Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Leukemia ; 32(3): 736-743, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29064484

RESUMO

Cell adhesion in the multiple myeloma (MM) microenvironment has been recognized as a major mechanism of MM cell survival and the development of drug resistance. Here we addressed the hypothesis that the protein junctional adhesion molecule-A (JAM-A) may represent a novel target and a clinical biomarker in MM. We evaluated JAM-A expression in MM cell lines and in 147 MM patient bone marrow aspirates and biopsies at different disease stages. Elevated JAM-A levels in patient-derived plasma cells were correlated with poor prognosis. Moreover, circulating soluble JAM-A (sJAM-A) levels were significantly increased in MM patients as compared with controls. Notably, in vitro JAM-A inhibition impaired MM migration, colony formation, chemotaxis, proliferation and viability. In vivo treatment with an anti-JAM-A monoclonal antibody (αJAM-A moAb) impaired tumor progression in a murine xenograft MM model. These results demonstrate that therapeutic targeting of JAM-A has the potential to prevent MM progression, and lead us to propose JAM-A as a biomarker in MM, and sJAM-A as a serum-based marker for clinical stratification.


Assuntos
Biomarcadores Tumorais , Molécula A de Adesão Juncional/sangue , Mieloma Múltiplo/sangue , Mieloma Múltiplo/mortalidade , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Medula Óssea/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Expressão Gênica , Humanos , Molécula A de Adesão Juncional/genética , Masculino , Camundongos , Terapia de Alvo Molecular , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Prognóstico
2.
Leukemia ; 31(4): 922-933, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27686868

RESUMO

Direct therapeutic targeting of oncogenic RAS is currently still impossible due to lack of suitable pharmacological inhibitors. Because specific blockade of druggable RAS effectors might represent an alternative treatment approach, we evaluated the role of the Raf complex for multiple myeloma (MM) pathobiology. We found frequent overexpression of the Raf isoforms (A-, B- and C-Raf) and downstream activation of MEK1,2/ERK1,2 in MM cells. Concomitant inhibition of all Raf isoforms (pan-Raf inhibition) by RNAi or pharmacological inhibitors was required to strongly induce apoptosis in human MM cell lines (HMCLs), in primary MM cells in vitro, and in a syngeneic MM mouse model in vivo. The anti-MM effect of pan-Raf inhibition did not correlate with the RAS mutation status, and functionally appeared to involve both MEK-dependent and -independent mechanisms. Furthermore, transcriptome analyses revealed that pan-Raf activity affects PI3K-dependent signalling, thus highlighting a functional link between the RAS/Raf and PI3K/mTOR/Akt pro-survival pathways. Accordingly, pharmacological inhibition of PI3K strongly enhanced the anti-MM effect of pan-Raf inhibition in MM cell lines and in primary MM cells in vitro and in vivo. Concomitant pan-Raf/PI3K inhibition was also effective in carfilzomib- and lenalidomide-resistant MM models underscoring that this attractive therapeutic anti-MM strategy is suitable for immediate clinical translation.


Assuntos
Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Transdução de Sinais , Proteínas ras/genética , Apoptose/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Isoenzimas , Lenalidomida , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Serina-Treonina Quinases TOR/metabolismo , Talidomida/análogos & derivados , Talidomida/farmacologia
3.
Blood Cancer J ; 3: e102, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23396385

RESUMO

Multiple myeloma (MM) is a largely incurable plasma cell malignancy with a poorly understood and heterogeneous clinical course. To identify potential, functionally relevant somatic mutations in MM, we performed whole-exome sequencing of five primary MM, corresponding germline DNA and six MM cell lines, and developed a bioinformatics strategy that also integrated published mutational data of 38 MM patients. Our analysis confirms that identical, recurrent mutations of single genes are infrequent in MM, but highlights that mutations cluster in important cellular pathways. Specifically, we show enrichment of mutations in adhesion molecules of MM cells, emphasizing the important role for the interaction of the MM cells with their microenvironment. We describe an increased rate of mutations in receptor tyrosine kinases (RTKs) and associated signaling effectors, for example, in EGFR, ERBB3, KRAS and MAP2K2, pointing to a role of aberrant RTK signaling in the development or progression of MM. The diversity of mutations affecting different nodes of a particular signaling network appears to be an intrinsic feature of individual MM samples, and the elucidation of intra- as well as interindividual redundancy in mutations that affect survival pathways will help to better tailor targeted therapeutic strategies to the specific needs of the MM patient.

4.
Cell Death Dis ; 2: e194, 2011 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-21850048

RESUMO

The huge majority of myeloma cell lines express TNFR2 while a substantial subset of them failed to show TNFR1 expression. Stimulation of TNFR1 in the TNFR1-expressing subset of MM cell lines had no or only a very mild effect on cellular viability. Surprisingly, however, TNF stimulation enhanced cell death induction by CD95L and attenuated the apoptotic effect of TRAIL. The contrasting regulation of TRAIL- and CD95L-induced cell death by TNF could be traced back to the concomitant NFκB-mediated upregulation of CD95 and the antiapoptotic FLIP protein. It appeared that CD95 induction, due to its strength, overcompensated a rather moderate upregulation of FLIP so that the net effect of TNF-induced NFκB activation in the context of CD95 signaling is pro-apoptotic. TRAIL-induced cell death, however, was antagonized in response to TNF because in this context only the induction of FLIP is relevant. Stimulation of TNFR2 in myeloma cells leads to TRAF2 depletion. In line with this, we observed cell death induction in TNFR1-TNFR2-costimulated JJN3 cells. Our studies revealed that the TNF-TNF receptor system adjusts the responsiveness of the extrinsic apoptotic pathway in myeloma cells by multiple mechanisms that generate a highly context-dependent net effect on myeloma cell survival.


Assuntos
Apoptose , Mieloma Múltiplo/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Linhagem Celular Tumoral , Proteína Ligante Fas/farmacologia , Humanos , NF-kappa B/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/metabolismo
5.
Oncogene ; 30(28): 3198-206, 2011 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-21478911

RESUMO

Multiple myeloma (MM) is a paradigm for a malignant disease that exploits external stimuli of the microenvironment for growth and survival. A thorough understanding of the complex interactions between malignant plasma cells and their surrounding requires a detailed analysis of the transcriptional response of myeloma cells to environmental signals. We determined the changes in gene expression induced by interleukin (IL)-6, tumor necrosis factor-α, IL-21 or co-culture with bone marrow stromal cells in myeloma cell lines. Among a limited set of genes that were consistently activated in response to growth factors, a prominent transcriptional target of cytokine-induced signaling in myeloma cells was the gene encoding the serine/threonine kinase serum/glucocorticoid-regulated kinase 1 (SGK1), which is a down-stream effector of PI3-kinase. We could demonstrate a rapid, strong and sustained induction of SGK1 in the cell lines INA-6, ANBL-6, IH-1, OH-2 and MM.1S as well as in primary myeloma cells. Pharmacologic inhibition of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway abolished STAT3 phosphorylation and SGK1 induction. In addition, small hairpin RNA (shRNA)-mediated knock-down of STAT3 reduced basal and induced SGK1 levels. Furthermore, downregulation of SGK1 by shRNAs resulted in decreased proliferation of myeloma cell lines and reduced cell numbers. On the molecular level, this was reflected by the induction of cell cycle inhibitory genes, for example, CDKNA1/p21, whereas positively acting factors such as CDK6 and RBL2/p130 were downregulated. Our results indicate that SGK1 is a highly cytokine-responsive gene in myeloma cells promoting their malignant growth.


Assuntos
Citocinas/farmacologia , Proteínas Imediatamente Precoces/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Proteínas Serina-Treonina Quinases/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Citocinas/metabolismo , Regulação para Baixo/genética , Humanos , Proteínas Imediatamente Precoces/deficiência , Janus Quinases/metabolismo , Mieloma Múltiplo/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Interferência de RNA , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
6.
Br J Cancer ; 102(11): 1578-91, 2010 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-20502461

RESUMO

BACKGROUND: Heat-shock protein 90 (Hsp90) has a crucial role in both the stabilisation and regulation of various proteins, including those related to radioresistance. Inhibition of Hsp90 may therefore provide a strategy for enhancing the radiosensitivity of tumour cells. This study explores the responses of four tumour cell lines (A549, GaMG, HT 1080 and SNB19) to combined treatment with ionising radiation (IR) and two novel inhibitors of Hsp90, NVP-AUY922 and NVP-BEP800. The techniques used included cell and colony counts, expression of Hsp90, Hsp70, Akt, survivin, cleaved caspase 3, p53, cell-cycle progression and associated proteins. DNA damage was analysed by histone gammaH2AX and Comet assays. RESULTS: We found that NVP-AUY922 and NVP-BEP800 enhanced radiosensitivity in all tested cell lines. In contrast, only two cell lines (HT 1080 and GaMG) exhibited an increased rate of apoptosis after drug pretreatment, as revealed by western blot. In all tested cell lines, the expression of histone gammaH2AX, a marker of DNA double-strand breaks, after combined drug-IR treatment was higher and its decay rate was slower than those after each single treatment modality. Drug-IR treatment also resulted in impaired cell-cycle progression, as indicated by S-phase depletion and G2/M arrest. In addition, the cell cycle-associated proteins, Cdk1 and Cdk4, were downregulated after Hsp90 inhibition. INTERPRETATION: These findings show that the novel inhibitors of Hsp90 can radiosensitise tumour cell lines of different entities through destabilisation and depletion of several Hsp90 client proteins, thus causing the depletion of S phase and G2/M arrest, increased DNA damage and repair protraction and, to some extent, apoptosis. The results might have important implications for the radiotherapy of solid tumours.


Assuntos
Ciclo Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Isoxazóis/farmacologia , Neoplasias/radioterapia , Pirimidinas/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Resorcinóis/farmacologia , Benzoquinonas/farmacologia , Ciclo Celular/genética , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular , Dano ao DNA/genética , Reparo do DNA/genética , Reparo do DNA/efeitos da radiação , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/metabolismo , DNA de Neoplasias/efeitos da radiação , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/efeitos da radiação , Avaliação Pré-Clínica de Medicamentos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Lactamas Macrocíclicas/farmacologia , Neoplasias/genética , Radiossensibilizantes/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/efeitos da radiação
7.
Leukemia ; 22(8): 1604-12, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18480838

RESUMO

We as well as others have recently shown that Hsp90 is overexpressed in multiple myeloma (MM) and critically contributes to tumour cell survival. Pharmacologic blockade of Hsp90 has consistently been found to induce MM cell death. However, most data have been obtained with MM cell lines whereas knowledge about the molecular effects of pharmacologic Hsp90 blockade in primary tumour cells is limited. Furthermore, these investigations have so far focused on geldanamycin derivatives. We analysed the biochemical effects of a novel diarylisoxazole-based Hsp90 inhibitor (NVP-AUY922) on signalling pathways and cell death in a large set of primary MM tumour samples and in MM cell lines. Treated cells displayed the molecular signature and pharmacodynamic properties for abrogation of Hsp90 function, such as downregulation of multiple survival pathways and strong upregulation of Hsp70. NVP-AUY922 treatment efficiently induced MM cell apoptosis and revealed both sensitive and resistant subgroups. Sensitivity was not correlated with TP53 mutation or Hsp70 induction levels and stromal cells from the bone marrow microenvironment were unable to abrogate NVP-AUY922-induced apoptosis of MM cells. Thus, NVP-AUY922 may be a promising drug for treatment of MM and clinical studies are warranted.


Assuntos
Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Isoxazóis/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Resorcinóis/farmacologia , Transdução de Sinais , Apoptose , Linhagem Celular Tumoral , Técnicas de Cocultura , Humanos , Isoxazóis/uso terapêutico , Mieloma Múltiplo/patologia , Resorcinóis/uso terapêutico
8.
Leukemia ; 21(4): 772-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17268519

RESUMO

The status of the p53 pathway in classical Hodgkin lymphoma (cHL) remains unclear, and a lack of proven TP53 mutations contrasts with often high expression levels of p53 protein. In this study, we demonstrate that pharmacologic activation of the p53 pathway with the murine double minute 2 (MDM2) antagonist nutlin-3 in Hodgkin lymphoma-derived cell lines leads to effective apoptosis induction and sensitizes the cells to other anticancer drugs. Cells with mutant p53 are resistant to nutlin-3, but sensitive to geldanamycin, a pharmacologic inhibitor of heat shock 90 kDa protein (HSP90), indicating that HSP90 inhibition can induce apoptosis in a p53-independent manner. Conversely, cells with defects in the HSP90/nuclear factor-kappa B pathway expressing wild-type p53 are more resistant to geldanamycin, but still sensitive to nutlin-3. Our results suggest that selective activation of p53 by MDM2 antagonists as a single agent or in combination with conventional chemotherapeutics and/or inhibitors of p53-independent survival pathways may offer effective treatment options for patients with cHL. Importantly, because nutlins and HSP90 inhibitors are non-genotoxic agents, their use might offer a means to reduce the genotoxic burden of current chemotherapeutic regimens.


Assuntos
Células de Reed-Sternberg/patologia , Proteína Supressora de Tumor p53/genética , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzoquinonas/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doença de Hodgkin/genética , Doença de Hodgkin/patologia , Humanos , Imidazóis/farmacologia , Lactamas Macrocíclicas/farmacologia , Camundongos , Piperazinas/farmacologia , RNA Interferente Pequeno/genética , Transcrição Gênica
9.
Verh Dtsch Ges Pathol ; 91: 330-7, 2007.
Artigo em Alemão | MEDLINE | ID: mdl-18314631

RESUMO

HSP90's are overexpressed in different cancer types and they probably are required to sustain aberrant signalling in malignant cells. Recently, pharmacological inhibition of HSP90 was found to suppress growth of myeloma cell lines and in primary myeloma cells. Therefore, we wanted to investigate the role of HSP90alpha and HSP90beta in the pathogenesis of malignant myeloma (MM) in more detail. Immunohistochemistry was employed to examine the expression of HSP90alpha and HSP90beta in MM. The importance of HSP90 for survival of MM -cells was investigated by SiRNA-mediated knockdown of HSP90 and blockade of the IL-6R/STAT3 and the MAPK signaling pathways in vitro. HSP90alpha and HSP90beta were overexpressed in majority of investigated MM cases, but not in MGUS or in normal plasma cells. SiRNA-mediated knockdown of HSP90 or treatment with the novel HSP90 inhibitor 17-DMAG attenuated the levels of STAT3 and phospho-ERK and decreased the viability of MM cells. The knockdown of HSP90alpha was sufficient to induce apoptosis. This effect was strongly increased when both HSP90s were targeted, indicating a cooperation of both. HSP90 critically contributes to myeloma survival in the context of its microenvironment and therefore strengthen the potential value of HSP90 as a therapeutic target.

10.
J Neurosci ; 20(2): 709-21, 2000 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-10632600

RESUMO

Four Dlx homeobox genes, Dlx1, Dlx2, Dlx5, and Dlx6 are expressed in the same primordia of the mouse forebrain with temporally overlapping patterns. The four genes are organized as two tail-to-tail pairs, Dlx1/Dlx2 and Dlx5/Dlx6, a genomic arrangement conserved in distantly related vertebrates like zebrafish. The Dlx5/Dlx6 intergenic region contains two sequences of a few hundred base pairs, remarkably well conserved between mouse and zebrafish. Reporter transgenes containing these two sequences are expressed in the forebrain of transgenic mice and zebrafish with patterns highly similar to endogenous Dlx5 and Dlx6 expression. The activity of the transgene is drastically reduced in mouse mutants lacking both Dlx1 and Dlx2, consistent with the decrease in endogenous Dlx5 and Dlx6 expression. These results suggest that cross-regulation by Dlx proteins, mediated by the intergenic sequences, is essential for Dlx5 and Dlx6 expression in the forebrain. This hypothesis is supported by cotransfection and DNA-protein binding experiments. We propose that the Dlx genes are part of a highly conserved developmental pathway that regulates forebrain development.


Assuntos
Elementos Facilitadores Genéticos , Genes Homeobox , Proteínas de Homeodomínio/genética , Prosencéfalo/metabolismo , Fatores de Transcrição , Proteínas de Peixe-Zebra , Animais , Sequência de Bases , Sequência Conservada , Ectoderma/metabolismo , Humanos , Íntrons , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Bulbo Olfatório/metabolismo , Prosencéfalo/embriologia , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Peixe-Zebra
11.
Proc Natl Acad Sci U S A ; 94(15): 8214-9, 1997 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-9223341

RESUMO

The molecular evolution of the opioid receptor family has been studied by isolating cDNAs that encode six distinct opioid receptor-like proteins from a lower vertebrate, the teleost fish Catostomus commersoni. One of these, which has been obtained in full-length form, encodes a 383-amino acid protein that exhibits greatest sequence similarity to mammalian mu-opioid receptors; the corresponding gene is expressed predominantly in brain and pituitary. Transfection of the teleost cDNA into HEK 293 cells resulted in the appearance of a receptor having high affinity for the mu-selective agonist [D-Ala2, MePhe4-Gly-ol5]enkephalin (DAMGO) (Kd = 0.63 +/- 0.15 nM) and for the nonselective antagonist naloxone (Kd = 3.1 +/- 1.3 nM). The receptor had negligible affinity for U50488 and [D-Pen2, D-Pen5]enkephalin (DPDPE), which are kappa- and delta-opioid receptor selective agonists, respectively. Stimulation of transfected cells with 1 microM DAMGO lowered forskolin-induced cAMP levels, an effect that could be reversed by naloxone. Experiments in Xenopus oocytes have demonstrated that the fish opioid receptor can, in an agonist-dependent fashion, activate a coexpressed mouse G-protein-gated inward-rectifying potassium channel (GIRK1). The identification of six distinct fish opioid receptor-like proteins suggests that additional mammalian opioid receptors remain to be identified at the molecular level. Furthermore, our data indicate that the mu-opioid receptor arose very early in evolution, perhaps before the appearance of vertebrates, and that the pharmacological and functional properties of this receptor have been conserved over a period of approximately 400 million years implying that it fulfills an important physiological role.


Assuntos
Evolução Molecular , Peixes/genética , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/metabolismo , Receptores Opioides mu/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Clonagem Molecular , DNA Complementar , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Humanos , Dados de Sequência Molecular , Ligação Proteica , Ensaio Radioligante , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Homologia de Sequência de Aminoácidos , Xenopus laevis
12.
Eur J Neurosci ; 9(11): 2414-22, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9464935

RESUMO

The pharmacological properties of homo-oligomeric channels formed by the GABA type A receptor-like rho1 and rho2 polypeptides are very reminiscent of those of the GABA type C receptors that have been extensively characterized in the retina. Similar receptors have been reported to occur in certain brain regions of a variety of vertebrate species. We have used in situ hybridization to investigate the expression patterns of the rho1- and rho2-polypeptide genes in the brain of the 1-day-old chick (Gallus domesticus) and the adult rat (Rattus norvegicus). Our results show that in the chick both the rho1- and rho2-subunit transcripts are present in the cerebellum, the optic tectum, the epithalamus and the nucleus pretectalis. However, the two messenger RNAs are often found in different populations of cells. Thus, only the rho1-subunit gene is expressed in the deep cerebellar nuclei, the dorsal thalamus, the ectostriatum and the tractus vestibulomesencephalicus, while only the rho2-subunit gene is transcribed in the nucleus habenularis lateralis and the nucleus isthmo-opticus. In contrast, neither of the rho-polypeptide messenger RNAs can be detected by in situ hybridization in the rat central nervous system. Reverse transcription-polymerase chain reaction amplification has been used to confirm the expression of the two rho-subunit genes in the chicken brain. Surprisingly, this highly sensitive technique also revealed transcription of these genes in the rat brain. We conclude that the rho1- and rho2-subunit genes are expressed at a much higher level in the avian brain than in the rat brain and that, at least in birds, subtypes of the GABA(C) receptor exist.


Assuntos
Química Encefálica/fisiologia , Galinhas/metabolismo , Receptores de GABA/biossíntese , Animais , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Química Encefálica/genética , Hibridização In Situ , Sondas de Oligonucleotídeos , Reação em Cadeia da Polimerase , RNA Mensageiro/biossíntese , Ratos , Receptores de GABA/genética , Especificidade da Espécie
13.
J Neurosci ; 16(9): 2869-80, 1996 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-8622118

RESUMO

We describe the isolation of a molluscan (Lymnaea stagnalis) full-length complementary DNA that encodes a mature polypeptide (which we have named Lym-eGluR2) with a predicted molecular weight of 105 kDa that exhibits 44-48% identity to the mammalian kainate-selective glutamate receptor GluR5, GluR6, and GluR7 subunits. Injection of in vitro-transcribed RNA from this clone into Xenopus laevis oocytes results in the robust expression of homo-oligomeric cation channels that can be gated by L-glutamate (EC50 = 1.2 +/- 0.3 micron) and several other glutamate receptor agonists; rank order of potency: glutamate >> kainate > ibotenate > AMPA. These currents can be blocked by the mammalian non-NMDA receptor antagonists 6,7-dinitroquinoxaline-2,3-dione, 6-cyano-7-nitroquinoxaline-2,3-dione, and 1-(4-chlorobenzoyl)piperazine-2,3-dicarboxylic acid. Ionic-replacement experiments have shown that the agonist-induced current is carried entirely by sodium and potassium ions. In situ hybridization has revealed that the Lym-eGluR2 transcript is present in all 11 ganglia of the Lymnaea CNS, including the 4-cluster motorneurons within the paired buccal ganglia. The pharmacological properties and deduced location of Lym-eGluR2 are entirely consistent with it being (a component of) the receptor, which has been identified previously on buccal motorneurons, that mediates the excitatory effects of glutamate released from neurons within the feeding central pattern generator.


Assuntos
Cátions/metabolismo , Comportamento Alimentar/fisiologia , Ácido Glutâmico/fisiologia , Ativação do Canal Iônico , Canais Iônicos/química , Canais Iônicos/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Canais Iônicos/genética , Dados de Sequência Molecular , Sondas de Oligonucleotídeos/genética , RNA Mensageiro/metabolismo , Relação Estrutura-Atividade
14.
Biophys Chem ; 46(3): 237-48, 1993 May.
Artigo em Inglês | MEDLINE | ID: mdl-8343570

RESUMO

Three mutants of the anaphylatoxin C5a were prepared with positions 2, 64 and 70, respectively, substituted by tryptophan. The last mutant was additionally labelled at Cys27 for fluorescence energy transfer (FET) measurements. The structural integrity and biological activity of the molecules were not affected. Fluorescence anisotropy decay (FAD) measurements showed that the rotational correlation time for tryptophan decreases in the order: [Trp2]rhC5a > [Trp64]rhC5a > [Trp70]rhC5a, indicating an increasing mobility of the side chain. Measurements of the fluorescence energy transfer from Trp70 to the 1,5-AEDANS group at Cys27 yielded a distance distribution of 2.4 +/- 0.8 nm. This value is compatible with the C-terminal chain being arranged as a slightly stretched helix pointing away from the body of the molecule.


Assuntos
Anafilatoxinas/química , Complemento C5a/química , Triptofano/química , Sequência de Aminoácidos , Anafilatoxinas/genética , Dicroísmo Circular , Complemento C5a/genética , Cisteína , Transferência de Energia , Polarização de Fluorescência , Corantes Fluorescentes , Humanos , Dados de Sequência Molecular , Mutação , Naftalenossulfonatos , Proteínas Recombinantes/química , Espectrofotometria Ultravioleta , Reagentes de Sulfidrila , Fatores de Tempo , Triptofano/genética
15.
Biochem J ; 288 ( Pt 1): 261-6, 1992 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-1445269

RESUMO

By site-directed mutagenesis of a human complement factor C5a cDNA clone, we have designed a hybrid anaphylatoxin in which three amino acid residues in the C-terminal sequence of human C5a were exchanged to create the native C-terminal human C3a (hC3a) sequence Leu-Gly-Leu-Ala-Arg. This hybrid anaphylatoxin rC5a-(1-69)-LGLAR exhibited true C3a and C5a activity when tested in the guinea pig ileum contraction assay. Quantitative measurements of ATP release from guinea pig platelets revealed about 1% intrinsic C3a activity for this hybrid, while the C5a activity was essentially unchanged. Competitive binding assays confirmed that the rC5a-(1-69)-LGLAR mutant was able to displace radioiodinated rhC5a with a KI of approx. 40 nM and hC3a with a KI of approx. 3.7 microM from guinea pig platelets. Since the C-termini of both human C3a and C5a anaphylatoxins are known to interact with their respective receptors, we conclude that the same peptidic sequence, LGLAR, is able to bind to and activate two different receptors, the C3a receptor as well as the C5a receptor. This clone provides a novel tool for the identification of further receptor-binding residues in both anaphylatoxins, since any mutants may be tested for altered C3a and C5a activity simultaneously.


Assuntos
Anafilatoxinas/farmacologia , Complemento C3a/farmacologia , Complemento C5a/farmacologia , Mutagênese Sítio-Dirigida , Trifosfato de Adenosina/sangue , Sequência de Aminoácidos , Anafilatoxinas/genética , Animais , Ligação Competitiva , Plaquetas/metabolismo , Complemento C3a/química , Complemento C3a/genética , Complemento C5a/química , Complemento C5a/genética , Cobaias , Íleo/fisiologia , Dados de Sequência Molecular , Contração Muscular , Receptor da Anafilatoxina C5a , Receptores de Complemento/metabolismo , Proteínas Recombinantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...