Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Gynecol Oncol ; 156(2): 308-314, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31822399

RESUMO

BACKGROUND: Selinexor is an oral inhibitor of the nuclear export protein Exportin 1 (XPO1) with demonstrated antitumor activity in solid and hematological malignancies. We evaluated the efficacy and safety of selinexor in heavily pretreated, recurrent gynecological malignancies. METHODS: In this phase 2 trial, patients received selinexor (35 or 50 mg/m2 twice-weekly [BIW] or 50 mg/m2 once-weekly [QW]) in 4-week cycles. Primary endpoint was disease control rate (DCR) including complete response (CR), partial response (PR) or stable disease (SD) ≥12 weeks. Secondary endpoints were progression-free survival (PFS), overall survival (OS) and safety. RESULTS: 114 patients with ovarian (N = 66), endometrial (N = 23) or cervical (N = 25) cancer were enrolled. Median number of prior regimens for ovarian, endometrial and cervical cancer was 6 (1-11), 2 (1-5), and 3 (1-6) respectively. DCR was 30% (ovarian 30%; endometrial 35%; cervical 24%), which included confirmed PRs in 8%, 9%, and 4% of patients with ovarian, endometrial, and cervical cancer respectively. Median PFS and OS for patients with ovarian, endometrial and cervical cancer were 2.6, 2.8 and 1.4 months, and 7.3, 7.0, and 5.0 months, respectively. Common Grade 3/4 adverse events (AEs) were thrombocytopenia (17%), fatigue (14%), anemia (10%), nausea (9%) and hyponatremia (9%). Patients with ovarian cancer receiving 50 mg/m2 QW had fewer high-grade AEs with similar efficacy as BIW treatment. CONCLUSIONS: Selinexor demonstrated single-agent activity and disease control in patients with heavily pretreated ovarian and endometrial cancers. Side effects were a function of dose level and treatment frequency, similar to previous reports, reversible and mitigated with supportive care.


Assuntos
Neoplasias dos Genitais Femininos/tratamento farmacológico , Hidrazinas/administração & dosagem , Carioferinas/antagonistas & inibidores , Recidiva Local de Neoplasia/tratamento farmacológico , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Triazóis/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Feminino , Neoplasias dos Genitais Femininos/metabolismo , Neoplasias dos Genitais Femininos/patologia , Humanos , Hidrazinas/efeitos adversos , Carioferinas/metabolismo , Pessoa de Meia-Idade , Gradação de Tumores , Recidiva Local de Neoplasia/patologia , Intervalo Livre de Progressão , Receptores Citoplasmáticos e Nucleares/metabolismo , Triazóis/efeitos adversos , Proteína Exportina 1
2.
J Cell Biol ; 150(4): 929-36, 2000 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-10953017

RESUMO

In the Xenopus embryo, blastomeres are joined by gap junctions that allow the movement of small molecules between neighboring cells. Previous studies using Lucifer yellow (LY) have reported asymmetries in the patterns of junctional communication suggesting involvement in dorso-ventral patterning. To explore that relationship, we systematically compared the transfer of LY and neurobiotin in embryos containing 16-128 cells. In all cases, the junction-permeable tracer was coinjected with a fluorescent dextran that cannot pass through gap junctions. Surprisingly, while LY appeared to transfer in whole-mount embryos, in no case did we observe junctional transfer of LY in fixed and sectioned embryos. The lack of correspondence between data obtained from whole-mounts and from sections results from two synergistic effects. First, uninjected blastomeres in whole-mounts reflect and scatter light originating from the intensely fluorescent injected cell, creating a diffuse background interpretable as dye transfer. Second, the heavier pigmentation in ventral blastomeres masks this scattered signal, giving the impression of an asymmetry in communication. Thus, inspection of whole-mount embryos is an unreliable method for the assessment of dye transfer between embryonic blastomeres. A rigorous and unambiguous demonstration of gap junctional intercellular communication demands both the coinjection of permeant and impermeant tracers followed by the examination of sectioned specimens. Whereas LY transfer was never observed, neurobiotin was consistently transferred in both ventral and dorsal aspects of the embryo, with no apparent asymmetry. Ventralization of embryos by UV irradiation and dorsalization by Xwnt-8 did not alter the patterns of communication. Thus, our results are not compatible with current models for a role of gap junctional communication in dorso-ventral patterning.


