Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 318
Filtrar
1.
Clin Exp Immunol ; 199(3): 255-262, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31386175

RESUMO

Regulatory T cells (Tregs ) are specialized in immune suppression and play a dominant role in peripheral immune tolerance. Treg  cell lineage development and function maintenance is determined by the forkhead box protein 3 (FoxP3) transcriptional factor, whose activity is fine-tuned by its post-translational modifications (PTMs) and interaction partners. In this review, we summarize current studies in the crystal structures, the PTMs and interaction partners of FoxP3 protein, and discuss how these insights may provide a roadmap for new approaches to modulate Treg  suppression, and new therapies to enhance immune tolerance in autoimmune diseases.


Assuntos
Doenças Autoimunes/imunologia , Fatores de Transcrição Forkhead/imunologia , Tolerância Imunológica/imunologia , Linfócitos T Reguladores/imunologia , Animais , Doenças Autoimunes/metabolismo , Cristalografia por Raios X , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/metabolismo , Humanos , Modelos Moleculares , Domínios Proteicos , Processamento de Proteína Pós-Traducional/imunologia , Linfócitos T Reguladores/metabolismo
2.
Oncogene ; 34(29): 3770-9, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25241898

RESUMO

Medulloblastoma (MB) is a highly malignant brain tumor that occurs primarily in children. Although surgery, radiation and high-dose chemotherapy have led to increased survival, many MB patients still die from their disease, and patients who survive suffer severe long-term side effects as a consequence of treatment. Thus, more effective and less toxic therapies for MB are critically important. Development of such therapies depends in part on identification of genes that are necessary for growth and survival of tumor cells. Survivin is an inhibitor of apoptosis protein that regulates cell cycle progression and resistance to apoptosis, is frequently expressed in human MB and when expressed at high levels predicts poor clinical outcome. Therefore, we hypothesized that Survivin may have a critical role in growth and survival of MB cells and that targeting it may enhance MB therapy. Here we show that Survivin is overexpressed in tumors from patched (Ptch) mutant mice, a model of Sonic hedgehog (SHH)-driven MB. Genetic deletion of survivin in Ptch mutant tumor cells significantly inhibits proliferation and causes cell cycle arrest. Treatment with small-molecule antagonists of Survivin impairs proliferation and survival of both murine and human MB cells. Finally, Survivin antagonists impede growth of MB cells in vivo. These studies highlight the importance of Survivin in SHH-driven MB, and suggest that it may represent a novel therapeutic target in patients with this disease.


Assuntos
Neoplasias Cerebelares/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Inibidoras de Apoptose/deficiência , Meduloblastoma/metabolismo , Proteínas Repressoras/deficiência , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Compostos de Bifenilo/farmacologia , Western Blotting , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/genética , Quimiorradioterapia , Criança , Proteínas Hedgehog/antagonistas & inibidores , Humanos , Imidazóis/farmacologia , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Inibidoras de Apoptose/genética , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Antígeno Ki-67/metabolismo , Meduloblastoma/tratamento farmacológico , Meduloblastoma/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Microscopia Confocal , Naftoquinonas/farmacologia , Piridinas/farmacologia , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Survivina , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Dis Esophagus ; 28(4): 394-403, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24708360

RESUMO

Human epidermal growth factor receptor 2 (HER2) is involved in the malignant progression of several human cancers, including esophageal adenocarcinoma (EAC). The purpose of this study was to evaluate HER2 overexpression and to explore the feasibility of confocal laser endomicroscopy for in vivo molecular imaging of HER2 status in an animal model of Barrett's-related EAC. Rats underwent esophagojejunostomy with gastric preservation. At 30 weeks post-surgery, the esophagus of 46 rats was studied; endoscopic and histological findings were correlated with HER2 immunofluorescence on excised biopsies and gross specimens. At this age, 23/46 rats developed Barrett's esophagus (BE), and 6/46 had cancer (four EAC and two squamous cell carcinomas). A significant overexpression of HER2 was observed in esophageal adenocarcinoma compared with normal squamous esophagus (9.4-fold) and BE (6.0-fold). AKT and its phosphorylated form were also overexpressed in cancer areas. Molecular imaging was performed at 80 weeks post-surgery in four rats after tail injection of fluorescent-labeled anti-HER2 antibody. At this age, 3/4 rats developed advance adenocarcinoma and showed in vivo overexpression of HER2 by molecular confocal laser endomicroscopy with heterogeneous distribution within cancer; no HER2 signal was observed in normal or Barrett's tissues. Therefore, HER2 overexpression is a typical feature of the surgical induced model of EAC that can be easily quantified in vivo using an innovative mini-invasive approach including confocal endomicroscopy; this approach may avoid limits of histological evaluation of HER2 status on 'blinded' biopsies.


Assuntos
Adenocarcinoma/metabolismo , Esôfago de Barrett/metabolismo , Neoplasias Esofágicas/metabolismo , Imagem Molecular/métodos , Adenocarcinoma/induzido quimicamente , Animais , Esôfago de Barrett/complicações , Biópsia , Carcinoma de Células Escamosas/metabolismo , Modelos Animais de Doenças , Endoscopia , Neoplasias Esofágicas/induzido quimicamente , Imunofluorescência , Microscopia Intravital/métodos , Microscopia Confocal/métodos , Ratos , Ratos Sprague-Dawley , Receptor ErbB-2 , Coloração e Rotulagem
4.
Oncogene ; 32(17): 2220-9, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22689061

RESUMO

The Yes-associated protein (YAP) is a transcriptional factor involved in tissue development and tumorigenesis. Although YAP has been recognized as a key element of the Hippo signaling pathway, the mechanisms that regulate YAP activities remain to be fully characterized. In this study, we demonstrate that the non-receptor type protein tyrosine phosphatase 14 (PTPN14) functions as a negative regulator of YAP. We show that YAP forms a protein complex with PTPN14 through the WW domains of YAP and the PPXY motifs of PTPN14. In addition, PTPN14 inhibits YAP-mediated transcriptional activities. Knockdown of YAP sensitizes cancer cells to various anti-cancer agents, such as cisplatin, the EGFR tyrosine kinase inhibitor erlotinib and the small-molecule antagonist of survivin, S12. YAP-targeted modalities may be used in combination with other cancer drugs to achieve maximal therapeutic effects.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Fosfoproteínas/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Quinazolinas/farmacologia , Aciltransferases , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Motivos de Aminoácidos , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cisplatino/farmacologia , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Técnicas de Silenciamento de Genes , Genes Reporter , Células HEK293 , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Camundongos , Células NIH 3T3 , Fragmentos de Peptídeos/química , Fosfoproteínas/química , Fosfoproteínas/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Proteínas Tirosina Fosfatases não Receptoras/química , RNA Interferente Pequeno/genética , Survivina , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteínas de Sinalização YAP
5.
Oncogene ; 31(15): 1938-48, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21892210

RESUMO

Survivin is a member of the inhibitor of apoptosis protein family and has an essential role in mitosis. Survivin is overexpressed in a large variety of human cancers and represents an attractive target for cancer therapy. Epidermal growth factor receptor and Her/neu-transformed human tumors in particular exhibit high levels of survivin. The survivin protein forms dimers through a conserved region that is critical for subcellular localization and biological functions of the protein. We identified small molecules that target a specific cavity adjacent to the survivin dimerization surfaces. S12, a lead compound identified in the screen, can bind to the survivin protein at the intended target site. Moreover, S12 alters spindle formation, causing mitotic arrest and cell death, and inhibits tumor growth in vitro and in vivo. Cell death occurs in premetaphase stage following mitotic arrest and is not a consequence of general toxicity. Thus, the study validates a novel therapeutic target site in the survivin protein and provides a promising strategy to develop a new class of therapeutic small molecules for the treatment of human cancers.


Assuntos
Sítio Alostérico , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Fuso Acromático/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Sobrevivência Celular/efeitos dos fármacos , Humanos , Proteínas Inibidoras de Apoptose/química , Camundongos , Camundongos Nus , Mitose/efeitos dos fármacos , Transplante de Neoplasias , Neoplasias , Survivina
6.
Oncogene ; 27(27): 3870-4, 2008 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-18264138

RESUMO

2C4 (Pertuzumab, Omnitarg) is a monoclonal antibody targeting p185(her2/neu), which is overexpressed in 30% of invasive breast cancer. 2C4 is currently in phase II clinical trials for several types of cancers. This antibody has been reported to disrupt the association between p185(her2/neu) and ErbB3. In our studies of epidermal growth factor receptor (EGFR)-p185(her2/neu) heterodimerization, we noted that 2C4 formed associations with the EGFR-p185(her2/neu) receptor complex. Our data argue against 2C4 as a universal heterodimerization blocker for p185(her2/neu), but indicate that cocktails of monoclonal antibodies binding distinct interaction surfaces of p185(her2/neu) will emerge as the most potent targeted therapy.


Assuntos
Anticorpos Monoclonais/metabolismo , Receptores ErbB/imunologia , Receptor ErbB-2/imunologia , Receptor ErbB-3/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Sítios de Ligação de Anticorpos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Sinergismo Farmacológico , Feminino , Humanos , Receptor ErbB-2/efeitos dos fármacos
7.
Oncogene ; 25(59): 7740-6, 2006 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-16785990

RESUMO

The anti-p185(her2/neu) peptidomimetic (AHNP) is a small exo-cyclic peptide derived from the anti-p185(her2/neu) rhumAb 4D5 (h4D5). AHNP mimics many but not all of the antitumor characteristics exhibited by h4D5. However, the pharmacokinetic profiles of AHNP are less than optimal for therapeutic or diagnostic purposes. To improve the binding affinity to p185(her2/neu) and the antitumor efficacy, we have engineered a fusion protein containing AHNP and a nonimmunoglobulin protein scaffold, streptavidin (SA). The recombinant protein, AHNP-SA (ASA) bound to p185(her2/neu) with high affinity, inhibited the proliferation of p185(her2/neu)-overexpressing cells, and reduced tumor growth induced by p185(her2/neu)-transformed cells. These data suggest that the bacterially produced tetrameric ASA can be used as an antibody-surrogate molecule. This class of molecule will play a role in the diagnosis and treatment of p185(her2/neu)-related tumors. Our studies establish a general principle by which a small biologically active synthetic exo-cyclic peptide can be engineered to enhance functional aspects by structured oligomerization and can be produced recombinantly using bacterial expression.


Assuntos
Anticorpos Monoclonais/biossíntese , Receptor ErbB-2/imunologia , Proteínas Recombinantes de Fusão/biossíntese , Estreptavidina/biossíntese , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Proliferação de Células/efeitos dos fármacos , Camundongos , Modelos Moleculares , Células NIH 3T3 , Neoplasias Experimentais/tratamento farmacológico , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia , Estreptavidina/química
8.
Oncogene ; 25(45): 6048-55, 2006 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-16682949

RESUMO

The centrosome modulates spindle formation and plays a critical role in guiding proper segregation of chromosomes during cell division. Centrosome aberrations, frequently seen in human tumors, may cause abnormal chromosome segregation and contribute to malignant transformation. To explore the components of the centrosomes, we previously identified a novel centrosomal protein called Su48. To further characterize the Su48-containing protein ensemble in the centrosome, we performed yeast two-hybrid screens and isolated a number of Su48-interacting molecules, including the centrosomal protein Nde1. Here, we demonstrate that Su48 can associate with Nde1. Moreover, we found that Nde1 is subjected to phosphorylation in vivo. In particular, we identified six putative Cdc2 phosphorylation sites in Nde1 and found that alteration of these sites diminishes phosphorylation by Cdc2 in vitro and affects the stability of Su48-Nde1 interactions and the centrosomal localization of Nde1. Ablation of Nde1 by gene specific small interfering RNA causes mitotic delay and cell death, coupled with a modest decrease in the incidence of the cells that harbor excessive centrosomes. Collectively, our findings indicate that Nde1 can form a protein complex with Su48 in the centrosome and plays an important role for successful mitosis.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ligação a DNA , Células HeLa , Humanos , Imunoprecipitação , Fosforilação , Fatores de Transcrição , Técnicas do Sistema de Duplo-Híbrido
9.
Oncogene ; 25(35): 4937-46, 2006 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-16547491

RESUMO

The centrosome, an organelle that functions as the major microtubule-organizing center, plays an essential role in the formation of the mitotic spindle and guiding accurate chromosome segregation. Centrosome aberrations are frequently associated with various forms of human cancers and it is thought that defects in this organelle contribute to genomic instability and malignant transformation. We recently identified and characterized a centrosome-localized protein complex that is comprised of Su48 and Nde1. Disruption of the normal function of these proteins leads to abnormal cell division. To extend our understanding of how this protein complex operates, we sought to identify Nde1-interacting molecules by the yeast two-hybrid screening method. Here, we demonstrate that both Nde1 and Su48 can associate with p78/MCRS1, a protein implicated in cancer development. We found that, whereas the majority of p78 localizes to the nucleus as reported in earlier studies, a fraction of the p78 protein can be detected in the centrosome. Moreover, we determined that a region containing the forkhead-associated domain of p78 is involved in association with Nde1 and Su48, as well as in centrosomal localization. We also provide evidence that the association between p78 and Nde1 is regulated by phosphorylation on Nde1. Furthermore, abrogation of the endogenous p78 function by small interfering RNA knockdown causes cell death and a modest delay in mitosis. These results indicate that a subset of the p78 proteins comprises a component of the centrosome and that p78 is essential for cell viability.


Assuntos
Proteínas de Transporte/metabolismo , Centrossomo/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Linhagem Celular , Sobrevivência Celular/fisiologia , Células HeLa , Humanos , Proteínas Nucleares/fisiologia , Proteínas de Ligação a RNA/fisiologia
10.
Cell Mol Biol (Noisy-le-grand) ; 49(2): 209-16, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12887103

RESUMO

Antibodies are often used to study the molecular basis of physiologic processes. Despite the widespread applications of monoclonal antibodies (mAb) from basic science to successful therapeutics in clinical settings their use is limited. Production of mAb is often cumbersome and creating diverse and therapeutic amounts of useful mAb is difficult. We have developed a methodology to reduce an antibody into much smaller peptidomimetics and have engineered the mimetics for increased serum half life and affinity. The novel species are termed "antibody like binding peptidomimetics" (ABiP). We developed the Anti-Her2/neu peptidomimetic (AHNP) which is a mimic of Herceptin, a mAb used for advanced breast cancer therapy. The AHNP has been used as a defining tool to develop immunodetection probes that exemplify a general process application. AHNP has been expressed as an oligomeric fusion protein with streptavidin. These Herceptin like ABiPs were used to detect the Her2/neu antigen at extremely low concentrations using the immunodetection amplification technique (IDAT) which our laboratory has also developed. A fully developed highly diverse library of ABiPs represents an alternative for panels of monoclonal antibodies and may also be useful for target validation, antigen detection, therapeutics and as a platform for drug development.


Assuntos
Anticorpos/imunologia , Técnicas de Sonda Molecular , Sondas Moleculares , Peptídeos/imunologia , Animais , RNA Polimerases Dirigidas por DNA/metabolismo , Humanos , Receptor ErbB-2/imunologia , Proteínas Virais
12.
Oncogene ; 20(42): 6018-25, 2001 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-11593409

RESUMO

SHP-2 is a ubiquitously expressed non-transmembrane tyrosine phosphatase with two SH2 domains. Multiple reverse-genetic studies have indicated that SHP-2 is a required component for organ and animal development. SHP-2 wild-type and homozygous mutant mouse fibroblast cells in which the N-terminal SH2 domain was target-deleted were used to examine the function of SHP-2 in regulating Phosphatidylinositol 3-Kinase (PI3K) activation by growth factors. In addition, SHP-2 and various mutants were introduced into human glioblastoma cells as well as SHP-2(-/-) mouse fibroblasts. We found that EGF stimulation and EGFR oncoprotein (DeltaEGFR) expression independently induced the co-immunoprecipitation of the p85 subunit of PI3K with SHP-2. Targeted deletion of the N-terminal SH2 domain of SHP-2 severely impaired PDGF- and IGF-induced Akt phosphorylation. Ectopic expression of SHP-2 in U87MG gliobastoma cells elevated EGF-induced Akt phosphorylation, and the effect was abolished by mutation of its N-terminal SH2 domain. Likewise, the reconstitution of SHP-2 expression in the SHP-2(-/-) cells enhanced Akt phosphorylation induced by EGF while rescuing that induced by PDGF and IGF. Further lipid kinase activity assays confirmed that SHP-2 modulation of Akt phosphorylation correlated with its regulation of PI3K activation. Based on these results, we conclude that SHP-2 is required for mediating PI3K/Akt activation, and the N-terminal SH2 domain is critically important for a "positive" role of SHP-2 in regulating PI3K pathway activation.


Assuntos
Substâncias de Crescimento/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Animais , Linhagem Celular , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Mutação , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/genética , Proteínas Proto-Oncogênicas c-akt , Proteínas Tirosina Fosfatases Contendo o Domínio SH2 , Transdução de Sinais , Somatomedinas/farmacologia , Transfecção , Células Tumorais Cultivadas , Domínios de Homologia de src
13.
Oncogene ; 20(34): 4640-9, 2001 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-11498787

RESUMO

We have identified the physical interaction between the Breast Cancer susceptibility gene product BRCA1 and the Hereditary Non-Polyposis Colorectal Cancer (HNPCC) and DNA mismatch repair (MMR) gene product hMSH2, both in vitro and in vivo. The BRCA1-hMSH2 association involved several well-defined regions of both proteins which include the adenosine nucleotide binding domain of hMSH2. Moreover, the interaction of BRCA1 with purified hMSH2-hMSH6 appears to be modulated by adenosine nucleotide much like G protein downstream interaction/signaling is modulated by guanosine nucleotide. BARD1, another BRCA1-interacting protein, was also found to interact with hMSH2. In addition, BRCA1 was found to associate with both hMSH3 and hMSH6, the heterodimeric partners of hMSH2. These observations implicate BRCA1/BARD1 as downstream effectors of the adenosine nucleotide-activated hMSH2-hMSH6 signaling complex, and suggest a global role for BRCA1 in DNA damage processing. The functional interaction between BRCA1 and hMSH2 may provide a partial explanation for the background of gynecological and colorectal cancer in both HNPCC and BRCA1 kindreds, respectively.


Assuntos
Difosfato de Adenosina/farmacologia , Trifosfato de Adenosina/farmacologia , Proteína BRCA1/metabolismo , Proteínas de Ligação a DNA , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor , Ubiquitina-Proteína Ligases , Proteína BRCA1/química , Sítios de Ligação , Proteínas de Transporte/metabolismo , Linhagem Celular , Humanos , Proteína 2 Homóloga a MutS , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/química
14.
DNA Cell Biol ; 20(6): 307-19, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11445002

RESUMO

The erbB receptor family consists of erbB1/epidermal growth factor receptor, erbB2/neu, erbB3, and erbB4, all of which have been implicated in cell proliferation, differentiation, and survival in several tissues. In the nervous system, these family members can function in a trophic capacity for certain subpopulations of neurons and some types of non-neuronal cells. Vestibular sensory epithelial cells and vestibular ganglion neurons are derived from ectodermal otic placode and are essential components of the peripheral vestibular system, the sensory system for balance. Recent studies in mammals suggest that certain ligands of the epidermal growth factor receptor can induce proliferation of vestibular sensory epithelial cells. We now show that vestibular ganglion neurons and vestibular sensory epithelial cells express all four erbB receptors in adult rats. Cultured vestibular ganglion neurons also expressed all four erbB family members and were therefore used to analyze the effects of modulating erbB signaling on differentiated vestibular ganglion neurons. Transforming growth factor-alpha (a ligand for epidermal growth factor receptor) and sensory and motor neuron-derived factor (a ligand for erbB3 and erbB4) promoted vestibular ganglion neuron viability, whereas epidermal growth factor (another ligand for epidermal growth factor receptor) did not. Glial growth factor 2 (another ligand for erbB3 and erbB4) and an antibody that blocks erbB2/neu-mediated signaling inhibited vestibular ganglion neuron viability. Collectively, these observations indicate that erbB signaling regulates the viability of differentiated otic placode-derived cells in mammals and suggest that exogenous modulation of erbB signaling in peripheral vestibular tissues may prove therapeutically useful in peripheral vestibular disorders.


Assuntos
Receptores ErbB/biossíntese , Gânglios Sensitivos/metabolismo , Neurônios/metabolismo , Receptor ErbB-2/biossíntese , Receptor ErbB-3/biossíntese , Vestíbulo do Labirinto/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Receptores ErbB/genética , Receptores ErbB/fisiologia , Imunofluorescência , Gânglios Sensitivos/citologia , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptor ErbB-2/genética , Receptor ErbB-2/fisiologia , Receptor ErbB-3/genética , Receptor ErbB-3/fisiologia , Receptor ErbB-4 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sáculo e Utrículo/metabolismo , Transdução de Sinais , Vestíbulo do Labirinto/citologia , Vestíbulo do Labirinto/crescimento & desenvolvimento
15.
J Med Chem ; 44(16): 2565-74, 2001 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-11472210

RESUMO

Overexpression of the HER2 receptor is observed in about 30% of breast and ovarian cancers and is often associated with an unfavorable prognosis. We have recently designed an anti-HER2 peptide (AHNP) based on the structure of the CDR-H3 loop of the anti-HER2 rhumAb 4D5 and showed that this peptide can mimic some functions of rhumAb 4D5. The peptide disabled HER2 tyrosine kinases in vitro and in vivo similar to the monoclonal antibody (Park, B.-W. et al. Nat. Biotechnol. 2000, 18, 194--198). AHNP has been shown to selectively bind to the extracellular domain of the HER2 receptor with a submicromolar affinity in Biacore assays. In the present paper, we demonstrate that in addition to being a structural and functional mimic of rhumAb 4D5, AHNP can also effectively compete with the antibody for binding to the HER2 receptor indicating a similar binding site for the peptide and the parental antibody. To further develop AHNP as an antitumor agent useful for preclinical trials and as a radiopharmaceutical to be used for tumor imaging, a number of derivatives of AHNP have been designed. Structure--function relationships have been studied using surface plasmon resonance technology. Some of the AHNP analogues have improved binding properties, solubility, and cytotoxic activity relative to AHNP. Residues in the exocyclic region of AHNP appear to be essential for high-affinity binding. Kinetic and equilibrium analysis of peptide-receptor binding for various AHNP analogues revealed a strong correlation between peptide binding characteristics and their biological activity. For AHNP analogues, dissociation rate constants have been shown to be better indicators of peptide biological activity than receptor-binding affinities. This study demonstrates a possibility of mimicking the well-documented antibody effects and its applications in tumor therapy by much smaller antibody-based cyclic peptides with potentially significant therapeutic advantages. Strategies used to improve binding properties of rationally designed AHNP analogues are discussed.


Assuntos
Anticorpos Monoclonais/química , Oligopeptídeos/síntese química , Peptídeos Cíclicos/síntese química , Receptor ErbB-2/efeitos dos fármacos , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Ligação Competitiva , Divisão Celular/efeitos dos fármacos , Desenho de Fármacos , Cinética , Camundongos , Modelos Moleculares , Mimetismo Molecular , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacologia , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/química , Receptor ErbB-2/metabolismo , Solubilidade , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície , Células Tumorais Cultivadas
16.
Proc Natl Acad Sci U S A ; 98(10): 5526-31, 2001 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-11320205

RESUMO

The extracellular domain of p185(c-neu) can be viewed as a complex structure of four subdomains, two of which are cysteine-rich subdomains. We have investigated the contribution of these distinct p185(c-neu) extracellular subdomains to p185/epidermal growth factor receptor (EGFR) heteromer formation and EGF-induced heteromeric signaling. Our studies indicate that at least two separate p185 subdomains, a region spanning subdomains I and II and subdomain IV are involved in association of p185 with the EGFR. We also demonstrated that subdomain IV reduced the heteromeric signaling and transforming activities induced by EGF after associating with EGFR. When 126 aa were deleted from subdomain IV, this small subdomain IV-derived fragment could still lead to heterodimers with EGFR and suppress EGF-induced mitogen-activated protein kinase activation and subsequent transformation abilities. These data provide information about trans-inhibitory mechanisms of mutant p185 species and also indicate that both the entire and a part of subdomain IV may represent a therapeutic target for erbB-overexpressing tumors. Finally, these studies define a basic feature of receptor-receptor associations that are determined by cystine-knot containing subdomains.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Receptor ErbB-2/metabolismo , Transdução de Sinais , Animais , Células COS , Dimerização , Ativação Enzimática , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Receptor ErbB-2/química
17.
Proc Natl Acad Sci U S A ; 98(10): 5497-502, 2001 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-11320219

RESUMO

We have developed an extremely sensitive technique, termed immuno-detection amplified by T7 RNA polymerase (IDAT) that is capable of monitoring proteins, lipids, and metabolites and their modifications at the single-cell level. A double-stranded oligonucleotide containing the T7 promoter is conjugated to an antibody (Ab), and then T7 RNA polymerase is used to amplify RNA from the double-stranded oligonucleotides coupled to the Ab in the Ab-antigen complex. By using this technique, we are able to detect the p185(her2/neu) receptor from the crude lysate of T6-17 cells at 10(-13) dilution, which is 10(9)-fold more sensitive than the conventional ELISA method. Single-chain Fv fragments or complementarity determining region peptides of the Ab also can be substituted for the Ab in IDAT. In a modified protocol, the oligonucleotide has been coupled to an Ab against a common epitope to create a universal detector species. With the linear amplification ability of T7 RNA polymerase, IDAT represents a significant improvement over immuno-PCR in terms of sensitivity and has the potential to provide a robotic platform for proteomics.


Assuntos
Proteínas/análise , Proteoma , Células 3T3 , Animais , Sequência de Bases , Western Blotting , Células Cultivadas , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Hipocampo/citologia , Hipocampo/metabolismo , Camundongos , Processamento de Proteína Pós-Traducional , Proteínas/metabolismo , Ratos , Receptor ErbB-2/análise , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
18.
Mini Rev Med Chem ; 1(1): 5-16, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-12369987

RESUMO

TNFalpha is an inflammatory mediator that is relevant to several autoimmune diseases. Macromolecular inhibitors of TNFalpha have proven therapeutically useful in some preliminary studies. We have developed small molecule TNFalpha antagonist based on the crystal structure of TNF receptor complex. The TNFalpha inhibitor is specific and mediates biological function similar to the inhibitory soluble TNF receptor. This review focuses on development of small molecule anti-TNFalpha mimetics by us and current status of other agents.


Assuntos
Anti-Inflamatórios/química , Anti-Inflamatórios/uso terapêutico , Doenças Autoimunes/tratamento farmacológico , Desenho de Fármacos , Oligopeptídeos/uso terapêutico , Receptores do Fator de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Sítios de Ligação , Humanos , Modelos Moleculares , Oligopeptídeos/química , Conformação Proteica , Receptores do Fator de Necrose Tumoral/química , Relação Estrutura-Atividade , Fator de Necrose Tumoral alfa/química
19.
Proc Natl Acad Sci U S A ; 97(24): 13257-62, 2000 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-11069296

RESUMO

We have found suppressor T cells that inhibit the proliferative response of naive CD4(+) T cells in T cell receptor (TCR) Vbeta8.1 transgenic mice rendered tolerant in vivo by inoculation of Mls-1(a)-positive cells. This suppression was mediated by CD4(+) T cells but not by CD8(+) T cells or double-negative (DN) cells, and splenic CD4(+) T cells from tolerant mice displayed a greater suppression than lymph node CD4(+) T cells. Cell contact was required for efficient suppression, and known inhibitory cytokines such as IL-4, IL-10, and transforming growth factor beta were not involved. Suppressor T cells inhibited IL-2 production by naive CD4(+) T cells, and the addition of exogenous IL-2 diminished the suppressed activity while having little activity on tolerant T cells. Suppression was abolished by the elimination of CD25(+) T cells in the tolerant CD4(+) T cell subset. CD25(+)CD4(+) T cells suppressed the proliferative response of the residual fraction of the nonanergic population, namely, 6C10(+)CD4(+) T cells still present in the tolerant mice. However, 6C10(-)CD4(+) T cells still had reduced reactivity to Mls-1(a) even after CD25(+)CD4(+) T cells were removed and exogenous IL-2 was added. Suppressor cells appear to affect only residual nonanergic cells in situ, thereby facilitating the maintenance of the unresponsive state in vivo. These data provide a framework for understanding suppressor T cells and explain the difficulties and variables in defining their activity in other systems, because suppressor T cells apparently control only a small population of nonanergic cells in the periphery and may be viewed as a homeostatic mechanism.


Assuntos
Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Antígenos Secundários de Estimulação de Linfócitos/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Tolerância Imunológica , Linfonodos/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Baço/imunologia
20.
Oncogene ; 19(35): 3999-4010, 2000 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-10962556

RESUMO

Several growth factors and cytokines, including EGF, are known to induce tyrosine phosphorylation of Signal Regulatory Proteins (SIRPs). Consistent with the idea that increased phosphorylation activates SIRP function, we overexpressed human SIRPalpha1 in U87MG glioblastoma cells in order to examine how SIRPalpha1 modulates EGFR signaling pathways. Endogenous EGFR proteins are overexpressed in U87MG cells and these cells exhibit survival and motility phenotypes that are influenced by EGFR kinase activity. Overexpression of the SIRPalpha1 cDNA diminished EGF-induced phosphoinositide-3-OH kinase (PI3-K) activation in U87MG cells. Reduced EGF-stimulated activation of PI3-K was mediated by interactions between carboxyl terminus of SIRPalpha1 and the Src homology-2 (SH2)-containing phosphotyrosine phosphatase, SHP2. SIRPalpha1 overexpression also reduced the EGF-induced association between SHP2 and the p85 regulatory subunit of PI3-K. Inhibition of transformation and enhanced apoptosis following gamma-irradiation were observed in SIRPalpha1-overexpressing U87MG cells, and enhanced apoptosis was associated with reduced levels of bcl-xL protein. Furthermore, SIRPalpha1-overexpressing U87MG cells displayed reduced cell migration and cell spreading that was mediated by association between SIRPalpha1 and SHP2. However, SIRPalpha1-overexpressing U87MG clonal derivatives exhibited no differences in cell growth or levels of mitogen-activated protein kinase (MAPK) activation. These data reveal a pathway that negatively regulates EGFR-induced PI3-K activation in glioblastoma cells and involves interactions between SHP2 and tyrosine phosphorylated SIRPalpha1. These results also suggest that negative regulation of PI3-K pathway activation by the SIRP family of transmembrane receptors may diminish EGFR-mediated motility and survival phenotypes that contribute to transformation of glioblastoma cells. Oncogene (2000) 19, 3999 - 4010.


Assuntos
Antígenos de Diferenciação , Neoplasias Encefálicas/metabolismo , Receptores ErbB/antagonistas & inibidores , Glioblastoma/metabolismo , Glicoproteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Molécula L1 de Adesão de Célula Nervosa , Moléculas de Adesão de Célula Nervosa/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Receptores Imunológicos , Transdução de Sinais/efeitos dos fármacos , Apoptose/genética , Apoptose/efeitos da radiação , Neoplasias Encefálicas/patologia , Movimento Celular , Transformação Celular Neoplásica/genética , Dano ao DNA , Ativação Enzimática , Receptores ErbB/fisiologia , Raios gama , Glioblastoma/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/antagonistas & inibidores , Moléculas de Adesão de Célula Nervosa/genética , Fenótipo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Transfecção , Proteína bcl-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA