Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Exp Neurol ; 343: 113788, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34147481

RESUMO

Increasing the intrinsic regeneration potential of neurons is the key to promote axon regeneration and repair of nerve injury. Therefore, identifying the molecular switches that respond to nerve injury may play critical role in improving intrinsic regeneration ability. The mechanisms by which injury unlocks the intrinsic axonal growth competence of mature neurons are not well understood. The present study identified the key regulatory genes after sciatic nerve crush injury by RNA sequencing (RNA-Seq) and found that the hub gene Vav1 was highly expressed at both early response and regenerative stages of sciatic nerve injury. Furthermore, Vav1 was required for axon regeneration of dorsal root ganglia (DRG) neurons and functional recovery. Krüppel-like factor 2 (Klf2) was induced by retrograde Ca2+ signaling from injured axons and could directly promote Vav1 transcription in adult DRG neurons. The increased Vav1 then promoted axon regeneration by activating Rac1 GTPase independent of its tyrosine phosphorylation. Collectively, these findings break through previous limited cognition of Vav1, and first reveal a crucial role of Vav1 as a molecular switch in response to axonal injury for promoting axon regeneration, which might further serve as a novel molecular therapeutic target for clinical nerve injury repair.


Assuntos
Axônios/fisiologia , Fatores de Transcrição Kruppel-Like/biossíntese , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/metabolismo , Proteínas Proto-Oncogênicas c-vav/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese , Animais , Células Cultivadas , Masculino , Traumatismos dos Nervos Periféricos/patologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores
2.
Mol Cancer ; 20(1): 77, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34006303

RESUMO

BACKGROUND: KDM6A, a histone demethylase, is frequently mutated in bladder cancer (BCa). However, the role and detailed molecular mechanism of KDM6A involved in bladder cancer progression remains unknown. METHODS: Tissue specimens were used to determine the expression levels and prognostic values of KDM6A and ARHGDIB. The MTT, colony formation, wound healing and Transwell migration and invasion assays were employed to detect the BCa cell proliferation, migration and invasion, respectively. Chemotaxis of macrophages was used to evaluate the ability of KDM6A to recruit macrophages. A subcutaneous tumour model and tail vein tumour injection in nude mice were used to assess the role of KDM6A in vivo. RNA sequencing, qPCR, Western blot, ChIP and phalloidin staining assay were performed to investigate the molecular functions of KDM6A. Dual-luciferase reporter assay was used to determine the effects of KDM6A and FOXA1 on the promoters of the ARHGDIB and KDM6A. RESULTS: We showed that the KDM6A inhibited the motility and invasiveness of the BCa cells. Mechanistically, KDM6A promotes the transcription of ARHGDIB by demethylating histone H3 lysine di/trimethylation (H3K27me2/3) and consequently leads to inhibition of Rac1. EZH2, which catalyses the methylation of H3K27, functions to silence ARHGDIB expression, and an EZH2 inhibitor can neutralize the metastatic effect caused by KDM6A deficiency. Furthermore, we demonstrated that FOXA1 directly binds to the KDM6A promoter and thus transactivates KDM6A, leading to diminished metastatic potential. CONCLUSION: Our findings establish the critical role of the FOXA1-KDM6A-ARHGDIB axis in restraining the malignancy of BCa and identify KDM6A and EZH2 as potential therapeutic targets in the management of BCa.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Histona Desmetilases/metabolismo , Neoplasias da Bexiga Urinária/patologia , Proteínas rac1 de Ligação ao GTP/biossíntese , Inibidor beta de Dissociação do Nucleotídeo Guanina rho/metabolismo , Animais , Movimento Celular/fisiologia , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia
3.
Cancer Biol Ther ; 21(10): 888-890, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-32866423

RESUMO

Tumor metabolism is exemplified by the increased rate of glucose utilization, a biochemical signature of cancer cells. The enhanced glucose hydrolysis enabled by the augmentation of glycolytic flux and the pentose phosphate pathway (PPP) plays a pivotal role in the growth and survival of neoplastic cells. In a recent report, it has been shown that in human breast cancer the GTP binding protein, Rac1 enables resistance to therapy, particularly against the DNA-damaging therapeutics. Significantly, the findings demonstrate that Rac1-dependent chemoresistance involves the upregulation of glycolytic flux as well as PPP. Using multiple approaches, the study demonstrates that disruption of Rac1 activity sensitizes cancer cells to DNA-damaging agents. More importantly, the data uncover a previously unknown PPP regulatory role of Rac1 in breast cancer. Finally, the authors also show the effectiveness and the feasibility of in vivo targeting of Rac1 to enhance the chemosensitivity of breast cancer. This elegant report provokes scientific curiosity to expand our understanding of the intricacies of the role and regulation of Rac1 in cancer.


Assuntos
Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas rac1 de Ligação ao GTP/biossíntese , Linhagem Celular Tumoral , Humanos , Terapia de Alvo Molecular , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Proteínas rac1 de Ligação ao GTP/genética
4.
Curr Neurovasc Res ; 17(3): 259-266, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32294039

RESUMO

OBJECTIVE: Ischemia-reperfusion (I/R) injury is a pathological feature of ischemic stroke. This study investigated the regulatory role of miR-485-5p in I/R injury. METHODS: SH-SY5Y cells were induced with oxygen and glucose deprivation/reoxygenation (OGD/R) to mimic I/R injury in vitro. Cells were transfected with designated constructs (miR-485- 5p mimics, miR-485-5p inhibitor, lentiviral vectors overexpressing Rac1 or their corresponding controls). Cell viability was evaluated using the MTT assay. The concentrations of lactate dehydrogenase, malondialdehyde, and reactive oxygen species were detected to indicate the degree of oxidative stress. Flow cytometry and caspase-3 activity assay were used for apoptosis assessment. Dual-luciferase reporter assay was performed to confirm that Rac family small GTPase 1 (Rac1) was a downstream gene of miR-485-5p. RESULTS: OGD/R resulted in decreased cell viability, elevated oxidative stress, increased apoptosis, and downregulated miR-485-5p expression in SH-SY5Y cells. MiR-485-5p upregulation alleviated I/R injury, evidenced by improved cell viability, decreased oxidative markers, and reduced apoptotic rate. OGD/R increased the levels of Rac1 and neurogenic locus notch homolog protein 2 (Notch2) signaling-related proteins in cells with normal miR-485-5p expression, whereas miR- 485-5p overexpression successfully suppressed OGD/R-induced upregulation of these proteins. Furthermore, the delivery of vectors overexpressing Rac1 in miR-485-5p mimics-transfected cells reversed the protective effect of miR-485-5p in cells with OGD/R-induced injury. CONCLUSION: This study showed that miR-485-5p protected cells following I/R injury via targeting Rac1/Notch2 signaling suggest that targeted upregulation of miR-485-5p might be a promising therapeutic option for the protection against I/R injury.


Assuntos
Isquemia Encefálica/metabolismo , MicroRNAs/biossíntese , Neurônios/metabolismo , Receptor Notch2/biossíntese , Traumatismo por Reperfusão/metabolismo , Proteínas rac1 de Ligação ao GTP/biossíntese , Isquemia Encefálica/patologia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Glucose/deficiência , Humanos , Neurônios/patologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais/fisiologia
5.
Mol Oncol ; 13(9): 2010-2030, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31314174

RESUMO

Development of chemoresistance remains a major challenge in treating esophageal squamous cell carcinoma (ESCC) patients despite treatment advances. However, the role of RAC1 in chemoresistance of ESCC and the underlying mechanisms remain largely unknown. In this study, we found that higher levels of RAC1 expression were associated with poorer prognosis in ESCC patients. Enhanced RAC1 expression increased cell proliferation, migration, and chemoresistance in vitro. Combination therapy using RAC1 inhibitor EHop-016 and cisplatin significantly promoted cell viability inhibition, G2/M phase cycle arrest, and apoptosis when compared to each monotherapy. Mechanistically, glycolysis was significantly downregulated in the RAC1 inhibitor monotherapy group and the combination group via inhibiting AKT/FOXO3a signaling when compared to the control group. Moreover, the silencing of RAC1 inhibited AKT/FOXO3a signaling and cell glycolysis while the upregulation of RAC1 produced an opposite effect. In murine xenograft models, the tumor volume and the expression of glycolytic enzymes were significantly reduced in combination therapy when compared to each monotherapy group. Overall, our study demonstrates that targeting RAC1 with an inhibitor overcomes cisplatin resistance in ESCC by suppressing glycolytic enzymes, which provides a promising strategy for treatment of ESCC in clinical practice.


Assuntos
Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Proteínas de Neoplasias/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese , Animais , Linhagem Celular Tumoral , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/metabolismo , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Inativação Gênica , Glicólise/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas rac1 de Ligação ao GTP/genética
6.
Biochem Pharmacol ; 164: 82-93, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30936017

RESUMO

Cardiotoxicity is the dose limiting adverse effect of anthracycline-based anticancer therapy. Inhibitor studies point to Rac1 as therapeutic target to prevent anthracycline-induced cardiotoxicity. Yet, supporting genetic evidence is still missing and the pathophysiological relevance of different cardiac cell types is unclear. Here, we employed a tamoxifen-inducible cardiomyocyte-specific rac1 knock-out mouse model (Rac1flox/flox/MHC-MerCreMer) to investigate the impact of Rac1 expression in cardiomyocytes on cardiac injury following doxorubicin treatment. Distinctive stress responses resulting from doxorubicin treatment were observed, including upregulation of systemic markers of inflammation (IL-6, IL-1α, MCP-1), cardiac damage (ANP, BNP), DNA damage (i.e. DNA double-strand breaks (DSB)), DNA damage response (DDR) and cell death. Measuring the acute doxorubicin response, the serum level of MCP-1 was elevated, cardiac mRNA expression of Hsp70 was reduced and cardiac DDR was specifically enhanced in Rac1 deficient mice. The frequency of apoptotic heart cells remained unaffected by Rac1. Employing a subactue model, the number of doxorubicin-induced DSB was significantly reduced if Rac1 is absent. Yet, the doxorubicin-triggered increase in serum ANP and BNP levels remained unaffected by Rac1. Overall, knock-out of rac1 in cardiomyocytes confers partial protection against doxorubicin-induced cardiac injury. Hence, the data provide first genetic evidence supporting the view that pharmacological targeting of Rac1 is useful to widen the therapeutic window of anthracycline-based anticancer therapy by alleviating acute/subacute cardiomyocyte damage. Furthermore, considering published data obtained from the use of pharmacological Rac1 inhibitors, the results of our study indicate that Rac1-regulated functions of cardiac cell types others than cardiomyocytes additionally influence the adverse outcomes of anthracycline treatment on the heart.


Assuntos
Antraciclinas/toxicidade , Cardiopatias/induzido quimicamente , Cardiopatias/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Neuropeptídeos/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese , Animais , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuropeptídeos/genética , Proteínas rac1 de Ligação ao GTP/genética
7.
J Neurochem ; 147(3): 409-428, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30091236

RESUMO

The Parkinson's disease (PD)-causative leucine-rich repeat kinase 2 (LRRK2) belongs to the Roco family of G-proteins comprising a Ras-of-complex (Roc) domain followed by a C-terminal of Roc (COR) domain in tandem (called Roc-COR domain). Two prokaryotic Roc-COR domains have been characterized as 'G proteins activated by guanine nucleotide-dependent dimerization' (GADs), which require dimerization for activation of their GTPase activity and bind guanine nucleotides with relatively low affinities. Additionally, LRRK2 Roc domain in isolation binds guanine nucleotides with relatively low affinities. As such, LRRK2 GTPase domain was predicted to be a GAD. Herein, we describe the design and high-level expression of human LRRK2 Roc-COR domain (LRRK2 Roc-COR). Biochemical analyses of LRRK2 Roc-COR reveal that it forms homodimers, with the C-terminal portion of COR mediating its dimerization. Furthermore, it co-purifies and binds Mg2+ GTP/GDP at 1 : 1 stoichiometry, and it hydrolyzes GTP with Km  and kcat  of 22 nM and 4.70 × 10-4  min-1 ,  respectively. Thus, even though LRRK2 Roc-COR forms GAD-like homodimers, it exhibits conventional Ras-like GTPase properties, with high-affinity binding of Mg2+ -GTP/GDP and low intrinsic catalytic activity. The PD-causative Y1699C mutation mapped to the COR domain was previously reported to reduce the GTPase activity of full-length LRRK2. In contrast, this mutation induces no change in the GTPase activity, and only slight perturbations in the secondary structure contents of LRRK2 Roc-COR. As this mutation does not directly affect the GTPase activity of the isolated Roc-COR tandem, it is possible that the effects of this mutation on full-length LRRK2 occur via other functional domains. Open Practices Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.


Assuntos
GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Genes ras/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Animais , Dimerização , Escherichia coli , Regulação Enzimológica da Expressão Gênica/genética , Nucleotídeos de Guanina/metabolismo , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/química , Magnésio/metabolismo , Camundongos , Mutação/genética , Neuropeptídeos/biossíntese , Neuropeptídeos/genética , Multimerização Proteica , Estrutura Secundária de Proteína/genética , Proteínas Recombinantes , Proteínas rac1 de Ligação ao GTP/biossíntese , Proteínas rac1 de Ligação ao GTP/genética
8.
Neoplasia ; 19(11): 868-884, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28938158

RESUMO

Directional cell migration is of fundamental importance to a variety of biological events, including metastasis of malignant cells. Herein, we specifically investigated SET oncoprotein, a subunit of the recently identified inhibitor of acetyltransferases (INHAT) complex and identified its role in the establishment of front-rear cell polarity and directional migration in Esophageal Squamous Cell Carcinoma (ESCC). We further define the molecular circuits that govern these processes by showing that SET modulated DOCK7/RAC1 and cofilin signaling events. Moreover, a detailed analysis of the spatial distribution of RAC1 and cofilin allowed us to decipher the synergistical contributions of the two in coordinating the advancing dynamics by measuring architectures, polarities, and cytoskeletal organizations of the lamellipodia leading edges. In further investigations in vivo, we identified their unique role at multiple levels of the invasive cascade for SET cell and indicate the necessity for their functional balance to enable efficient invasion as well. Additionally, SET epigenetically repressed miR-30c expression by deacetylating histones H2B and H4 on its promoter, which was functionally important for the biological effects of SET in our cell-context. Finally, we corroborated our findings in vivo by evaluating the clinical relevance of SET signaling in the metastatic burden in mice and a large series of patients with ESCC at diagnosis, observing it's significance in predicting metastasis formation. Our findings uncovered a novel signaling network initiated by SET that epigenetically modulated ESCC properties and suggest that targeting the regulatory axis might be a promising strategy to inhibit migration and metastasis.


Assuntos
Carcinoma de Células Escamosas/genética , Movimento Celular/genética , Epigênese Genética/genética , Neoplasias Esofágicas/genética , Histona Acetiltransferases/genética , Chaperonas de Histonas/genética , Oncogenes/genética , Fatores de Transcrição/genética , Animais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteínas de Ligação a DNA , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Proteínas Ativadoras de GTPase/biossíntese , Proteínas Ativadoras de GTPase/genética , Fatores de Troca do Nucleotídeo Guanina , Células HEK293 , Histona Acetiltransferases/biossíntese , Chaperonas de Histonas/biossíntese , Humanos , Masculino , Camundongos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Ratos , Fatores de Transcrição/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese , Proteínas rac1 de Ligação ao GTP/genética
9.
Tumour Biol ; 39(7): 1010428317710908, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28671041

RESUMO

The change of cell polarity is usually associated with invasion and metastasis. Partial reverse cell polarity in IDC-NOS may play a role in lymphatic tumor spread. Rac1 is a kind of polarity related protein. It plays an important role in invasion and metastasis in tumors. We here investigated the expression of Rac1 and partial reverse cell polarity status in breast cancer and evaluated their value for prognosis in breast cancer. The association of the expression of Rac1 and MUC-1 with clinicopathological parameters and prognostic significance was evaluated in 162 cases of IDC-NOS paraffin-embedded tissues by immunohistochemical method. The Rac1 messenger RNA expression was measured by real-time polymerase chain reaction in 30 breast cancer patients, which was divided into two groups of partial reverse cell polarity and no partial reverse cell polarity. We found that lymph node metastasis of partial reverse cell polarity patients was higher than no partial reverse cell polarity patients (Z = -4.030, p = 0.000). Rac1 was upregulated in partial reverse cell polarity group than no partial reverse cell polarity group (Z = -3.164, p = 0.002), and there was correlationship between the expression of Rac1 and partial reverse cell polarity status (rs = 0.249, p = 0.001). The level of Rac1 messenger RNA expression in partial reverse cell polarity group was significantly higher compared to no partial reverse cell polarity group (t = -2.527, p = 0.017). Overexpression of Rac1 and partial reverse cell polarity correlates with poor prognosis of IDC-NOS patients (p = 0.011). Partial reverse cell polarity and lymph node metastasis remained as independent predictors for poor disease-free survival of IDC-NOS (p = 0.023, p = 0.046). Our study suggests that partial reverse cell polarity may lead to poor prognosis of breast cancer. Overexpression of Rac1 may lead to polarity change in IDC-NOS of the breast. Therefore, Rac1 could be a therapeutic target for breast cancer.


Assuntos
Biomarcadores Tumorais/biossíntese , Carcinoma Ductal de Mama/genética , Prognóstico , Proteínas rac1 de Ligação ao GTP/biossíntese , Adulto , Idoso , Biomarcadores Tumorais/genética , Carcinoma Ductal de Mama/patologia , Polaridade Celular , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Estadiamento de Neoplasias , Proteínas rac1 de Ligação ao GTP/genética
10.
Tumour Biol ; 39(5): 1010428317698392, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28459214

RESUMO

Several studies have proved that Vav2 gene is associated with the carcinogenesis of some tumors, but the relationship between Vav2 gene and gastric cancer remains unclear. Purpose of this study is to detect the expression of Vav2 protein in gastric cancer tissues and to evaluate the clinical value of Vav2. Furthermore, both effect of Vav2 gene on invasion and metastasis of gastric cancer cells and its mechanism are investigated in vitro. Results showed that positive rate of Vav2 protein was significantly higher in gastric cancer tissues than in adjacent tissues and notably higher in metastatic lymph nodes than in gastric cancer tissues. Results of western blot were consistent with immunohistochemistry. Expression of Vav2 protein in gastric cancer tissues was related to degree of tumor differentiation, lymph node metastasis, and clinical stages. Inhibition of endogenous Vav2 in BGC823 cells led to significantly decreased cell activity, migration, and invasion ability in vitro, and expression of Rac1, MMP-2, and MMP-9 decreased, whereas expression of TIMP-1 increased. We concluded that Vav2 might promote invasion and metastasis of gastric cancer by regulating some invasion and metastasis-related genes.


Assuntos
Invasividade Neoplásica/genética , Proteínas Proto-Oncogênicas c-vav/genética , Neoplasias Gástricas/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , Masculino , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Proteínas Proto-Oncogênicas c-vav/antagonistas & inibidores , Neoplasias Gástricas/patologia , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese
11.
J Am Heart Assoc ; 6(3)2017 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-28246076

RESUMO

BACKGROUND: Endothelial dysfunction contributes significantly to the development of vascular diseases. However, a therapy able to reduce this derangement still needs to be identified. We evaluated the effects of pharmacological inhibition of Rac1, a small GTPase protein promoting oxidative stress, in human endothelial dysfunction. METHODS AND RESULTS: We performed vascular reactivity studies to test the effects of NSC23766, a Rac1 inhibitor, on endothelium-dependent vasorelaxation of saphenous vein segments collected from 85 subjects who had undergone surgery for venous insufficiency and from 11 patients who had undergone peripheral vascular surgery. The endothelium-dependent vasorelaxation of the varicose segments of saphenous veins collected from patients with venous insufficiency was markedly impaired and was also significantly lower than that observed in control nonvaricose vein tracts from the same veins. Rac1 activity, reactive oxygen species levels, and reduced nicotine adenine dinucleotide phosphate (NADPH) oxidase activity were significantly increased in varicose veins, and NSC23766 was able to significantly improve endothelium-dependent vasorelaxation of dysfunctional saphenous vein portions in a nitric oxide-dependent manner. These effects were paralleled by a significant reduction of NADPH oxidase activity and activation of endothelial nitric oxide synthase. Finally, we further corroborated this data by demonstrating that Rac1 inhibition significantly improves venous endothelial function and reduces NADPH oxidase activity in saphenous vein grafts harvested from patients with vascular diseases undergoing peripheral bypass surgery. CONCLUSIONS: Rac1 pharmacological inhibition rescues endothelial function and reduces oxidative stress in dysfunctional veins. Rac1 inhibition may represent a potential therapeutic intervention to reduce human endothelial dysfunction and subsequently vascular diseases in various clinical settings.


Assuntos
Aminoquinolinas/farmacologia , Endotélio Vascular/fisiopatologia , Pirimidinas/farmacologia , Veia Safena/fisiopatologia , Vasodilatação/efeitos dos fármacos , Insuficiência Venosa/fisiopatologia , Proteínas rac1 de Ligação ao GTP/biossíntese , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença Crônica , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Humanos , Immunoblotting , Masculino , Pessoa de Meia-Idade , NADP/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Veia Safena/efeitos dos fármacos , Veia Safena/metabolismo , Insuficiência Venosa/metabolismo , Adulto Jovem , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores
12.
Dev Cell ; 38(5): 522-35, 2016 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-27623383

RESUMO

An important feature of the mammary gland is its ability to undergo repeated morphological changes during each reproductive cycle with profound tissue expansion in pregnancy and regression in involution. However, the mechanisms that determine the tissue's cyclic regenerative capacity remain elusive. We have now discovered that Cre-Lox ablation of Rac1 in mammary epithelia causes gross enlargement of the epithelial tree and defective alveolar regeneration in a second pregnancy. Architectural defects arise because loss of Rac1 disrupts clearance in involution following the first lactation. We show that Rac1 is crucial for mammary alveolar epithelia to switch from secretion to a phagocytic mode and rapidly remove dying neighbors. Moreover, Rac1 restricts the extrusion of dying cells into the lumen, thus promoting their eradication by live phagocytic neighbors while within the epithelium. Without Rac1, residual milk and cell corpses flood the ductal network, causing gross dilation, chronic inflammation, and defective future regeneration.


Assuntos
Inflamação/genética , Glândulas Mamárias Humanas/metabolismo , Regeneração/genética , Proteínas rac1 de Ligação ao GTP/genética , Animais , Apoptose/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Epitélio/patologia , Feminino , Humanos , Inflamação/patologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/patologia , Camundongos Knockout , Fagócitos/metabolismo , Gravidez , Proteínas rac1 de Ligação ao GTP/biossíntese
13.
Biochim Biophys Acta ; 1866(1): 51-63, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27345584

RESUMO

A recently acknowledged morphological pathway to colorectal cancer originates from precursor polyps with a serrated appearance due to branching and folding of the colon epithelium. This serrated origin accounts for up to 30% of all colorectal tumors but these are heterogeneous regarding molecular characteristics and patient outcome. Here we review the current knowledge about the classification of this tumor subtype and its association with five key features: mutation status of the BRAF or KRAS genes, the CpG island methylation phenotype, microsatellite instability, immune cell infiltration, and overexpression of GTPase RAC1b. Subsequently, available therapeutic approaches for targeting these molecular characteristics are presented and critically discussed.


Assuntos
Neoplasias Colorretais/genética , Metilação de DNA/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas rac1 de Ligação ao GTP/genética , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/patologia , Ilhas de CpG/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Instabilidade de Microssatélites , Mutação , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/biossíntese
14.
Invest Ophthalmol Vis Sci ; 57(6): 2522-32, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27159442

RESUMO

PURPOSE: To characterize whether the activation of Rac1 is involved in the inflammatory effects produced by Amadori-glycated albumin (AGA) in retinal microglia and to further explore the pathologic pathways of AGA-induced retinal microglial activation and inflammation via a microRNA-dependent mechanism. METHODS: Primary rat retinal microglia were separated and cultured. The levels of TNF-α mRNA and soluble TNF-α produced by the retinal microglia in response to AGA were measured with quantitative RT-PCR (qRT-PCR) and ELISA. In addition, the GTPase activity of Rac1 was measured using a Rac activation assay kit. Luciferase reporter assays were used to validate the regulation of a putative target of microRNA-124 (miR-124). RESULTS: Amadori-glycated albumin significantly stimulated the expression of TNF-α mRNA and protein in cultured retinal microglial cells in a dose- and time-dependent manner. MicroRNA-124 expression was consistently suppressed by AGA, and the inhibitory effect was controlled by histone deacetylases (HDACs). Amadori-glycated albumin induced an increase in Rac1 activation in a dose- and time-dependent manner. Furthermore, our data indicated that Rac1 activation-mediated reactive oxygen species production stimulates p65 NF-κB phosphorylation and induces TNF-α release from retinal microglial cells. Finally, we demonstrated that miR-124 directly controls Rac1 expression. CONCLUSIONS: The current study indicated that AGA-induced retinal microglial activation and inflammation occur via a miR-124-dependent mechanism.


Assuntos
Retinopatia Diabética/genética , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , Microglia/metabolismo , Células Ganglionares da Retina/metabolismo , Albumina Sérica/farmacologia , Proteínas rac1 de Ligação ao GTP/genética , Animais , Animais Recém-Nascidos , Western Blotting , Contagem de Células , Células Cultivadas , Diabetes Mellitus Experimental , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Ensaio de Imunoadsorção Enzimática , Produtos Finais de Glicação Avançada , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , MicroRNAs/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Células Ganglionares da Retina/patologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Proteínas rac1 de Ligação ao GTP/biossíntese , Albumina Sérica Glicada
15.
Mol Med Rep ; 13(5): 4414-20, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27052944

RESUMO

The Ras-related C3 botulinum toxin substrate 1 (Rac1)-WASP-family verprolin-homologous protein-2 (WAVE2)-actin-related protein 2/3 (Arp2/3) signaling pathway has been identified to be involved in cell migration and invasion in various types of cancer cell. Cofilin­1 (CFL­1), which is regulated by the Rac1­WAVE2­Arp2/3 signaling pathway, may promote radioresistance in glioma. Therefore, the present study aimed to investigate the potential role of the Rac1­WAVE2­Arp2/3 signaling pathway in radioresistance in U251 human glioma cells and elucidate its affect on CFL­1 expression. Western blot analysis was performed to evaluate the protein expression of CFL­1. In the present study, Rac1 was inhibited by NSC 23766, WAVE2 was inhibited by transfection with short hairpin (sh)RNA­WAVE2 using Lipofectamine™ 2000 and Arp2/3 was inhibited by CK­666. Cell viability was measured using the 3­(4,5­dimethylthiazol­2­yl)-2,5­diphenyltetrazolium bromide assay, the cell migration ability was examined by a wound­healing assay, and the cell invasion ability was assessed using a Transwell culture chamber system. The results showed that inhibition of the Rac1­WAVE2­Arp2/3 signaling pathway using NSC 23766, shRNA­WAVE2 or CK­666 reduced the cell viability, migration and invasion abilities in U251 human glioma cells, concordant with a reduced expression of CFL­1. Furthermore, the expression of CFL­1 was significantly increased in radioresistant U251 glioma cells when compared with normal U251 human glioma cells. These findings indicate that inhibition of the Rac1­WAVE2­Arp2/3 signaling pathway may promote radiosensitivity, which may partially result from the downregulation of CFL­1 in U251 human glioma cells.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/biossíntese , Cofilina 1/biossíntese , Regulação para Baixo/efeitos da radiação , Raios gama , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioma/metabolismo , Proteínas de Neoplasias/biossíntese , Tolerância a Radiação , Transdução de Sinais/efeitos da radiação , Família de Proteínas da Síndrome de Wiskott-Aldrich/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Linhagem Celular Tumoral , Cofilina 1/genética , Glioma/genética , Glioma/patologia , Glioma/radioterapia , Humanos , Proteínas de Neoplasias/genética , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética , Proteínas rac1 de Ligação ao GTP/genética
16.
Tumour Biol ; 37(8): 11049-55, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26897750

RESUMO

Rab23 overexpression has been implicated in several human cancers. However, its biological roles and molecular mechanism in astrocytoma have not been elucidated. The aim of this study is to explore clinical significance and biological roles of Rab23 in astrocytoma. We observed negative Rab23 staining in normal astrocytes and positive staining in 39 out of 86 (45 %) astrocytoma specimens using immunohistochemistry. The positive rate of Rab23 was higher in grades III and IV (56.5 %, 26/46) than grades I + II astrocytomas (32.5 %, 13/40, p < 0.05). Transfection of Rab23 plasmid was performed induced A172 cell proliferation, colony formation, invasion, and migration, while Rab23 depletion with siRNA reduced these abilities of U87 cells. In addition, we found that Rab23 transfection upregulated while its depletion reduced Rac1 activity. Treatment of transfected cells with a Rac1 inhibitor decreased Rac1 activity and invasion. In conclusion, Rab23 serves as an important oncoprotein in human astrocytoma by regulating cell invasion and migration through Rac1 activity.


Assuntos
Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Movimento Celular , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas rab de Ligação ao GTP/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese , Adulto , Idoso , Western Blotting , Movimento Celular/genética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Reação em Cadeia da Polimerase em Tempo Real , Regulação para Cima
17.
Oncotarget ; 7(9): 10498-512, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26871290

RESUMO

Diallyl disulfide (DADS) has been shown to have multi-targeted antitumor activities. We have previously discovered that it has a repressive effect on LIM kinase-1 (LIMK1) expression in gastric cancer MGC803 cells. This suggests that DADS may inhibit epithelial-mesenchymal transition (EMT) by downregulating LIMK1, resulting in the inhibition of invasion and growth in gastric cancer. In this study, we reveal that LIMK1 expression is correlated with tumor differentiation, invasion depth, clinical stage, lymph node metastasis, and poor prognosis. DADS downregulated the Rac1-Pak1/Rock1-LIMK1 pathway in MGC803 cells, as shown by decreased p-LIMK1 and p-cofilin1 levels, and suppressed cell migration and invasion. Knockdown and overexpression experiments performed in vitro demonstrated that downregulating LIMK1 with DADS resulted in restrained EMT that was coupled with decreased matrix metalloproteinase-9 (MMP-9) and increased tissue inhibitor of metalloproteinase-3 (TIMP-3) expression. In in vitro and in vivo experiments, the DADS-induced suppression of cell proliferation was enhanced and antagonized by the knockdown and overexpression of LIMK1, respectively. Similar results were observed for DADS-induced changes in the expression of vimentin, CD34, Ki-67, and E-cadherin in xenografted tumors. These results indicate that downregulation of LIMK1 by DADS could explain the inhibition of EMT, invasion and proliferation in gastric cancer cells.


Assuntos
Compostos Alílicos/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dissulfetos/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Quinases Lim/metabolismo , Neoplasias Gástricas/patologia , Animais , Antígenos CD34/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Cofilina 1/metabolismo , Regulação para Baixo , Humanos , Antígeno Ki-67/metabolismo , Quinases Lim/biossíntese , Quinases Lim/genética , Metástase Linfática , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/patologia , Transplante de Neoplasias , Neoplasias Gástricas/mortalidade , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Transplante Heterólogo , Vimentina/metabolismo , Quinases Ativadas por p21/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese , Quinases Associadas a rho/biossíntese
18.
Cell Death Differ ; 23(1): 169-81, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26184908

RESUMO

Successful embryo implantation requires functional luminal epithelia to establish uterine receptivity and blastocyst-uterine adhesion. During the configuration of uterine receptivity from prereceptive phase, the luminal epithelium undergoes dynamic membrane reorganization and depolarization. This timely regulated epithelial membrane maturation and precisely maintained epithelial integrity are critical for embryo implantation in both humans and mice. However, it remained largely unexplored with respect to potential signaling cascades governing this functional epithelial transformation prior to implantation. Using multiple genetic and cellular approaches combined with uterine conditional Rac1 deletion mouse model, we demonstrated herein that Rac1, a small GTPase, is spatiotemporally expressed in the periimplantation uterus, and uterine depletion of Rac1 induces premature decrease of epithelial apical-basal polarity and defective junction remodeling, leading to disrupted uterine receptivity and implantation failure. Further investigations identified Pak1-ERM as a downstream signaling cascade upon Rac1 activation in the luminal epithelium necessary for uterine receptivity. In addition, we also demonstrated that Rac1 via P38 MAPK signaling ensures timely epithelial apoptotic death at postimplantation. Besides uncovering a potentially important molecule machinery governing uterine luminal integrity for embryo implantation, our finding has high clinical relevance, because Rac1 is essential for normal endometrial functions in women.


Assuntos
Proteínas de Ligação a DNA/genética , Implantação do Embrião/genética , Neuropeptídeos/genética , Fatores de Transcrição/genética , Quinases Ativadas por p21/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas rac1 de Ligação ao GTP/genética , Animais , Blastocisto/metabolismo , Proteínas de Ligação a DNA/biossíntese , Implantação do Embrião/fisiologia , Endométrio/crescimento & desenvolvimento , Endométrio/metabolismo , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Feminino , Humanos , Camundongos , Neuropeptídeos/biossíntese , Transdução de Sinais/genética , Fatores de Transcrição/biossíntese , Útero/metabolismo , Útero/fisiologia , Quinases Ativadas por p21/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese
19.
Toxins (Basel) ; 7(11): 4610-21, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26556375

RESUMO

Eukaryotic cells secrete extracellular vesicles (EVs), either constitutively or in a regulated manner, which represent an important mode of intercellular communication. EVs serve as vehicles for transfer between cells of membrane and cytosolic proteins, lipids and RNA. Furthermore, certain bacterial protein toxins, or possibly their derived messages, can be transferred cell to cell via EVs. We have herein demonstrated that eukaryotic EVs represent an additional route of cell-to-cell propagation for the Escherichia coli protein toxin cytotoxic necrotizing factor 1 (CNF1). Our results prove that EVs from CNF1 pre-infected epithelial cells can induce cytoskeleton changes, Rac1 and NF-κB activation comparable to that triggered by CNF1. The observation that the toxin is detectable inside EVs derived from CNF1-intoxicated cells strongly supports the hypothesis that extracellular vesicles can offer to the toxin a novel route to travel from cell to cell. Since anthrax and tetanus toxins have also been reported to engage in the same process, we can hypothesize that EVs represent a common mechanism exploited by bacterial toxins to enhance their pathogenicity.


Assuntos
Toxinas Bacterianas/farmacologia , Toxinas Bacterianas/uso terapêutico , Proteínas de Escherichia coli/farmacologia , Proteínas de Escherichia coli/uso terapêutico , Vesículas Extracelulares/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Citoesqueleto/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Humanos , NF-kappa B/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/biossíntese , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos
20.
Tumour Biol ; 36(12): 9589-97, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26142737

RESUMO

The importance of tumor-stromal cell interactions in breast tumor progression and invasion is well established. Here, an evaluation of differential genomic profiles of carcinoma-associated fibroblasts (CAFs) compared to fibroblasts derived from tissues adjacent to fibroadenomas (NAFs) revealed altered focal adhesion pathways. These data were validated through confocal assays. To verify the possible role of fibroblasts in lymph node invasion, we constructed a tissue microarray consisting of primary breast cancer samples and corresponding lymph node metastasis and compared the expression of adhesion markers RhoA and Rac1 in fibroblasts located at these different locations. Two distinct tissue microarrays were constructed from the stromal component of 43 primary tumors and matched lymph node samples, respectively. Fibroblasts were characterized for their expression of α-smooth muscle actin (α-SMA) and vimentin. Moreover, we verified the level of these proteins in the stromal compartment from normal adjacent tissue and in non-compromised lymph nodes. Our immunohistochemistry revealed that 59 % of fibroblasts associated with primary tumors and 41 % of the respective metastatic lymph nodes (p = 0.271) displayed positive staining for RhoA. In line with this, 57.1 % of fibroblasts associated with primary tumors presented Rac1-positive staining, and the frequency of co-positivity within the lymph nodes was 42.9 % (p = 0.16). Expression of RhoA and Rac1 was absent in fibroblasts of adjacent normal tissue and in compromised lymph nodes. Based on our findings that no significant changes were observed between primary and metastatic lymph nodes, we suggest that fibroblasts are active participants in the invasion of cancer cells to lymph nodes and support the hypothesis that metastatic tumor cells continue to depend on their microenvironment.


Assuntos
Neoplasias da Mama/genética , Invasividade Neoplásica/genética , Proteínas rac1 de Ligação ao GTP/biossíntese , Proteína rhoA de Ligação ao GTP/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Linfonodos/metabolismo , Linfonodos/patologia , Metástase Linfática , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Proteínas rac1 de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA