contrast, treatment with Ang II for 12 or 24 h induced dose-dependent cell proliferation without affecting RhoGDI expression. RNA interference of either RhoGDI1 or RhoGDI2 blocked proliferation induced by 12 or 24 h treatment of Ang II. Moreover, Ang II-dependent degradation at 6 and 48 h correlated with RhoGDI ubiquitination and inversely correlated with RhoGDI SUMOylation and cell proliferation. Treatment with specific inhibitors suggests that ubiquitin and SUMO competitively bind to RhoGDI1 and RhoGDI2 to reciprocally regulate RhoGDI stability and HA-VSMC proliferation. Furthermore, inhibition of the Ang II receptor 1 (AT1 receptor), but not the Ang II receptor 2, blocked Ang II-dependent RhoGDI stabilization and proliferation at 12 and 24 h. In mice, Ang II infusion increased the intima-media thickness, collagen and myofiber production and VSMC proliferation, and these effects were shown to be dependent on RhoGDI1, RhoGDI2 and AT1 receptor. Ang II infusion exerted no significant effect on RhoGDI1 and RhoGDI2 protein levels, which were decreased after AT1 receptor inhibition. CONCLUSIONS: Together, the results of this study reveal a novel mechanism by which Ang II regulates RhoGDI stability by SUMOylation and ubiquitination via AT1 receptor activation and thus affects VSMC proliferation and vascular remodeling.


Assuntos
Angiotensina II/farmacologia , Proliferação de Células/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/agonistas , Ubiquitinação , Remodelação Vascular/efeitos dos fármacos , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Inibidor beta de Dissociação do Nucleotídeo Guanina rho/metabolismo , Animais , Células Cultivadas , Humanos , Masculino , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neointima , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , Proteólise , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais , Sumoilação , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Inibidor beta de Dissociação do Nucleotídeo Guanina rho/genética
12.
Nat Commun ; 10(1): 112, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30631060

RESUMO

Wilms tumor gene on the X chromosome (WTX) is a putative tumor suppressor gene in Wilms tumor, but its expression and functions in other tumors are unclear. Colorectal cancer (CRC) is the third leading cause of cancer-related deaths in women and the second leading cause in men in the United States. We demonstrated that WTX frequently lost in CRC which was highly correlated with cell proliferation, tumor invasion and metastasis. Mechanistically, WTX loss disrupts the interaction between RhoGDIα and CDC42 by losing of the binding with RhoGDIα and triggers the activation of CDC42 and its downstream cascades, which promotes CRC development and liver metastasis. The aberrant upregulation of miR-20a/miR-106a were identified as the reason of WTX loss in CRC both in vivo and in vitro. These study defined the mechanism how miR-20a/miR-106a-mediated WTX loss regulates CRC progression and metastasis, and provided a potential therapeutic target for preventing CRC progression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias do Colo/genética , MicroRNAs/genética , Proteínas Supressoras de Tumor/genética , Proteína cdc42 de Ligação ao GTP/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HT29 , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Transdução de Sinais/genética , Transplante Heterólogo , Proteínas Supressoras de Tumor/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
13.
Oncogene ; 37(7): 861-872, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29059157

RESUMO

Eph receptors and their corresponding ephrin ligands have been associated with regulating cell-cell adhesion and motility, and thus have a critical role in various biological processes including tissue morphogenesis and homeostasis, as well as pathogenesis of several diseases. Aberrant regulation of Eph/ephrin signaling pathways is implicated in tumor progression of various human cancers. Here, we show that a Rho family GTPase regulator, Rho guanine nucleotide dissociation inhibitor 1 (RhoGDI1), can interact with ephrinB1, and this interaction is enhanced upon binding the extracellular domain of the cognate EphB2 receptor. Deletion mutagenesis revealed that amino acids 327-334 of the ephrinB1 intracellular domain are critical for the interaction with RhoGDI1. Stimulation with an EphB2 extracellular domain-Fc fusion protein (EphB2-Fc) induces RhoA activation and enhances the motility as well as invasiveness of wild-type ephrinB1-expressing cells. These Eph-Fc-induced effects were markedly diminished in cells expressing the mutant ephrinB1 construct (Δ327-334) that is ineffective at interacting with RhoGDI1. Furthermore, ephrinB1 depletion by siRNA suppresses EphB2-Fc-induced RhoA activation, and reduces motility and invasiveness of the SW480 and Hs578T human cancer cell lines. Our study connects the interaction between RhoGDI1 and ephrinB1 to the promotion of cancer cell behavior associated with tumor progression. This interaction may represent a therapeutic target in cancers that express ephrinB1.


Assuntos
Biomarcadores Tumorais/metabolismo , Movimento Celular , Neoplasias/patologia , Receptor EphB2/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Humanos , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Receptor EphB2/genética , Células Tumorais Cultivadas , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Proteína rhoA de Ligação ao GTP/genética
14.
Sci Rep ; 7: 43798, 2017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28252009

RESUMO

Nicotine can induce the abnormal migration and proliferation of vascular smooth muscle cells (VSMCs). We have previously shown that cytoskeletal proteins and RhoGDIA, a negative regulator of the Rho GTPase pathway, are involved in the nicotine-induced dysfunction of VSMCs. Here, we found that nicotine can activate the Rho GTPase pathway and induce the synthesis of the cytoskeletal proteins in VSMCs through the activation of intracellular downstream signaling pathways, including targets such as MYPT1, PAK1 and PI3K/AKT. Upon nicotine treatment, the mRNA level of RhoGDIA is increased but protein level is decreased both in vitro and in vivo, which suggested a mechanism of post-translational regulation. By the dual luciferase reporter assay, we identified the microRNA-200b (miR-200b) as a modulator of the behavioural changes of VSMCs in response to nicotine through targeting RhoGDIA directly. Introducing miR-200b inhibitors into cultured VSMCs significantly attenuated cell proliferation and migration. Additionally, we found that hypomethylation in the CpG island shore region of miR-200b was responsible for the nicotine-induced miR-200b up-regulation in VSMCs. The study demonstrates that nicotine facilitates VSMC dysfunction through a miR-200b/RhoGDIA/cytoskeleton module through the hypomethylation of miR-200b promoter and suggests that epigenetic modifications may play an important role in the pathological progression.


Assuntos
Citoesqueleto/efeitos dos fármacos , MicroRNAs/genética , Miócitos de Músculo Liso/efeitos dos fármacos , Nicotina/farmacologia , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Regiões 3' não Traduzidas/genética , Animais , Sequência de Bases , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Citoesqueleto/metabolismo , Estimulantes Ganglionares/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Masculino , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
15.
PLoS One ; 11(11): e0166715, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27835684

RESUMO

The Rac1 GTPase plays key roles in cytoskeletal organization, cell motility and a variety of physiological and disease-linked responses. Wild type Rac1 signaling entails dissociation of the GTPase from cytosolic Rac1-Rho GDP dissociation inhibitor (GDI) complexes, translocation to membranes, activation by exchange factors, effector binding, and activation of downstream signaling cascades. Out of those steps, membrane translocation is the less understood. Using transfections of a expression cDNA library in cells expressing a Rac1 bioreporter, we previously identified a cytoskeletal feedback loop nucleated by the F-actin binding protein coronin 1A (Coro1A) that promotes Rac1 translocation to the plasma membrane by facilitating the Pak-dependent dissociation of Rac1-Rho GDI complexes. This screening identified other potential regulators of this process, including WDR26, basigin, and TMEM8A. Here, we show that WDR26 promotes Rac1 translocation following a Coro1A-like and Coro1A-dependent mechanism. By contrast, basigin and TMEM8A stabilize Rac1 at the plasma membrane by inhibiting the internalization of caveolin-rich membrane subdomains. This latter pathway is F-actin-dependent but Coro1A-, Pak- and Rho GDI-independent.


Assuntos
Basigina/genética , Membrana Celular/metabolismo , Glicoproteínas de Membrana/genética , Proteínas dos Microfilamentos/genética , Proteínas/genética , Proteínas rac1 de Ligação ao GTP/genética , Actinas/genética , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Basigina/metabolismo , Células COS , Caveolina 1/genética , Caveolina 1/metabolismo , Fracionamento Celular , Chlorocebus aethiops , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células HeLa , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Transporte Proteico , Proteínas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
16.
Oncotarget ; 7(34): 55110-55127, 2016 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-27391153

RESUMO

Hepatitis B virus (HBV) X protein (HBx), a trans-regulator, is frequently expressed in truncated form without carboxyl-terminus in hepatocellular carcinoma (HCC), but its functional mechanisms are not fully defined. In this report, we investigated frequency of this natural HBx mutant in HCCs and its functional significance. In 102 HBV-infected patients with HCC, C-terminal truncation of HBx, in contrast to full-length HBx, were more prevalent in tumors (70.6%) rather than adjacent non-tumorous tissues (29.4%) (p = 0.0032). Furthermore, two naturally-occurring HBx variants (HBxΔ31), which have 31 amino acids (aa) deleted (codons 123-125/124-126) at C-terminus were identified in tumors and found that the presence of HBxΔ31 significantly correlated with intrahepatic metastasis. We also show that over-expression of HBxΔ31 enhanced hepatoma cell invasion in vitro and metastasis in vivo compared to full-length HBx. Interestingly, HBxΔ31 exerts this function via down-regulating Maspin, RhoGDIα and CAPZB, a set of putative metastasis-suppressors in HCC, in part, by enhancing the binding of transcriptional repressor, myc-associated zinc finger protein (MAZ) to the promoters through physical association with MAZ. Notably, these HBxΔ31-repressed proteins were also significantly lower expression in a subset of HCC tissues with C-terminal HBx truncation than the adjacent non-tumorous tissues, highlighting the clinical significance of this novel HBxΔ31-driven metastatic molecular cascade. Our data suggest that C-terminal truncation of HBx, particularly breakpoints at 124aa, plays a role in enhancing hepatoma cell invasion and metastasis by deregulating a set of metastasis-suppressors partially through MAZ, thus uncovering a novel mechanism for the progression of HBV-associated hepatocarcinogenesis.


Assuntos
Carcinoma Hepatocelular/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Mutação , Transativadores/genética , Adulto , Animais , Proteína de Capeamento de Actina CapZ/genética , Proteína de Capeamento de Actina CapZ/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Feminino , Células Hep G2 , Hepatite B/genética , Hepatite B/virologia , Vírus da Hepatite B/genética , Vírus da Hepatite B/fisiologia , Interações Hospedeiro-Patógeno/genética , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica , Serpinas/genética , Serpinas/metabolismo , Transativadores/química , Transplante Heterólogo , Proteínas Virais Reguladoras e Acessórias , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
17.
Artigo em Inglês | MEDLINE | ID: mdl-27302765

RESUMO

Long-term administration of antipsychotic drugs (APDs) has been theorized to effect drug-induced changes in protein expression in the brain. Our previous findings revealed that ADPs can regulate Rho GDP-dissociation inhibitor 1 (RhoGDI1) expression in glial cells. To reveal whether APDs (haloperidol, risperidone, and clozapine) might regulate cell functions in rat brain by affecting RhoGDI1, RhoGDI1 regulation, RhoGDI1-related Rho family protein, and also MLC2 in brain of 7-day APD treatment rat were examined. Increased expression of RhoGDI1 and RhoA and decreased expression of MLC2, p-MLC2 and ARP2/3 were found in the cortex of APD-treated rats. The activation of RhoA in APD-treated rat cortex was also found. The regulation of RhoGDI1-induced protein expression and its relation to intracellular stress filament production and cell migration were further examined in APD-treated C6 and B35 cells. APD-induced RhoA expression and activation in C6 cells and Cdc42 expression and activation in B35 cells were investigated. In C6 cells, ARP2/3, ROCK1, pMLC2, and PFN1 expressions were decreased, and N-WASP expression was increased by any of the three APDs. In B35 cells, haloperidol decreased ROCK1 expression, but risperidone increased ROCK1 expression. MLC2, p-MLC2, and PFN1 expressions were decreased in B35 cells treated with either risperidone or clozapine. N-WASP expression was decreased by haloperidol and clozapine. We also found all three APDs enhance C6 and B35 F-actin condensation and migration ability.


Assuntos
Antipsicóticos/farmacologia , Citoesqueleto/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Animais , Encéfalo/citologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Serina/metabolismo , Transdução de Sinais/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética
18.
Oncotarget ; 7(15): 19483-98, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26761212

RESUMO

RNA-RNA and protein-RNA interactions are essential for post-transcriptional regulation in normal development and may be deregulated in cancer initiation and progression. The RNA-binding protein PCBP2, an oncogenic protein in human malignant gliomas, is an essential regulator of mRNA and miRNA biogenesis, stability and activity. Here, we identified Rho GDP dissociation inhibitor α (ARHGDIA) as a target mRNA that binds to PCBP2, and we uncovered the role of ARHGDIA as a putative metastasis suppressor through analyses of in vitro and in vivo models of EMT and metastasis. Furthermore, we demonstrated that ARHGDIA is a potential target of miR-151-5p and miR-16 in gliomas. The interaction between PCBP2 and the 3'UTR of the ARHGDIA mRNA may induce a local change in RNA structure that favors subsequent binding of miR-151-5p and miR-16, thus leading to the suppression of ARHGDIA expression. PCBP2 may facilitate miR-151-5p and miR-16 promotion of glioma cell migration and invasion through mitigating the function of ARHGDIA.


Assuntos
Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Glioma/genética , MicroRNAs/genética , Proteínas de Ligação a RNA/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Regiões 3' não Traduzidas/genética , Adulto , Idoso , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Feminino , Glioma/metabolismo , Glioma/patologia , Células HeLa , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Ligação Proteica , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transplante Heterólogo , Adulto Jovem , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
19.
Small GTPases ; 7(2): 107-21, 2016 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-26726844

RESUMO

Nephrotic syndrome (NS) describes a group of kidney disorders in which there is injury to podocyte cells, specialized cells within the kidney's glomerular filtration barrier, allowing proteins to leak into the urine. Three mutations in ARHGDIA, which encodes Rho GDP dissociation inhibitor α (GDIα), have been reported in patients with heritable NS and encode the following amino acid changes: ΔD185, R120X, and G173V. To investigate the impact of these mutations on podocyte function, endogenous GDIα was knocked-down in cultured podocytes by shRNA and then the cells were re-transfected with wild-type or mutant GDIα constructs. Among the 3 prototypical Rho-GTPases, Rac1 was markedly hyperactivated in podocytes with any of the 3 mutant forms of GDIα while the activation of RhoA and Cdc42 was modest and variable. All three mutant GDIα proteins resulted in slow podocyte motility, suggesting that podocytes are sensitive to the relative balance of Rho-GTPase activity. In ΔD185 podocytes, both random and directional movements were impaired and kymograph analysis of the leading edge showed increased protrusion and retraction of leading edge (phase switching). The mutant podocytes also showed impaired actin polymerization, smaller cell size, and increased cellular projections. In the developing kidney, GDIα expression increased as podocytes matured. Conversely, active Rac1 was detected only in immature, but not in mature, podocytes. The results indicate that GDIα has a critical role in suppressing Rac1 activity in mature podocytes, to prevent podocyte injury and nephrotic syndrome.


Assuntos
Mutação , Síndrome Nefrótica/genética , Podócitos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Actinas/química , Animais , Movimento Celular/genética , Tamanho Celular , Ativação Enzimática/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Podócitos/citologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Multimerização Proteica , Estrutura Quaternária de Proteína , Proteólise , Regulação para Cima , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/deficiência
20.
Sci Rep ; 5: 16911, 2015 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-26593060

RESUMO

Spinal cord injury (SCI) is frequently accompanied by a degree of spontaneous functional recovery. The underlying mechanisms through which such recovery is generated remain elusive. In this study, we observed a significant spontaneous motor function recovery 14 to 28 days after spinal cord transection (SCT) in rats. Using a comparative proteomics approach, caudal to the injury, we detected difference in 20 proteins. Two of these proteins, are eukaryotic translation initiation factor 5A1 (eIF5A1) that is involved in cell survival and proliferation, and Rho GDP dissociation inhibitor alpha (RhoGDIα), a member of Rho GDI family that is involved in cytoskeletal reorganization. After confirming the changes in expression levels of these two proteins following SCT, we showed that in vivo eIF5A1 up-regulation and down-regulation significantly increased and decreased, respectively, motor function recovery. In vitro, eIF5A1 overexpression in primary neurons increased cell survival and elongated neurite length while eIF5A1 knockdown reversed these results. We found that RhoGDIα up-regulation and down-regulation rescues the effect of eIF5A1 down-regulation and up-regulation both in vivo and in vitro. Therefore, we have identified eIF5A1/RhoGDIα pathway as a new therapeutic target for treatment of spinal cord injured patients.


Assuntos
Fatores de Iniciação de Peptídeos/genética , Proteínas de Ligação a RNA/genética , Recuperação de Função Fisiológica/fisiologia , Transdução de Sinais/genética , Traumatismos da Medula Espinal/genética , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Proliferação de Células/genética , Sobrevivência Celular/genética , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Feminino , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Atividade Motora/fisiologia , Neurônios/metabolismo , Neurônios/ultraestrutura , Fatores de Iniciação de Peptídeos/antagonistas & inibidores , Fatores de Iniciação de Peptídeos/metabolismo , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/metabolismo , Ratos , Ratos Sprague-Dawley , Remissão Espontânea , Medula Espinal/metabolismo , Medula Espinal/patologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/antagonistas & inibidores , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Fator de Iniciação de Tradução Eucariótico 5A
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...