Assuntos
Blastômeros/fisiologia , Comunicação Celular/fisiologia , Embrião não Mamífero/fisiologia , Junções Comunicantes/fisiologia , Xenopus laevis/embriologia , Animais , Biotina/análogos & derivados , Blastômeros/citologia , Blastômeros/ultraestrutura , Citoplasma/fisiologia , Citoplasma/ultraestrutura , Embrião não Mamífero/citologia , Embrião não Mamífero/ultraestrutura , Junções Comunicantes/ultraestrutura , RNA Mensageiro/genética , Transcrição Gênica
3.
J Cell Biol ; 106(3): 585-96, 1988 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-3126192

RESUMO

A cDNA clone encoding a portion of Drosophila nuclear lamins Dm1 and Dm2 has been identified by screening a lambda-gt11 cDNA expression library using Drosophila lamin-specific monoclonal antibodies. Two different developmentally regulated mRNA species were identified by Northern blot analysis using the initial cDNA as a probe, and full-length cDNA clones, apparently corresponding to each message, have been isolated. In vitro transcription of both full-length cDNA clones in a pT7 transcription vector followed by in vitro translation in wheat germ lysate suggests that both clones encode lamin Dm0, the polypeptide precursor of lamins Dm1 and Dm2. Nucleotide sequence analyses confirm the impression that both cDNA clones code for the identical polypeptide, which is highly homologous with human lamins A and C as well as with mammalian intermediate filament proteins. The two clones differ in their 3'-untranslated regions. In situ hybridization of lamin cDNA clones to Drosophila polytene chromosomes shows only a single locus of hybridization at or near position 25F on the left arm of chromosome 2. Southern blot analyses of genomic DNA are consistent with the notion that a single or only a few highly similar genes encoding Drosophila nuclear lamin Dm0 exist in the genome.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster/genética , Genes , Proteínas Nucleares/genética , Precursores de Proteínas/genética , RNA Mensageiro/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA/genética , Feminino , Imunoensaio , Laminas , Dados de Sequência Molecular , Hibridização de Ácido Nucleico , Biossíntese de Proteínas , Homologia de Sequência do Ácido Nucleico , Transcrição Gênica
4.
Leukemia ; 31(1): 143-150, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27211268

RESUMO

Acute myeloid leukemia (AML) is a clonal hematologic malignant disease of developing myeloid cells that have acquired aberrant survival, uncontrolled proliferation and a block in normal hematopoietic cell differentiation. Standard chemotherapy often induces remissions in AML patients, but the disease frequently relapses due to incomplete targeting of leukemia-initiating cells (LICs), emphasizing the need for novel effective treatments. Exportin 1 (XPO1)-mediated nuclear export, which is inhibited by the drug selinexor, is an attractive new therapeutic target in AML. Selinexor has shown impressive activity in Phase I/II clinical trials for AML. Here we report the anti-leukemic efficacy and tolerability of KPT-8602, a second-generation XPO1 inhibitor. KPT-8602 demonstrates substantially reduced brain penetration compared to selinexor, with resultant attenuation of the central nervous system mediated side effects of anorexia and weight loss. Due to its improved tolerability profile, KPT-8602 can be given daily compared to the two or three times weekly regimen of selinexor, and exhibits greater anti-leukemic efficacy against both leukemic blasts and LICs in AML patient-derived xenograft models. Importantly, normal hematopoietic stem and progenitor cell (HSPC) frequency is not significantly reduced by KPT-8602, providing a therapeutic window for elimination of relapse-driving LICs while sparing normal HSPCs. These findings strongly endorse clinical testing of KPT-8602 in patients with relapsed and refractory AML.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Carioferinas/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Crise Blástica/tratamento farmacológico , Crise Blástica/patologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Xenoenxertos , Humanos , Hidrazinas , Leucemia Mieloide Aguda/patologia , Camundongos , Triazóis , Proteína Exportina 1
5.
Leukemia ; 30(1): 190-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26202935

RESUMO

Currently available combination chemotherapy for acute myeloid leukemia (AML) often fails to result in long-term remissions, emphasizing the need for novel therapeutic strategies. We reasoned that targeted inhibition of a prominent nuclear exporter, XPO1/CRM1, could eradicate self-renewing leukemia-initiating cells (LICs) whose survival depends on timely XPO1-mediated transport of specific protein and RNA cargoes. Using an immunosuppressed mouse model bearing primary patient-derived AML cells, we demonstrate that selinexor (KPT-330), an oral antagonist of XPO1 that is currently in clinical trials, has strong activity against primary AML cells while sparing normal stem and progenitor cells. Importantly, limiting dilution transplantation assays showed that this cytotoxic activity is not limited to the rapidly proliferating bulk population of leukemic cells but extends to the LICs, whose inherent drug resistance and unrestricted self-renewal capacity has been implicated in the difficulty of curing AML patients with conventional chemotherapy alone.


Assuntos
Hidrazinas/farmacologia , Carioferinas/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Triazóis/farmacologia , Animais , Humanos , Terapia de Imunossupressão , Leucemia Mieloide Aguda/patologia , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína Exportina 1
6.
Oncogene ; 9(4): 1241-5, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7510864

RESUMO

p53 has at least two conformations that differ in their immunoreactivity. They consist of the functional tumor suppressor form, characteristic of the wild type p53 and the mutant form, generated by changes in the primary amino acid sequence of the protein. It has been previously shown that the wild type p53 protein also acquires the mutant conformation upon certain changes in growth conditions. Here we report that similar epitopic changes can be induced in crude cell lysate by addition of vanadate anions at 1 mM final concentration. A panel of anti p53 antibodies was used to discriminate between the different immunoreactive forms of wild type p53 in SV80 fibroblasts. It was found that addition of sodium vanadate to the lysis buffer converted part of p53 molecules into a mutant conformation that is recognized by the PAb 240 monoclonal antibodies. The effect of vanadate on p53 conformation was prominent even if it was added to the cell lysates after 15 min of pre-incubation at 37 degrees C. This further excluded its possible role as phosphatase inhibitor in the system and suggested a direct interaction with the p53 protein itself. Based on these data we recommend to avoid using sodium vanadate as a phosphatase inhibitor in experiments where in vivo conformational changes of wild type p53 are studied.


Assuntos
Epitopos , Genes p53 , Conformação de Ácido Nucleico/efeitos dos fármacos , Vanadatos/farmacologia , Anticorpos Monoclonais , Linhagem Celular Transformada , Humanos , Mutação
7.
Mech Dev ; 63(2): 199-209, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9203142

RESUMO

Xwnt-2b is a novel member of the Wnt gene family and is 73-74% similar to human and mouse Wnt-2 proteins. Starting from stage 15, Xwnt-2b transcripts are localized to a non-contiguous stripe in the anterior neural plate of the Xenopus embryo. In the tailbud, Xwnt-2b is expressed along the dorsoanterior side of the prosencephalon-mesencephalon boundary. At the tadpole stages, the brain-specific expression fades, but the total amount of Xwnt-2b mRNA does not decline due to activation of its expression in non-brain areas. Microinjection of Xwnt-2b mRNA into a ventral blastomere of 4-8-cell embryos results in the formation of complete secondary body axes. These results suggest that Xwnt-2b is a member of the axis-inducing Wnts and that it is involved in brain development and in later organogenesis.


Assuntos
Padronização Corporal/genética , Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/genética , Proteínas do Tecido Nervoso/genética , Proteínas de Xenopus , Xenopus/genética , Sequência de Aminoácidos , Animais , Encéfalo/embriologia , Glicoproteínas/biossíntese , Proteínas de Homeodomínio/metabolismo , Humanos , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/metabolismo , Ratos , Homologia de Sequência de Aminoácidos , Proteína Wnt2 , Peixe-Zebra
8.
Cell Signal ; 9(3-4): 291-8, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9218130

RESUMO

Transforming growth factory beta (TGF-beta) is a potent growth inhibitor of epithelial cells. One of the strategies used to elucidate the anti-proliferative mode of action of TGF-beta is to find out whether the receptor-generated signals interact with components of the basic machinery of the cell cycle. In this study we examined whether p53 and two other cycle inhibitory genes that can be transactivated by p53 are affected by TGF-beta 1 in epithelial cells. We show that TGF-beta 1 signalling controls the intracellular localization as well as the phosphorylation pattern and the stability of p53 protein. TGF-beta signalling also elevates the expression of p21/waf-1 and gadd45. The observed modifications in the protein suggest that p53 is involved in mediation of TGF-beta 1 growth inhibition. However, in TGF-beta 1 growth inhibited cells, wild type p53 is not required for the accumulation of the two p53 downstream targets p21/waf-1 and gadd45.


Assuntos
Ciclinas/metabolismo , Inibidores do Crescimento/farmacologia , Proteínas/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/genética , Genes cdc , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Camundongos , Fosforilação , Proteínas/genética , RNA Mensageiro/metabolismo , Ativação Transcricional , Proteínas GADD45
9.
Leukemia ; 28(1): 155-65, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23588715

RESUMO

The key nuclear export protein CRM1/XPO1 may represent a promising novel therapeutic target in human multiple myeloma (MM). Here we showed that chromosome region maintenance 1 (CRM1) is highly expressed in patients with MM, plasma cell leukemia cells and increased in patient cells resistant to bortezomib treatment. CRM1 expression also correlates with increased lytic bone and shorter survival. Importantly, CRM1 knockdown inhibits MM cell viability. Novel, oral, irreversible selective inhibitors of nuclear export (SINEs) targeting CRM1 (KPT-185, KPT-330) induce cytotoxicity against MM cells (ED50<200 nM), alone and cocultured with bone marrow stromal cells (BMSCs) or osteoclasts (OC). SINEs trigger nuclear accumulation of multiple CRM1 cargo tumor suppressor proteins followed by growth arrest and apoptosis in MM cells. They further block c-myc, Mcl-1, and nuclear factor κB (NF-κB) activity. SINEs induce proteasome-dependent CRM1 protein degradation; concurrently, they upregulate CRM1, p53-targeted, apoptosis-related, anti-inflammatory and stress-related gene transcripts in MM cells. In SCID mice with diffuse human MM bone lesions, SINEs show strong anti-MM activity, inhibit MM-induced bone lysis and prolong survival. Moreover, SINEs directly impair osteoclastogenesis and bone resorption via blockade of RANKL-induced NF-κB and NFATc1, with minimal impact on osteoblasts and BMSCs. These results support clinical development of SINE CRM1 antagonists to improve patient outcome in MM.


Assuntos
Carioferinas/antagonistas & inibidores , Mieloma Múltiplo/terapia , Osteoclastos/patologia , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Humanos , Mieloma Múltiplo/patologia , Proteína Exportina 1
12.
Genomics ; 8(2): 217-24, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2123469

RESUMO

A complete nucleotide sequence of a 4.2-kb genomic fragment containing the Drosophila lamin gene and flanking sequences is presented. Primer extension experiments and sequence analysis revealed that transcription starts from a single promoter. The lamin maternal 2.8-kb transcript and the 3.0-kb zygotic transcript are generated from two alternative polyadenylation sites. The gene contains four exons. The first intron is 7 bp upstream of the first AUG site. The two other introns are located within the alpha-helical rod domain of the protein: one in coil 1B in the 42-amino-acid domain that is absent in vertebrate cytoplasmic intermediate filament proteins and the other in coil 2 at a position different from intron positions within the vertebrate intermediate filament genes. Together with the sequence homology analysis, the data suggest either that the lamin gene was the ancestral gene of intermediate filament genes or that the lamin gene diverged from other intermediate filament genes early in evolution.


Assuntos
Drosophila melanogaster/genética , Proteínas Nucleares/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Evolução Biológica , Regulação da Expressão Gênica , Genes , Proteínas de Filamentos Intermediários/genética , Laminas , Dados de Sequência Molecular , Família Multigênica , Sequências Reguladoras de Ácido Nucleico , Homologia de Sequência do Ácido Nucleico
13.
J Cell Sci ; 112 ( Pt 14): 2391-6, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10381394

RESUMO

Innexins comprise a large family of genes that are believed to encode invertebrate gap junction channel-forming proteins. However, only two Drosophila innexins have been directly tested for the ability to form intercellular channels and only one of those was active. Here we tested the ability of Caenorhabditis elegans family members INX-3 and EAT-5 to form intercellular channels between paired Xenopus oocytes. We show that expression of INX-3 but not EAT-5, induces electrical coupling between the oocyte pairs. In addition, analysis of INX-3 voltage and pH gating reveals a striking degree of conservation in the functional properties of connexin and innnexin channels. These data strongly support the idea that innexin genes encode intercellular channels.


Assuntos
Proteínas de Caenorhabditis elegans , Conexinas/metabolismo , Proteínas de Helminto/metabolismo , Canais Iônicos/metabolismo , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Comunicação Celular , Conexinas/genética , Feminino , Junções Comunicantes/metabolismo , Expressão Gênica , Genes de Helmintos , Proteínas de Helminto/genética , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Ativação do Canal Iônico , Canais Iônicos/genética , Potenciais da Membrana , Oócitos/metabolismo , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA