Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
FASEB J ; 33(9): 9959-9973, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31199673

RESUMO

VEGF stimulates the formation of new blood vessels by inducing endothelial cell (EC) proliferation and migration. Brefeldin A (BFA)-inhibited guanine nucleotide-exchange protein (BIG)1 and 2 accelerate the replacement of bound GDP with GTP to activate ADP-ribosylation factor (Arf)1, which regulates vesicular transport between the Golgi and plasma membrane. Although it has been reported that treating cells with BFA interferes with Arf1 activation to inhibit VEGF secretion, the role of BIG1 and BIG2 in VEGF trafficking and expression, EC migration and proliferation, and vascular development remains unknown. Here, we found that inactivation of Arf1 reduced VEGF secretion but did not affect the levels of VEGF protein. Interestingly, however, BIG1 and BIG2 knockdown significantly decreased the levels of VEGF mRNA and protein in glioblastoma U251 cells and HUVECs. Furthermore, depletion of BIG1 and BIG2 inhibited HUVEC angiogenesis by diminishing cell migration. Angioblast migration and intersegmental vessel sprouting were also impaired when the BIG2 homolog, Arf guanine nucleotide exchange factor (arfgef)2, was knocked down in zebrafish with endothelial expression of green fluorescent protein (GFP). Depletion of arfgef2 by clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) also caused defects in vascular development of zebrafish embryos. Taken together, these data reveal that BIG1 and BIG2 participate in endothelial cell angiogenesis.-Lu, F.-I., Wang, Y.-T., Wang, Y.-S., Wu, C.-Y., Li, C.-C. Involvement of BIG1 and BIG2 in regulating VEGF expression and angiogenesis.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/fisiologia , Neovascularização Fisiológica/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator 1 de Ribosilação do ADP/antagonistas & inibidores , Fator 1 de Ribosilação do ADP/fisiologia , Animais , Sistemas CRISPR-Cas , Movimento Celular , Embrião não Mamífero/irrigação sanguínea , Desenvolvimento Embrionário , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Técnicas de Silenciamento de Genes , Genes Reporter , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Neovascularização Fisiológica/genética , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
2.
Nat Chem Biol ; 15(4): 358-366, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30742123

RESUMO

Peripheral membrane proteins orchestrate many physiological and pathological processes, making regulation of their activities by small molecules highly desirable. However, they are often refractory to classical competitive inhibition. Here, we demonstrate that potent and selective inhibition of peripheral membrane proteins can be achieved by small molecules that target protein-membrane interactions by a noncompetitive mechanism. We show that the small molecule Bragsin inhibits BRAG2-mediated Arf GTPase activation in vitro in a manner that requires a membrane. In cells, Bragsin affects the trans-Golgi network in a BRAG2- and Arf-dependent manner. The crystal structure of the BRAG2-Bragsin complex and structure-activity relationship analysis reveal that Bragsin binds at the interface between the PH domain of BRAG2 and the lipid bilayer to render BRAG2 unable to activate lipidated Arf. Finally, Bragsin affects tumorsphere formation in breast cancer cell lines. Bragsin thus pioneers a novel class of drugs that function by altering protein-membrane interactions without disruption.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Fator 1 de Ribosilação do ADP/metabolismo , Linhagem Celular Tumoral , GTP Fosfo-Hidrolases , Proteínas Ativadoras de GTPase , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Células HeLa , Humanos , Bicamadas Lipídicas , Glicoproteínas de Membrana/metabolismo , Nucleotídeos , Domínios de Homologia à Plecstrina/fisiologia , Ligação Proteica , Transdução de Sinais , Relação Estrutura-Atividade , Sulfotransferases/metabolismo
3.
Neuro Oncol ; 19(1): 22-30, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27402815

RESUMO

BACKGROUND: Astrocytomas are the most common primary human brain tumors. Receptor tyrosine kinases (RTKs), including tyrosine receptor kinase B (TrkB, also known as tropomyosin-related kinase B; encoded by neurotrophic tyrosine kinase receptor type 2 [NTRK2]), are frequently mutated by rearrangement/fusion in high-grade and low-grade astrocytomas. We found that activated TrkB can contribute to the development of astrocytoma and might serve as a therapeutic target in this tumor type. METHODS: To identify RTKs capable of inducing astrocytoma formation, a library of human tyrosine kinases was screened for the ability to transform murine Ink4a-/-/Arf-/- astrocytes. Orthotopic allograft studies were conducted to evaluate the effects of RTKs on the development of astrocytoma. Since TrkB was identified as a driver of astrocytoma formation, the effect of the Trk inhibitors AZD1480 and RXDX-101 was assessed in astrocytoma cells expressing activated TrkB. RNA sequencing, real-time PCR, western blotting, and enzyme-linked immunosorbent assays were conducted to characterize NTRK2 in astrocytomas. RESULTS: Activated TrkB cooperated with Ink4a/Arf loss to induce the formation of astrocytomas through a mechanism mediated by activation of signal transducer and activator of transcription 3 (STAT3). TrkB activation positively correlated with Ccl2 expression. TrkB-induced astrocytomas remained dependent on TrkB signaling for survival, highlighting a role of NTRK2 as an addictive oncogene. Furthermore, the QKI-NTRK2 fusion associated with human astrocytoma transformed Ink4a-/-/Arf-/- astrocytes, and this process was also mediated via STAT3 signaling. CONCLUSIONS: Our findings provide evidence that constitutively activated NTRK2 alleles, notably the human tumor-associated QKI-NTRK2 fusion, can cooperate with Ink4a/Arf loss to drive astrocytoma formation. Therefore, we propose NTRK2 as a potential therapeutic target in the subset of astrocytoma patients defined by QKI-NTRK2 fusion.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Astrócitos/patologia , Astrocitoma/patologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptor trkB/metabolismo , Animais , Astrócitos/enzimologia , Astrocitoma/enzimologia , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Camundongos Knockout , Transdução de Sinais
5.
Cell Microbiol ; 18(8): 1121-33, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26814617

RESUMO

GBF1 is a host factor required for hepatitis C virus (HCV) replication. GBF1 functions as a guanine nucleotide exchange factor for G-proteins of the Arf family, which regulate membrane dynamics in the early secretory pathway and the metabolism of cytoplasmic lipid droplets. Here we established that the Arf-guanine nucleotide exchange factor activity of GBF1 is critical for its function in HCV replication, indicating that it promotes viral replication by activating one or more Arf family members. Arf involvement was confirmed with the use of two dominant negative Arf1 mutants. However, siRNA-mediated depletion of Arf1, Arf3 (class I Arfs), Arf4 or Arf5 (class II Arfs), which potentially interact with GBF1, did not significantly inhibit HCV infection. In contrast, the simultaneous depletion of both Arf4 and Arf5, but not of any other Arf pair, imposed a significant inhibition of HCV infection. Interestingly, the simultaneous depletion of both Arf4 and Arf5 had no impact on the activity of the secretory pathway and induced a compaction of the Golgi and an accumulation of lipid droplets. A similar phenotype of lipid droplet accumulation was also observed when GBF1 was inhibited by brefeldin A. In contrast, the simultaneous depletion of both Arf1 and Arf4 resulted in secretion inhibition and Golgi scattering, two actions reminiscent of GBF1 inhibition. We conclude that GBF1 could regulate different metabolic pathways through the activation of different pairs of Arf proteins.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Hepacivirus/fisiologia , Hepatite C/virologia , Replicação Viral , Linhagem Celular Tumoral , Hepatite C/enzimologia , Interações Hospedeiro-Patógeno , Humanos , Gotículas Lipídicas , Domínios Proteicos , Transporte Proteico , Via Secretória
6.
Int J Mol Sci ; 16(12): 29305-14, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26690137

RESUMO

Small G-protein adenosine diphosphate (ADP)-ribosylation factors (ARFs) regulate a variety of cellular functions, including actin cytoskeleton remodeling, plasma membrane reorganization, and vesicular transport. Here, we propose the functional roles of ARF1 in multiple stages of osteoclast differentiation. ARF1 was upregulated during receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation and transiently activated in an initial stage of their differentiation. Differentiation of ARF1-deficient osteoclast precursors into mature osteoclasts temporarily increased in pre-maturation stage of osteoclasts followed by reduced formation of mature osteoclasts, indicating that ARF1 regulates the osteoclastogenic process. ARF1 deficiency resulted in reduced osteoclast precursor proliferation and migration as well as increasing cell-cell fusion. In addition, ARF1 silencing downregulated c-Jun N-terminal kinase (JNK), Akt, osteopontin, and macrophage colony-stimulating factor (M-CSF)-receptor c-Fms as well as upregulating several fusion-related genes including CD44, CD47, E-cadherin, and meltrin-α. Collectively, we showed that ARF1 stimulated proliferation and migration of osteoclast precursors while suppressing their fusion, suggesting that ARF1 may be a plausible inter-player that mediates the transition to osteoclast fusion at multiple steps during osteoclast differentiation.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Osteoclastos/fisiologia , Animais , Fusão Celular , Células Cultivadas , Masculino , Camundongos Endogâmicos C57BL , Transdução de Sinais
7.
Cell Signal ; 27(10): 2035-2044, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26169956

RESUMO

Mitogen-activated protein kinases (MAPK) can be activated by a number of biochemical pathways through distinct signaling molecules. We have recently revealed a novel function for the Ras-like small GTPase ADP-ribosylation factor 1 (ARF1) in mediating the activation of Raf1-MEK-ERK1/2 pathway by G protein-coupled receptors [Dong C, Li C and Wu G (2011) J Biol Chem 286, 43,361-43,369]. Here, we have further defined the underlying mechanism and the possible function of ARF1-mediated MAPK pathway. We demonstrated that the blockage of ARF1 activation and the disruption of ARF1 localization to the Golgi by mutating Thr48, a highly conserved residue involved in the exchange of GDP for GTP, and the myristoylation site Gly2 abolished ARF1's ability to activate ERK1/2. In addition, treatment with Golgi structure disrupting agents markedly attenuated ARF1-mediated ERK1/2 activation. Furthermore, ARF1 significantly promoted cell proliferation. More interestingly, ARF1 activated 90kDa ribosomal S6 kinase 1 (RSK1) without influencing Elk-1 activation and ERK2 translocation to the nuclei. These data demonstrate that, once activated, ARF1 activates the MAPK pathway likely using the Golgi as a main platform, which in turn activates the cytoplasmic RSK1, leading to cell proliferation.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sequência de Aminoácidos , Proliferação de Células , Ativação Enzimática , Complexo de Golgi/enzimologia , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Transporte Proteico , Receptor A2B de Adenosina/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Proteínas Elk-1 do Domínio ets/metabolismo
8.
Cancer Prev Res (Phila) ; 8(6): 492-501, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25813526

RESUMO

Loss of parietal cells initiates the development of spasmolytic polypeptide-expressing metaplasia (SPEM), a precancerous lesion in stomach. CD44 variant (CD44v) that enhances the ability to defend against reactive oxygen species (ROS) in epithelial cells is expressed de novo in SPEM of K19-Wnt1/C2mE mice, a transgenic model of gastric tumorigenesis, and is required for the efficient development of SPEM and gastric tumor in these animals. The role of ROS and its downstream signaling in CD44-dependent gastric tumorigenesis has remained unknown, however. With the use of the K19-Wnt1/C2mE mouse, we now show that parietal cells in the inflamed stomach are highly sensitive to oxidative stress and manifest activation of p38(MAPK) signaling by ROS. Oral treatment with the antioxidant ascorbic acid or genetic ablation of the Ink4a/Arf locus, a major downstream target of ROS-p38(MAPK) signaling, inhibited parietal cell loss and the subsequent gastric tumorigenesis. Our results indicate that signaling activated by oxidative stress in parietal cells plays a key role in CD44-dependent gastric tumorigenesis. .


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Transformação Celular Neoplásica/patologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Receptores de Hialuronatos/metabolismo , Estresse Oxidativo , Células Parietais Gástricas/patologia , Neoplasias Gástricas/patologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Técnicas Imunoenzimáticas , Metaplasia/metabolismo , Metaplasia/patologia , Camundongos , Camundongos Knockout , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células Parietais Gástricas/metabolismo , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Proteína Wnt1/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Blood ; 125(9): 1444-51, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25499760

RESUMO

Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) is initiated and driven by the oncogenic fusion protein BCR-ABL, a constitutively active tyrosine kinase. Despite major advances in the treatment of this highly aggressive disease with potent inhibitors of the BCR-ABL kinase such as dasatinib, patients in remission frequently relapse due to persistent minimal residual disease possibly supported, at least in part, by salutary cytokine-driven signaling within the hematopoietic microenvironment. Using a mouse model of Ph+ ALL that accurately mimics the genetics, clinical behavior, and therapeutic response of the human disease, we show that a combination of 2 agents approved by the US Food and Drug Administration (dasatinib and ruxolitinib, which inhibit BCR-ABL and Janus kinases, respectively), significantly extends survival by targeting parallel signaling pathways. Although the BCR-ABL kinase cancels the cytokine requirement of immature leukemic B cells, dasatinib therapy restores cytokine dependency and sensitizes leukemic cells to ruxolitinib. As predicted, ruxolitinib alone had no significant antileukemic effect in this model, but it prevented relapse when administered with dasatinib. The combination of dasatinib, ruxolitinib, and the corticosteroid dexamethasone yielded more durable remissions, in some cases after completion of therapy, avoiding the potential toxicity of other cytotoxic chemotherapeutic agents.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Janus Quinases/antagonistas & inibidores , Recidiva Local de Neoplasia/tratamento farmacológico , Cromossomo Filadélfia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Fator 1 de Ribosilação do ADP/fisiologia , Animais , Western Blotting , Dasatinibe , Dexametasona/administração & dosagem , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Proteínas de Fusão bcr-abl/genética , Humanos , Interleucina-7/genética , Interleucina-7/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Nitrilas , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Indução de Remissão , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Taxa de Sobrevida , Tiazóis/administração & dosagem , Células Tumorais Cultivadas
10.
Mol Biol Cell ; 25(1): 17-29, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24196838

RESUMO

Invasion of tumor cells is a key step in metastasis that depends largely on the ability of these cells to degrade the extracellular matrix. Although we have showed that the GTPase ADP-ribosylation factor 1 (ARF1) is overexpressed in highly invasive breast cancer cell lines and that epidermal growth factor stimulation can activate this ARF isoform to regulate migration as well as proliferation, the role of this small GTP-binding protein has not been addressed in the context of invasiveness. Here we report that modulation of ARF1 expression and activity markedly impaired the ability of M.D. Anderson-metastatic breast-231 cells, a prototypical highly invasive breast cancer cell line, to degrade the extracellular matrix by controlling metalloproteinase-9 activity. In addition, we demonstrate that this occurs through inhibition of invadopodia maturation and shedding of membrane-derived microvesicles, the two key structures involved in invasion. To further define the molecular mechanisms by which ARF1 controls invasiveness, we show that ARF1 acts to modulate RhoA and RhoC activity, which in turn affects myosin light-chain (MLC) phosphorylation. Together our findings underscore for the first time a key role for ARF1 in invasion of breast cancer cells and suggest that targeting the ARF/Rho/MLC signaling axis might be a promising strategy to inhibit invasiveness and metastasis.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Neoplasias da Mama/enzimologia , Cadeias Leves de Miosina/metabolismo , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Extensões da Superfície Celular/metabolismo , Micropartículas Derivadas de Células/metabolismo , Fator de Crescimento Epidérmico/fisiologia , Receptores ErbB/metabolismo , Feminino , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Invasividade Neoplásica , Neoplasias Hormônio-Dependentes/enzimologia , Neoplasias Hormônio-Dependentes/patologia
11.
Blood ; 119(3): 717-26, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-22101896

RESUMO

Alterations of the BM microenvironment have been shown to occur after chemoradiotherapy, during aging, and after genetic manipulations of telomere length. Nevertheless, whether BM stromal cells adopt senescent features in response to these events is unknown. In the present study, we provide evidence that exposure to ionizing radiation (IR) leads murine stromal BM cells to express senescence markers, namely senescence-associated ß-galactosidase and increased p16(INK4a)/p19(ARF) expression. Long (8 weeks) after exposure of mice to IR, we observed a reduction in the number of stromal cells derived from BM aspirates, an effect that we found to be absent in irradiated Ink4a/arf-knockout mice and to be mostly independent of the CFU potential of the stroma. Such a reduction in the number of BM stromal cells was specific, because stromal cells isolated from collagenase-treated bones were not reduced after IR. Surprisingly, we found that exposure to IR leads to a cellular nonautonomous and Ink4a/arf-dependent effect on lymphopoiesis. Overall, our results reveal the distinct sensitivity of BM stromal cell populations to IR and suggest that long-term residual damage to the BM microenvironment can influence hematopoiesis in an Ink4a/arf-dependent manner.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Medula Óssea/efeitos da radiação , Senescência Celular/efeitos da radiação , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Homeostase/efeitos da radiação , Radiação Ionizante , Células Estromais/efeitos da radiação , Animais , Apoptose , Western Blotting , Medula Óssea/metabolismo , Medula Óssea/patologia , Diferenciação Celular , Proliferação de Células , Feminino , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/efeitos da radiação , Linfopoese/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Células Estromais/metabolismo , Células Estromais/patologia
12.
Neuro Oncol ; 13(12): 1277-87, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21926087

RESUMO

Faithful replication and DNA repair are vital for maintenance of genome integrity. RAD51 is a central protein in homologous recombination repair and during replication, when it protects and restarts stalled replication forks. Aberrant RAD51 expression occurs in glioma, and high expression has been shown to correlate with prolonged survival. Furthermore, genes involved in DNA damage response (DDR) are mutated or deleted in human glioblastomas, corroborating the importance of proper DNA repair to suppress gliomagenesis. We have analyzed DDR and genomic instability in PDGF-B-induced gliomas and investigated the role of RAD51 in glioma development. We show that PDGF-B-induced gliomas display genomic instability and that co-expression of RAD51 can suppress PDGF-B-induced tumorigenesis and prolong survival. Expression of RAD51 inhibited proliferation and genomic instability of tumor cells independent of Arf status. Our results demonstrate that the RAD51 pathway can prevent glioma initiation and maintain genome integrity of induced tumors, suggesting reactivation of the RAD51 pathway as a potential therapeutic avenue.


Assuntos
Neoplasias Encefálicas/prevenção & controle , Instabilidade Genômica , Glioma/prevenção & controle , Hiperplasia/prevenção & controle , Proteínas Proto-Oncogênicas c-sis/metabolismo , Rad51 Recombinase/metabolismo , Fator 1 de Ribosilação do ADP/fisiologia , Animais , Western Blotting , Encéfalo/citologia , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Galinhas , Dano ao DNA , Reparo do DNA , Replicação do DNA , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Glioma/genética , Glioma/patologia , Proteínas de Fluorescência Verde/genética , Recombinação Homóloga , Humanos , Hiperplasia/genética , Hiperplasia/patologia , Técnicas Imunoenzimáticas , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-sis/genética , Rad51 Recombinase/genética , Células-Tronco/metabolismo
13.
Oncogene ; 30(36): 3846-61, 2011 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-21478909

RESUMO

The ADP-ribosylation factors (ARFs) 1 and 6 are small GTP-binding proteins, highly expressed and activated in several breast cancer cell lines and are associated with enhanced migration and invasiveness. In this study, we report that ARF1 has a critical role in cell proliferation. Depletion of this GTPase or expression of a dominant negative form, which both resulted in diminished ARF1 activity, led to sustained cell-growth arrest. This cellular response was associated with the induction of senescent markers in highly invasive breast cancer cells as well as in control mammary epithelial cells by a mechanism regulating retinoblastoma protein (pRB) function. When examining the role of ARF1, we found that this GTPase was highly activated in normal proliferative conditions, and that a limited amount could be found in the nucleus, associated with the chromatin of MDA-MB-231 cells. However, when cells were arrested in the G(0)/G(1) phase or transfected with a dominant negative form of ARF1, the total level of activated ARF1 was markedly reduced and the GTPase significantly enriched in the chromatin. Using biochemical approaches, we demonstrated that the GDP-bound form of ARF1 directly interacted with pRB, but not other members of this family of proteins. In addition, depletion of ARF1 or expression of ARF1T(31)N resulted in the constitutive association of pRB and E2F1, thereby stabilizing the interaction of E2F1 as well as pRB at endogenous sites of target gene promoters, preventing expression of E2F target genes, such as cyclin D1, Mcm6 and E2F1, important for cell-cycle progression. These novel findings provide direct physiological and molecular evidence for the role of ARF1 in controlling cell proliferation, dependent on its ability to regulate pRB/E2F1 activity and gene expression for enhanced proliferation and breast cancer progression.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica , Retinoblastoma/metabolismo , Animais , Células COS , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Chlorocebus aethiops , Fator de Transcrição E2F1/metabolismo , Feminino , Humanos , Modelos Biológicos , Proteína do Retinoblastoma/metabolismo
14.
Toxicol Lett ; 203(3): 210-8, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21421027

RESUMO

Cadmium (Cd) is nephrotoxic. Circulating Cd-metallothionein complexes (CdMT) are filtered by the kidney, reabsorbed by proximal tubule cells (PTC) via receptor-mediated endocytosis, and trafficked to lysosomes which results in apoptosis. ADP-ribosylation factors (Arfs) regulate vesicular trafficking. Arf1 is traditionally associated with the secretory pathway, but has been recently found involved in endocytotic trafficking in PTC. Hence, the role of Arf1 was investigated in MT-1 and transferrin (Tf) endocytosis, and in CdMT-1-induced cell death in a PTC line by overexpressing Arf1-wildtype (WT) or dominant-negative mutant Arf1-T31N. Endogenous Arf1 distribution in PTC was punctate throughout the cytosol, but was not detected in the plasma membrane. Arf1 colocalized with markers for sorting to late endosomes (Rab7, CLC6). Arf1 weakly overlapped with the late endosomal/lysosomal marker CLC7, but not with markers for early (Rab5, CLC5) and recycling endosomes (Rab11). Arf1-T31N significantly attenuated CdMT-1 toxicity by ∼60% when compared to Arf1-WT. However, overexpression of Arf1-T31N did not prevent internalization of Alexa Fluor 546-coupled Tf or MT-1 which accumulated in an EEA1-positive early endocytotic compartment, but not in Arf1-WT overexpressing cells. We conclude that Arf1 is involved in trafficking of protein-metal complexes, including CdMT, to late endosomes/lysosomes in renal PTC.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Endossomos/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Metalotioneína/toxicidade , Fator 1 de Ribosilação do ADP/análise , Animais , Transporte Biológico , Células Cultivadas , Endocitose , Metalotioneína/metabolismo , Transporte Proteico , Ratos , Transferrina/metabolismo , Proteínas de Transporte Vesicular/análise , Proteínas rab de Ligação ao GTP/análise , proteínas de unión al GTP Rab7
16.
Cancer Prev Res (Phila) ; 2(9): 800-6, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19723896

RESUMO

Despite advances in screening and treatment, colorectal cancer remains the second leading cause of cancer-related death in the United States. Cyclin-dependent kinases (Cdk) are deregulated in colorectal cancer by silencing of the Cdk inhibitor p16(Ink4a) and other mechanisms. We tested whether the small molecule Cdk inhibitor SNS-032 (formerly BMS-387032), which targets Cdk2, Cdk7, and Cdk9, can prevent intestinal tumorigenesis in mouse models. We generated mice with high intestinal tumor loads by combining the multiple intestinal neoplasia (Min) mutation with Ink4a/Arf mutations and inducing colitis with dextran sulfate sodium. p16-null Min mice (n = 17) began dextran sulfate sodium treatment at week 5 and i.p. injection of carrier or SNS-032 at week 6. Mice were sacrificed at week 12. SNS-032 was well tolerated and reduced colon tumor burden to 36% of that in carrier-treated mice (P < 0.001). We then extended the study to Ink4/Arf-null Min mice (n = 14) and increased the drug dose frequency. SNS-032 treatment reduced the intestinal tumor number to 25% and intestinal tumor burden to 16% of carrier-treated mice (P < 0.0001). DNA synthesis in non-neoplastic and tumor epithelial cells, detected by bromodeoxyuridine incorporation, was modestly reduced by acute SNS-032 treatment. The mitotic index, detected by histone H3 phosphorylation, was distinctly decreased (P < 0.03), and apoptosis, detected by caspase 3 activation, was increased (P < 0.005). These results show the chemoprevention of intestinal tumorigenesis by SNS-032. Our findings support further study of Cdk inhibitors for chemoprevention and therapy of colon cancer.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Neoplasias Intestinais/prevenção & controle , Oxazóis/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Tiazóis/uso terapêutico , Fator 1 de Ribosilação do ADP/fisiologia , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Colite/enzimologia , Colite/patologia , Colite/prevenção & controle , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Feminino , Neoplasias Intestinais/enzimologia , Neoplasias Intestinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/efeitos dos fármacos
17.
Autophagy ; 5(3): 397-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19221462

RESUMO

Autophagy plays a critical role in the initiation and progression of tumors. The exact nature of this role, however, is complex. Autophagy is suppressive to tumor initiation, and reduces genomic instability. Genes with key roles in autophagy are mutated in human cancer, and knockout mice for certain autophagy genes are predisposed to cancer. Conversely, established tumors appear to utilize autophagy in order to survive periods of metabolic or hypoxic stress. Consistent with this, small molecule inhibitors of autophagy like chloroquine are effective anticancer agents for certain tumor types. The consensus appears to be that autophagy suppresses tumor initiation, but promotes the survival of established tumors. But this premise may be over-simplified. Several groups have recently shown that the ARF tumor suppressor can induce autophagy. While some groups have found that ARF-mediated autophagy is cytotoxic to tumor cells, we have shown that ARF's autophagy function may promote the survival and progression of certain tumors. We have previously shown that silencing ARF limits autophagy and the development of p53-null lymphomas. In this addendum, we show this is not true for primary p53-null sarcoma cells. Rather, ARF-silencing enhances sarcoma development. These data suggest that the survival-benefit of ARF, and possibly also of autophagy, may be restricted to certain tumor types.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Neoplasias/metabolismo , Fator 1 de Ribosilação do ADP/metabolismo , Animais , Antineoplásicos/farmacologia , Autofagia/genética , Cloroquina/farmacologia , Inativação Gênica , Genes p53 , Humanos , Camundongos , Modelos Biológicos , Transplante de Neoplasias , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
18.
J Biol Chem ; 283(52): 36425-34, 2008 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18990689

RESUMO

Activation of intracellular signaling pathways by growth factors is one of the major causes of cancer development and progression. Recent studies have demonstrated that monomeric G proteins of the Ras family are key regulators of cell proliferation, migration, and invasion. Using an invasive breast cancer cell lines, we demonstrate that the ADP-ribosylation factor 1 (ARF1), a small GTPase classically associated with the Golgi, is an important regulator of the biological effects induced by epidermal growth factor. Here, we show that this ARF isoform is activated following epidermal growth factor stimulation and that, in MDA-MB-231 cells, ARF1 is found in dynamic plasma membrane ruffles. Inhibition of endogenous ARF1 expression results in the inhibition of breast cancer cell migration and proliferation. The underlying mechanism involves the activation of the phosphatidylinositol 3-kinase pathway. Our data demonstrate that depletion of ARF1 markedly impairs the recruitment of the phosphatidylinositol 3-kinase catalytic subunit (p110alpha) to the plasma membrane, and the association of the regulatory subunit (p85alpha) to the activated receptor. These results uncover a novel molecular mechanism by which ARF1 regulates breast cancer cell growth and invasion during cancer progression.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Neoplasias da Mama/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fator 1 de Ribosilação do ADP/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/metabolismo , Catálise , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Classe I de Fosfatidilinositol 3-Quinases , Progressão da Doença , Ativação Enzimática , Complexo de Golgi/metabolismo , Humanos , Microscopia Confocal
19.
Cancer Res ; 67(15): 7350-7, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17671205

RESUMO

Cellular senescence is emerging as an important in vivo anticancer response elicited by multiple stresses, including currently used chemotherapeutic drugs. Nutlin-3a is a recently discovered small-molecule antagonist of the p53-destabilizing protein murine double minute-2 (MDM2) that induces cell cycle arrest and apoptosis in cancer cells with functional p53. Here, we report that nutlin-3a induces cellular senescence in murine primary fibroblasts, oncogenically transformed fibroblasts, and fibrosarcoma cell lines. No evidence of drug-induced apoptosis was observed in any case. Nutlin-induced senescence was strictly dependent on the presence of functional p53 as revealed by the fact that cells lacking p53 were completely insensitive to the drug, whereas cells lacking the tumor suppressor alternative reading frame product of the CDKN2A locus underwent irreversible cell cycle arrest. Interestingly, irreversibility was achieved in neoplastic cells faster than in their corresponding parental primary cells, suggesting that nutlin-3a and oncogenic signaling cooperate in activating p53. Our current results suggest that senescence could be a major cellular outcome of cancer therapy by antagonists of the p53-MDM2 interaction, such as nutlin-3a.


Assuntos
Senescência Celular , Fibroblastos/metabolismo , Fibrossarcoma/patologia , Imidazóis/farmacologia , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Fator 1 de Ribosilação do ADP/genética , Fator 1 de Ribosilação do ADP/fisiologia , Animais , Ciclo Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Fibroblastos/citologia , Fibrossarcoma/metabolismo , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
20.
Curr Med Chem ; 14(17): 1815-27, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17627519

RESUMO

One of the outstanding fundamental questions in cancer cell biology concerns how cells coordinate cellular growth (or macromolecular synthesis) with cell cycle progression and mitosis. Intuitively, rapidly dividing cells must have some control over these processes; otherwise cells would continue to shrink in volume with every passing cycle, similar to the cytoreductive divisions seen in the very early stages of embryogenesis. The problem is easily solved in unicellular organisms, such as yeast, as their growth rates are entirely dependent on nutrient availability. Multicellular organisms such as mammals, however, must have acquired additional levels of control, as nutrient availability is seldom an issue and the organism has a prodigious capacity to store necessary metabolites in the form of glycogen, lipids, and protein. Furthermore, the specific needs and specialized architecture of tissues must constrain growth for growth's sake; if not, the necessary function of the organ could be lost. While certainly a myriad of mechanisms for preventing this exist via initiating cell death (e.g. apoptosis, autophagy, necrosis), these all depend on some external cue, such as death signals, hypoxia, lack of nutrients or survival signals. However there must also be some cell autonomous method for surveying against inappropriate growth signals (such as oncogenic stress) that occur in a stochastic fashion, possibly as a result of random mutations. The ARF tumor suppressor seems to fulfill that role, as its expression is near undetectable in normal tissues, yet is potently induced by oncogenic stress (such as overexpression of oncogenic Ras or myc). As a result of induced expression of ARF, the tumor suppressor protein p53 is stabilized and promotes cell cycle arrest. Mutations or epigenetic alterations of the INK4a/Arf locus are second only to p53 mutations in cancer cells, and in some cancers, alterations in both Arf and p53 observed, suggesting that these two tumor suppressors act coordinately to prevent unwarranted cell growth and proliferation. The aim of this review is to characterize the current knowledge in the field about both p53-dependent and independent functions of ARF as well as to summarize the present models for how ARF might control rates of cell proliferation and/or macromolecular synthesis. We will discuss potential therapeutic targets in the ARF pathway, and some preliminary attempts at enhancing or restoring the activity of this important tumor suppressor.


Assuntos
Fator 1 de Ribosilação do ADP/efeitos dos fármacos , Fator 1 de Ribosilação do ADP/fisiologia , Antineoplásicos/farmacologia , Genes Supressores de Tumor/efeitos dos fármacos , Genes Supressores de Tumor/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator 1 de Ribosilação do ADP/genética , Animais , Nucléolo Celular/genética , Nucléolo Celular/fisiologia , Genes p53/fisiologia , Humanos , Neoplasias/genética , Neoplasias/fisiopatologia , Peptídeos/síntese química , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Ribossomos/fisiologia , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA