Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Obstet Gynecol ; 140(5): 758-767, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36201776

RESUMO

OBJECTIVE: To describe the testing rate, patient characteristics, temporal trends, timing, and results of germline and somatic BRCA testing in patients with ovarian cancer using real-world data. METHODS: We included a cross-sectional subset of adult patients diagnosed with ovarian cancer between January 1, 2011, and November 30, 2018, who received frontline treatment and were followed for at least 1 year in a real-world database. The primary outcome was receipt of BRCA testing, classified by biosample source as germline (blood or saliva) or somatic (tissue). Lines of therapy (frontline, second line, third line) were derived based on dates of surgery and chemotherapy. Descriptive statistics were analyzed. RESULTS: Among 2,557 patients, 72.2% (n=1,846) had at least one documented BRCA test. Among tested patients, 62.5% (n=1,154) had only germline testing, 10.6% (n=197) had only somatic testing, and 19.9% (n=368) had both. Most patients had testing before (9.7%, n=276) or during (48.6%, n=1,521) frontline therapy, with 17.6% (n=273) tested during second-line and 12.7% (n=129) tested during third-line therapy. Patients who received BRCA testing, compared with patients without testing, were younger (mean age 63 years vs 66 years, P <.001) and were more likely to be treated at an academic practice (10.4% vs 7.0%, P =.01), with differences by Eastern Cooperative Oncology Group performance score ( P <.001), stage of disease ( P <.001), histology ( P <.001), geography ( P <.001), and type of frontline therapy ( P <.001), but no differences based on race or ethnicity. The proportion of patients who received BRCA testing within 1 year of diagnosis increased from 24.6% of patients in 2011 to 75.6% of patients in 2018. CONCLUSION: In a large cohort of patients with ovarian cancer, significant practice disparities existed in testing for actionable BRCA mutations. Despite increased testing over time, many patients did not receive testing, suggesting missed opportunities to identify patients appropriate for targeted therapy and genetic counseling.


Assuntos
Proteína BRCA1 , Neoplasias Ovarianas , Humanos , Adulto , Feminino , Pessoa de Meia-Idade , Proteína BRCA1/genética , Estudos Transversais , Testes Genéticos/métodos , Carcinoma Epitelial do Ovário/genética , Neoplasias Ovarianas/patologia , Células Germinativas/patologia
2.
J Surg Oncol ; 120(4): 786-793, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31368160

RESUMO

BACKGROUND AND OBJECTIVES: The single-arm ROSiA study evaluated frontline bevacizumab for advanced ovarian cancer. We explored how discordant surgically and radiologically assessed postoperative residual disease affects outcomes. METHODS: After debulking surgery, 1021 patients received 4 to 8 cycles of carboplatin-paclitaxel plus bevacizumab until progression or up to 24 months. The primary endpoint was safety; progression-free survival (PFS) was a secondary endpoint. We performed post hoc exploratory PFS analyses in four subgroups: surgeon-reported no visible residuum (NVR) without target lesions; surgeon-reported NVR with target lesions; macroscopic (≤1 cm) residuum; and >1 cm residuum. RESULTS: Surgical and radiological assessments were concordant in 94% of patients; 61 patients (6%; 21% of those with surgeon-reported NVR) had NVR with target lesions. Median PFS was numerically longest in patients with concordant surgically/radiologically assessed NVR (35.5 months), intermediate for surgeon-reported NVR with target lesions (31.8 months), and shortest for visible residuum (27.9 and 20.2 months for visible residuum ≤1 and >1 cm, respectively). One-year and 2-year PFS rates showed the same pattern. CONCLUSIONS: These analyses suggest that prognosis is potentially worse in patients with radiologically detected target lesions despite surgeon-reported NVR compared with concordant NVR by both assessment methods. Postsurgical imaging may add valuable prognostic information.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bevacizumab/uso terapêutico , Neoplasia Residual/mortalidade , Neoplasias Ovarianas/mortalidade , Cirurgiões/estatística & dados numéricos , Tomografia Computadorizada por Raios X/métodos , Adenocarcinoma de Células Claras/diagnóstico por imagem , Adenocarcinoma de Células Claras/mortalidade , Adenocarcinoma de Células Claras/patologia , Antineoplásicos Imunológicos/uso terapêutico , Carcinossarcoma/diagnóstico por imagem , Carcinossarcoma/mortalidade , Carcinossarcoma/patologia , Feminino , Seguimentos , Humanos , Neoplasia Residual/diagnóstico por imagem , Neoplasia Residual/patologia , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/patologia , Prognóstico , Taxa de Sobrevida
3.
Oncotarget ; 8(58): 98371-98383, 2017 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-29228696

RESUMO

BACKGROUND: The VEGF/VEGFR and the HGF/cMET pathways are key mediators of the interplay of tumor cells and their microenvironment. However, inhibition of VEGF has been shown to produce only limited clinical benefit and inhibition of the activation of cMET by HGF has not translated into clinical benefit in pivotal trials. MP0250, a DARPin® molecule that specifically inhibits both VEGF and HGF has been developed to explore the clinical potential of dual inhibition of these pathways. RESULTS: MP0250 binding to VEGF and HGF inhibited downstream signalling through VEGFR2 and cMET resulting in inhibition of proliferation of VEGF- and HGF-dependent cells. Antitumor activity was demonstrated in VEGF- and HGF-dependent xenograft and syngeneic models with activity superior to that of individual VEGF- and HGF-blocking DARPin® molecules. Combination therapy studies showed potentiation of the antitumor activity of chemotherapy and immunotherapy agents, including an anti-PD1 antibody. MATERIALS AND METHODS: Potency of MP0250 was assessed in cellular models and in a variety of xenograft models as monotherapy or in combination with standard-of-care drugs. CONCLUSIONS: Dual inhibition of VEGF and HGF by MP0250 produced powerful single agent and combination antitumor activity. This, together with increasing understanding of the role of the HGF/cMET pathway in resistance to VEGF (and other agents), supports testing of MP0250 in the clinic.

4.
MAbs ; 9(8): 1262-1269, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29035637

RESUMO

MP0250 is a multi-domain drug candidate currently being tested in clinical trials for the treatment of cancer. It comprises one anti-vascular endothelial growth factor-A (VEGF-A), one anti-hepatocyte growth factor (HGF), and two anti-human serum albumin (HSA) DARPin® domains within a single polypeptide chain. While there is first clinical validation of a single-domain DARPin® drug candidate, little is known about DARPin® drug candidates comprising multiple domains. Here, we show that MP0250 can be expressed at 15 g/L in soluble form in E. coli high cell-density fermentation, it is stable in soluble/frozen formulation for 2 years as assessed by reverse phase HPLC, it has picomolar potency in inhibiting VEGF-A and HGF in ELISA and cellular assays, and its domains are simultaneously active as shown by surface plasmon resonance. The inclusion of HSA-binding DARPin® domains leads to a favorable pharmacokinetic profile in mouse and cynomolgus monkey, with terminal half-lives of ∼ 30 hours in mouse and ∼ 5 days in cynomolgus monkey. MP0250 is thus a highly potent drug candidate that could be particularly useful in oncology. Beyond MP0250, the properties of MP0250 indicate that multi-domain DARPin® proteins can be valuable next-generation drug candidates.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos/imunologia , Antineoplásicos/imunologia , Proteínas Recombinantes de Fusão/imunologia , Administração Intravenosa , Animais , Repetição de Anquirina/genética , Repetição de Anquirina/imunologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Desenho de Fármacos , Feminino , Meia-Vida , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/imunologia , Humanos , Infusões Intravenosas , Macaca fascicularis , Masculino , Camundongos Endogâmicos BALB C , Ligação Proteica/imunologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacocinética , Albumina Sérica Humana/genética , Albumina Sérica Humana/imunologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia
5.
Nat Cell Biol ; 16(7): 685-94, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24880666

RESUMO

The mechanisms that allow colon cancer cells to form liver and lung metastases, and whether KRAS mutation influences where and when metastasis occurs, are unknown. We provide clinical and molecular evidence showing that different MAPK signalling pathways are implicated in this process. Whereas ERK2 activation provides colon cancer cells with the ability to seed and colonize the liver, reduced p38 MAPK signalling endows cancer cells with the ability to form lung metastasis from previously established liver lesions. Downregulation of p38 MAPK signalling results in increased expression of the cytokine PTHLH, which contributes to colon cancer cell extravasation to the lung by inducing caspase-independent death in endothelial cells of the lung microvasculature. The concerted acquisition of metastatic traits in the colon cancer cells together with the sequential colonization of liver and lung highlights the importance of metastatic lesions as a platform for further dissemination.


Assuntos
Neoplasias do Colo/patologia , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Metástase Neoplásica , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Células Cultivadas , Neoplasias do Colo/fisiopatologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Humanos , Camundongos , Mutação , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas ras/genética , Proteínas ras/metabolismo
6.
EMBO Mol Med ; 5(11): 1759-74, 2013 11.
Artigo em Inglês | MEDLINE | ID: mdl-24115572

RESUMO

The p38 MAPK pathway is an important regulator of many cellular responses. It is well established that p38 MAPK signalling negatively regulates epithelial cell transformation, but enhanced p38 MAPK activity has been also correlated with bad clinical prognosis in some tumour types. Here, we provide genetic and pharmacological evidence showing that p38 MAPK inhibition cooperates with the chemotherapeutic agent cisplatin to kill tumour cells. We show that p38 MAPK inhibition results in ROS upregulation, which in turn activates the JNK pathway via inactivation of phosphatases, sensitizing human tumour cells to cisplatin-induced apoptosis. Using a mouse model for breast cancer, we confirm that inhibition of p38 MAPK cooperates with cisplatin treatment to reduce tumour size and malignancy in vivo. Taken together, our results illustrate a new function of p38 MAPK that helps tumour cells to survive chemotherapeutic drug treatments, and reveal that the combination of p38 MAPK inhibitors with cisplatin can be potentially exploited for cancer therapy.


Assuntos
Apoptose/efeitos dos fármacos , Cisplatino/administração & dosagem , MAP Quinase Quinase 4/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , MAP Quinase Quinase 4/genética , Camundongos , Camundongos Transgênicos , Neoplasias/genética , Neoplasias/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/genética
7.
Biochem J ; 434(3): 549-58, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21226672

RESUMO

p38α MAPK (mitogen-activated protein kinase) plays an important tumour suppressor role, which is mediated by both its negative effect on cell proliferation and its pro-apoptotic activity. Surprisingly, most tumour suppressor mechanisms co-ordinated by p38α have been reported to occur at the post-translational level. This contrasts with the important role of p38α in the regulation of transcription and the profound changes in gene expression that normally occur during tumorigenesis. We have analysed whole-genome expression profiles of Ras-transformed wild-type and p38α-deficient cells and have identified 202 genes that are potentially regulated by p38α in transformed cells. Expression analysis has confirmed the regulation of these genes by p38α in tumours, and functional validation has identified several of them as probable mediators of the tumour suppressor effect of p38α on Ras-induced transformation. Interestingly, approx. 10% of the genes that are negatively regulated by p38α in transformed cells contribute to EGF (epidermal growth factor) receptor signalling. Our results suggest that inhibition of EGF receptor signalling by transcriptional targets of p38α is an important function of this signalling pathway in the context of tumour suppression.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Transcrição Gênica , Animais , Proliferação de Células , Transformação Celular Neoplásica/patologia , Células Cultivadas , Receptores ErbB/fisiologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Perfilação da Expressão Gênica , Genes ras , Humanos , Camundongos , Camundongos Nus , Proteína Quinase 14 Ativada por Mitógeno/genética , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
8.
Mol Cell Biol ; 29(12): 3332-43, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19364817

RESUMO

Contact inhibition is a fundamental process in multicellular organisms aimed at inhibiting proliferation at high cellular densities through poorly characterized intracellular signals, despite availability of growth factors. We have previously identified the protein kinase p38alpha as a novel regulator of contact inhibition, as p38alpha is activated upon cell-cell contacts and p38alpha-deficient cells are impaired in both confluence-induced proliferation arrest and p27(Kip1) accumulation. Here, we establish that p27(Kip1) plays a key role downstream of p38alpha to arrest proliferation at high cellular densities. Surprisingly, p38alpha does not directly regulate p27(Kip1) expression levels but leads indirectly to confluent upregulation of p27(Kip1) and cell cycle arrest via the inhibition of mitogenic signals originating from the epidermal growth factor receptor (EGFR). Hence, confluent activation of p38alpha uncouples cell proliferation from mitogenic stimulation by inducing EGFR degradation through downregulation of the EGFR-stabilizing protein Sprouty2 (Spry2). Accordingly, confluent p38alpha-deficient cells fail to downregulate Spry2, providing them in turn with sustained EGFR signaling that facilitates cell overgrowth and oncogenic transformation. Our results provide novel mechanistic insight into the role of p38alpha as a sensor of cell density, which induces confluent cell cycle arrest via the Spry2-EGFR-p27(Kip1) network.


Assuntos
Inibição de Contato/fisiologia , Receptores ErbB/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Animais , Sequência de Bases , Contagem de Células , Ciclo Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/deficiência , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Primers do DNA/genética , Regulação para Baixo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana , Camundongos , Camundongos Knockout , Proteína Quinase 14 Ativada por Mitógeno/deficiência , Modelos Biológicos , Células NIH 3T3 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais
9.
Cancer Cell ; 14(6): 427-9, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-19061832

RESUMO

AKT, a protein kinase frequently hyperactivated in cancer, plays an important role in cell survival and contributes to tumor cell resistance to cytotoxic therapies. A new study in this issue of Cancer Cell shows that AKT also induces the accumulation of oxygen radicals, which can be exploited to selectively kill cancer cells containing high levels of AKT activity.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias/metabolismo , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose , Sobrevivência Celular , Humanos , Modelos Biológicos , Transdução de Sinais
10.
EMBO J ; 26(8): 2115-26, 2007 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-17380123

RESUMO

The p38 mitogen-activated protein kinase (MAPK) signaling pathway plays an important role in stress-induced cell-fate decisions by orchestrating responses that go from cell-cycle arrest to apoptosis. We have identified a new p38 MAPK-regulated protein that we named p18(Hamlet), which becomes stabilized and accumulates in response to certain genotoxic stresses such as UV or cisplatin treatment. Overexpression of p18(Hamlet) is sufficient to induce apoptosis, whereas its downregulation reduces the apoptotic response to these DNA damage-inducing agents. We show that p18(Hamlet) interacts with p53 and stimulates the transcription of several proapoptotic p53 target genes such as PUMA and NOXA. This correlates with enhanced p18(Hamlet)-induced recruitment of p53 to the promoters. In proliferating cells, low steady-state levels of p18(Hamlet) are probably maintained by a p53-dependent negative feedback loop. Therefore, p18(Hamlet) is a new cell-fate regulator that links the p38 MAPK and p53 pathways and contributes to the establishment of p53-regulated stress responses.


Assuntos
Apoptose/fisiologia , Ciclo Celular/fisiologia , Dano ao DNA , Regulação da Expressão Gênica/fisiologia , Transativadores/genética , Transativadores/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Sequência de Aminoácidos , Proteínas Reguladoras de Apoptose/metabolismo , Northern Blotting , Western Blotting , Imunoprecipitação da Cromatina , Análise por Conglomerados , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Regulação da Expressão Gênica/genética , Humanos , Luciferases , Dados de Sequência Molecular , Filogenia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Cancer Cell ; 11(2): 191-205, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17292829

RESUMO

p38alpha is a stress-activated protein kinase that negatively regulates malignant transformation induced by oncogenic H-Ras, although the mechanisms involved are not fully understood. Here, we show that p38alpha is not a general inhibitor of oncogenic signaling, but that it specifically modulates transformation induced by oncogenes that produce reactive oxygen species (ROS). This inhibitory effect is due to the ROS-induced activation of p38alpha early in the process of transformation, which induces apoptosis and prevents the accumulation of ROS and their carcinogenic effects. Accordingly, highly tumorigenic cancer cell lines have developed a mechanism to uncouple p38alpha activation from ROS production. Our results indicate that oxidative stress sensing plays a key role in the inhibition of tumor initiation by p38alpha.


Assuntos
Transformação Celular Neoplásica , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Neoplasias/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Sequência de Aminoácidos , Animais , Apoptose , Células Cultivadas , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/fisiologia , Genes ras/genética , Glutationa Transferase/metabolismo , Humanos , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , NADPH Oxidases/metabolismo , Neoplasias/patologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo
12.
Cell Signal ; 18(11): 1897-905, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16574378

RESUMO

Cell migration is critical for many processes, such as angiogenesis, inflammation, development and wound healing, and is also involved in tumour progression and metastasis. Here we show that CXCL12, complement factor 5a (C5a), hepatocyte growth factor (HGF) and platelet-derived growth factor (PDGF)-BB, which stimulate cell migration, also activate p38alpha MAPK. Pharmacological inhibition of this protein kinase with SB 203580 or BIRB 0796, or the genetic ablation of p38alpha MAPK, blocked cell migration induced by the aforementioned chemo-attractants. Macrophages from mice lacking one or more of the other p38 MAPK isoforms showed normal cell migration in response to C5a. We also show that the activation of p38alpha MAPK in response to CXCL12 requires the p21-activated protein kinases (PAK)-1 and PAK-2. MAPKAP-K2 is a protein kinase that is activated by p38alpha MAPK. Reducing its expression using RNA interference blocked CXCL12-induced HeLa cell migration, while macrophages from mice that do not express MAPKAP-K2 failed to migrate in response to C5a. Moreover, RNA interference against the small heat shock protein 27 (HSP27), a physiological substrate of MAPKAP-K2, blocked the CXCL12-induced cell migration. These results demonstrate a general and essential role of the PAK-p38alpha MAPK-MAPKAP-K2-HSP27 signalling pathway in mediating the effects of chemotactic stimuli on cell migration.


Assuntos
Movimento Celular/fisiologia , Quimiocinas CXC/metabolismo , Complemento C5a/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Células Cultivadas , Quimiocina CXCL12 , Fibroblastos/metabolismo , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Modelos Biológicos , Transdução de Sinais/fisiologia , Quinases Ativadas por p21
13.
Proteomics ; 6 Suppl 1: S262-71, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16534743

RESUMO

Oncogenic Ras signaling has been long known to play an important role in tumorigenesis and human cancer. In this report, we have used the sensitive 2-D-DIGE coupled to MS for the identification of proteins differentially expressed at the cell membrane level between oncogenic H-RasV12-transformed wild-type and p38alpha-deficient mouse embryo fibroblasts (MEFs). Following trifluoroethanol solubilization, 76 proteins were found to be differentially regulated. After PMF, 63 spots containing 42 different proteins were unequivocally identified by MALDI-TOF MS coupled with database interrogation. As expected, many of them were membrane proteins. Six proteins were selected for further validation studies based on their potential functional link with malignant transformation and signal transduction. These were prohibitin (PHB), protein disulfide isomerase 3 (PDIA3), focal adhesion kinase 2 (FAK2), c-GMP dependent protein kinase 2 (KGP2), NADH-ubiquinone oxidoreductase 30 kDa subunit (NUGM) and translationally controlled tumor protein (TCTP). All these proteins were up-regulated in the membranes of H-RasV12-transformed p38alpha-/-cells, except for prohibitin, which was down-regulated. An excellent correlation was found between DIGE results and Western blot studies, indicating the reliability of the 2-D-DIGE analysis. The available evidence about the putative function of the identified proteins supports the emerging role of p38alpha as a negative regulator of tumorigenesis. Further studies are in progress to elucidate the implications of these findings in the regulation of H-Ras-induced transformation by p38alpha signaling.


Assuntos
Transformação Celular Neoplásica , Fibroblastos/enzimologia , Genes ras , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Proteômica , Animais , Células Cultivadas , Fibroblastos/patologia , Técnicas de Transferência de Genes , Humanos , Camundongos , Proibitinas , Proteoma/metabolismo , Frações Subcelulares , Proteína Tumoral 1 Controlada por Tradução
14.
Oncogene ; 24(53): 7941-5, 2005 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-16027723

RESUMO

Proliferation of nontransformed cells is regulated by cell-cell contacts, which are referred to as contact-inhibition. Despite its generally accepted importance for cell cycle control, knowledge about the intracellular signalling pathways involved in contact inhibition is scarce. In the present work we show that p38alpha mitogen-activated protein kinase (MAPK) is involved in the growth-inhibitory signalling cascade of contact inhibition in fibroblasts. p38alpha activity is increased in confluent cultures of human fibroblasts compared to proliferating cultures. Time course studies show a sustained activation of p38alpha in response to cell-cell contacts in contrast to a transient activation after serum stimulation. The induction of contact inhibition by addition of glutaraldehyde-fixed cells is impaired by pharmacological inhibition of p38 as well as in p38alpha-/- fibroblasts. Further evidence for a central role of p38alpha in contact inhibition comes from the observation that p38alpha-/- fibroblasts show a higher saturation density compared to wild-type (wt) fibroblasts, which is reversed by reconstituted expression of p38alpha. In agreement with a defect in contact inhibition, p27(Kip1) accumulation is impaired in p38alpha-/- fibroblasts compared to wt fibroblasts. Hence, our work shows a new role for p38alpha in contact inhibition and provides a mechanistic basis for the recently proposed tumour suppressive function of this MAPK pathway.


Assuntos
Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Técnicas de Cultura de Células , Proliferação de Células , Transformação Celular Neoplásica , Inibição de Contato , Fibroblastos/fisiologia , Humanos , Neoplasias/fisiopatologia , Transdução de Sinais
15.
J Comb Chem ; 7(2): 246-52, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15762752

RESUMO

Stabilization of tetrameric transthyretin (TTR) by binding of small ligands is a current strategy aimed at inhibiting amyloid fibrillogenesis in transthyretin-associated pathologies, such as senile systemic amyloidosis (SSA) and familial amyloidotic polyneuropathy (FAP). A kinetic assay is developed for rapid evaluation of compounds as potential in vitro inhibitors in a high-throughput screening format. It is based on monitoring the time-dependent increase of absorbance due to turbidity occurring by acid-induced protein aggregation. The method uses the highly amyloidogenic Y78F mutant of human transthyretin (heterogously expressed in Escherichia coli cells). Initial rates of protein aggregation at different inhibitor concentrations follow a monoexponential dose-response curve from which inhibition parameters are calculated. For the assay development, thyroid hormones and nonsteroidal antiinflamatory drugs were chosen among other reference compounds. Some of them are already known to be in vitro inhibitors of TTR amyloidogenesis. Analysis time is optimized to last 1.5 h, and the method is implemented in microtiter plates for screening of libraries of potential fibrillogenesis inhibitors.


Assuntos
Amiloide/metabolismo , Amiloidose/metabolismo , Desenho de Fármacos , Pré-Albumina/antagonistas & inibidores , Proteínas Recombinantes/antagonistas & inibidores , Anti-Inflamatórios não Esteroides/farmacologia , Relação Dose-Resposta a Droga , Escherichia coli/genética , Humanos , Cinética , Pré-Albumina/genética , Proteínas Recombinantes/genética , Hormônios Tireóideos/farmacologia , Triclosan/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...