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.
J Med Chem ; 62(24): 11232-11259, 2019 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-31724864

RESUMO

The treatment of tumors driven by overexpression or amplification of MYC oncogenes remains a significant challenge in drug discovery. Here, we present a new strategy toward the inhibition of MYC via the disruption of the protein-protein interaction between MYC and its chromatin cofactor WD Repeat-Containing Protein 5. Blocking the association of these proteins is hypothesized to disrupt the localization of MYC to chromatin, thus disrupting the ability of MYC to sustain tumorigenesis. Utilizing a high-throughput screening campaign and subsequent structure-guided design, we identify small-molecule inhibitors of this interaction with potent in vitro binding affinity and report structurally related negative controls that can be used to study the effect of this disruption. Our work suggests that disruption of this protein-protein interaction may provide a path toward an effective approach for the treatment of multiple tumors and anticipate that the molecules disclosed can be used as starting points for future efforts toward compounds with improved drug-like properties.


Assuntos
Descoberta de Drogas , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Ácido Salicílico/química , Bibliotecas de Moléculas Pequenas/farmacologia , Sulfonamidas/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligação Proteica , Conformação Proteica , Proteínas Proto-Oncogênicas c-myc/metabolismo , Repetições WD40
2.
ACS Chem Biol ; 14(3): 325-331, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30735352

RESUMO

Activating mutations in RAS can lead to oncogenesis by enhancing downstream signaling, such as through the MAPK and PI3K pathways. Therefore, therapeutically targeting RAS may perturb multiple signaling pathways simultaneously. One method for modulating RAS signaling is to target the activity of the guanine nucleotide exchange factor SOS1. Our laboratory has discovered compounds that bind to SOS1 and activate RAS. Interestingly, these SOS1 agonist compounds elicit biphasic modulation of ERK phosphorylation and simultaneous inhibition of AKT phosphorylation levels. Here, we utilized multiple chemically distinct compounds to elucidate whether these effects on MAPK and PI3K signaling by SOS1 agonists were mechanistically linked. In addition, we used CRISPR/Cas9 gene-editing to generate clonally derived SOS1 knockout cells and identified a potent SOS1 agonist that rapidly elicited on-target molecular effects at substantially lower concentrations than those causing off-target effects. Our findings will allow us to further define the on-target utility of SOS1 agonists.


Assuntos
Benzimidazóis/química , Indóis/química , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Quinazolinas/química , Proteína SOS1/agonistas , Benzimidazóis/metabolismo , Proteína 9 Associada à CRISPR/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Edição de Genes , Humanos , Indóis/metabolismo , Quinazolinas/metabolismo
3.
J Med Chem ; 61(19): 8875-8894, 2018 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-30205005

RESUMO

Son of sevenless homologue 1 (SOS1) is a guanine nucleotide exchange factor that catalyzes the exchange of GDP for GTP on RAS. In its active form, GTP-bound RAS is responsible for numerous critical cellular processes. Aberrant RAS activity is involved in ∼30% of all human cancers; hence, SOS1 is an attractive therapeutic target for its role in modulating RAS activation. Here, we describe a new series of benzimidazole-derived SOS1 agonists. Using structure-guided design, we discovered small molecules that increase nucleotide exchange on RAS in vitro at submicromolar concentrations, bind to SOS1 with low double-digit nanomolar affinity, rapidly enhance cellular RAS-GTP levels, and invoke biphasic signaling changes in phosphorylation of ERK 1/2. These compounds represent the most potent series of SOS1 agonists reported to date.


Assuntos
Benzimidazóis/farmacologia , Descoberta de Drogas/normas , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteína SOS1/agonistas , Proteína SOS1/metabolismo , Benzimidazóis/química , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Troca do Nucleotídeo Guanina/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Fosforilação , Conformação Proteica , Proteínas Proto-Oncogênicas p21(ras)/química , Relação Estrutura-Atividade
4.
ACS Med Chem Lett ; 9(9): 941-946, 2018 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-30258545

RESUMO

Proteins in the RAS family are important regulators of cellular signaling and, when mutated, can drive cancer pathogenesis. Despite considerable effort over the last 30 years, RAS proteins have proven to be recalcitrant therapeutic targets. One approach for modulating RAS signaling is to target proteins that interact with RAS, such as the guanine nucleotide exchange factor (GEF) son of sevenless homologue 1 (SOS1). Here, we report hit-to-lead studies on quinazoline-containing compounds that bind to SOS1 and activate nucleotide exchange on RAS. Using structure-based design, we refined the substituents attached to the quinazoline nucleus and built in additional interactions not present in the initial HTS hit. Optimized compounds activate nucleotide exchange at single-digit micromolar concentrations in vitro. In HeLa cells, these quinazolines increase the levels of RAS-GTP and cause signaling changes in the mitogen-activated protein kinase/extracellular regulated kinase (MAPK/ERK) pathway.

5.
J Med Chem ; 61(14): 6002-6017, 2018 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-29856609

RESUMO

Deregulated RAS activity, often the result of mutation, is implicated in approximately 30% of all human cancers. Despite this statistic, no clinically successful treatment for RAS-driven tumors has yet been developed. One approach for modulating RAS activity is to target and affect the activity of proteins that interact with RAS, such as the guanine nucleotide exchange factor (GEF) son of sevenless homologue 1 (SOS1). Here, we report on structure-activity relationships (SAR) in an indole series of compounds. Using structure-based design, we systematically explored substitution patterns on the indole nucleus, the pendant amino acid moiety, and the linker unit that connects these two fragments. Best-in-class compounds activate the nucleotide exchange process at submicromolar concentrations in vitro, increase levels of active RAS-GTP in HeLa cells, and elicit signaling changes in the mitogen-activated protein kinase-extracellular regulated kinase (MAPK-ERK) pathway, resulting in a decrease in pERK1/2T202/Y204 protein levels at higher compound concentrations.


Assuntos
Desenho de Fármacos , Indóis/química , Indóis/farmacologia , Piperidinas/química , Proteína SOS1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas ras/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Conformação Proteica , Proteína SOS1/química , Relação Estrutura-Atividade , Proteínas ras/química
6.
Mol Cancer Ther ; 17(5): 1051-1060, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29440291

RESUMO

Oncogenic mutation of RAS results in aberrant cellular signaling and is responsible for more than 30% of all human tumors. Therefore, pharmacologic modulation of RAS has attracted great interest as a therapeutic strategy. Our laboratory has recently discovered small molecules that activate Son of Sevenless (SOS)-catalyzed nucleotide exchange on RAS and inhibit downstream signaling. Here, we describe how pharmacologically targeting SOS1 induced biphasic modulation of RAS-GTP and ERK phosphorylation levels, which we observed in a variety of cell lines expressing different RAS-mutant isoforms. We show that compound treatment caused an increase in phosphorylation at ERK consensus motifs on SOS1 that was not observed with the expression of a non-phosphorylatable S1178A SOS1 mutant or after pretreatment with an ERK inhibitor. Phosphorylation at S1178 on SOS1 is known to inhibit the association between SOS1 and GRB2 and disrupt SOS1 membrane localization. Consistent with this, we show that wild-type SOS1 and GRB2 dissociated in a time-dependent fashion in response to compound treatment, and conversely, this interaction was enhanced with the expression of an S1178A SOS1 mutant. Furthermore, in cells expressing either S1178A SOS1 or a constitutively membrane-bound CAAX box tagged SOS1 mutant, we observed elevated RAS-GTP levels over time in response to compound, as compared with the biphasic changes in RAS-GTP exhibited in cells expressing wild-type SOS1. These results suggest that small molecule targeting of SOS1 can elicit a biphasic modulation of RAS-GTP and phospho-ERK levels through negative feedback on SOS1 that regulates the interaction between SOS1 and GRB2. Mol Cancer Ther; 17(5); 1051-60. ©2018 AACR.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Retroalimentação Fisiológica/efeitos dos fármacos , Proteína SOS1/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas ras/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Estrutura Molecular , Mutação de Sentido Incorreto , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteína SOS1/genética , Bibliotecas de Moléculas Pequenas/química , Proteínas ras/genética
7.
Anal Biochem ; 548: 44-52, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29444450

RESUMO

K-RAS is mutated in approximately 30% of human cancers, resulting in increased RAS signaling and tumor growth. Thus, RAS is a highly validated therapeutic target, especially in tumors of the pancreas, lung and colon. Although directly targeting RAS has proven to be challenging, it may be possible to target other proteins involved in RAS signaling, such as the guanine nucleotide exchange factor Son of Sevenless (SOS). We have previously reported on the discovery of small molecules that bind to SOS1, activate SOS-mediated nucleotide exchange on RAS, and paradoxically inhibit ERK phosphorylation (Burns et al., PNAS, 2014). Here, we describe the discovery of additional, structurally diverse small molecules that also bind to SOS1 in the same pocket and elicit similar biological effects. We tested >160,000 compounds in a fluorescence-based assay to assess their effects on SOS-mediated nucleotide exchange. X-Ray structures revealed that these small molecules bind to the CDC25 domain of SOS1. Compounds that elicited high levels of nucleotide exchange activity in vitro increased RAS-GTP levels in cells, and inhibited phospho ERK levels at higher treatment concentrations. The identification of structurally diverse SOS1 binding ligands may assist in the discovery of new molecules designed to target RAS-driven tumors.


Assuntos
Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteína SOS1/metabolismo , Células HeLa , Humanos , Proteínas Proto-Oncogênicas p21(ras)/química , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteína SOS1/química , Proteína SOS1/genética
9.
Ann Am Thorac Soc ; 12(4): 505-11, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25741996

RESUMO

RATIONALE: Although expert communication between intensive care unit clinicians with patients or surrogates improves patient- and family-centered outcomes, fellows in critical care medicine do not feel adequately trained to conduct family meetings. OBJECTIVES: We aimed to develop, implement, and evaluate a communication skills program that could be easily integrated into a U.S. critical care fellowship. METHODS: We developed four simulation cases that provided communication challenges that critical care fellows commonly face. For each case, we developed a list of directly observable tasks that could be used by faculty to evaluate fellows during each simulation. We developed a didactic curriculum of lectures/case discussions on topics related to palliative care, end-of-life care, communication skills, and bioethics; this month-long curriculum began and ended with the fellows leading family meetings in up to two simulated cases with direct observation by faculty who were not blinded to the timing of the simulation. Our primary measures of effectiveness were the fellows' self-reported change in comfort with leading family meetings after the program was completed and the quality of the communication as measured by the faculty evaluators during the family meeting simulations at the end of the month. MEASUREMENTS AND MAIN RESULTS: Over 3 years, 31 critical care fellows participated in the program, 28 of whom participated in 101 family meeting simulations with direct feedback by faculty facilitators. Our trainees showed high rates of information disclosure during the simulated family meetings. During the simulations done at the end of the month compared with those done at the beginning, our fellows showed significantly improved rates in: (1) verbalizing an agenda for the meeting (64 vs. 41%; Chi-square, 5.27; P = 0.02), (2) summarizing what will be done for the patient (64 vs. 39%; Chi-square, 6.21; P = 0.01), and (3) providing a follow-up plan (60 vs. 37%; Chi-square, 5.2; P = 0.02). More than 95% of our participants (n = 27) reported feeling "slightly" or "much" more comfortable with discussing foregoing life-sustaining treatment and leading family discussions after the month-long curriculum. CONCLUSIONS: A communication skills program can be feasibly integrated into a critical care training program and is associated with improvements in fellows' skills and comfort with leading family meetings.


Assuntos
Competência Clínica , Cuidados Críticos , Currículo , Bolsas de Estudo/métodos , Relações Médico-Paciente , Relações Profissional-Família , Pneumologia/educação , Adulto , Comunicação , Estado Terminal , Feminino , Humanos , Masculino , Desenvolvimento de Programas
10.
ANZ J Surg ; 82(10): 671-4, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22946902

RESUMO

BACKGROUND: There is relatively little in the medical literature relating to complaints about the healthcare process. The aim of this study was to report the frequency and content of patient complaints against a University Hospital Surgical Department. In particular, the study aimed to relate the number of complaints to the number of health-care episodes and to determine the frequency of patient safety incidents and subsequent medico-legal action. METHODS: Retrospective interrogation of a prospectively maintained Complaints Department database at a University Hospital for the calendar year 2009. RESULTS: Complaints relating to 360 aspects of the health-care journey in 113 patients were made. This translated into one complaint per 400 health-care episodes. Concerns about clinical care were cited in 31%, delays in the health-care process in 30%, communication issues in 19%, the institutional environment in 8% and poor discharge planning in 6%. Overall, 16 complaints (4%) were raised as patient safety incidents. Eighty-three per cent of complaints were addressed by a telephone conversation or a single letter response, 13% by a face-to-face meeting. Two per cent resulted in subsequent medico-legal action. CONCLUSIONS: Although perceived in a negative way by health-care professionals, only 1 in 400 health-care episodes resulted in a complaint. Only a small number related to patient safety incidents or resulted in medico-legal instructions. Attention should focus on developing effective strategies to improve patient satisfaction with all aspects of the patient journey.


Assuntos
Hospitais Universitários , Auditoria Médica , Erros Médicos/estatística & dados numéricos , Segurança do Paciente/estatística & dados numéricos , Satisfação do Paciente/estatística & dados numéricos , Relações Profissional-Paciente , Centro Cirúrgico Hospitalar , Comunicação , Humanos , Erros Médicos/legislação & jurisprudência , Segurança do Paciente/legislação & jurisprudência , Estudos Retrospectivos , Reino Unido
11.
J Biol Chem ; 287(11): 8552-60, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22219183

RESUMO

Marinesco-Sjögren syndrome (MSS) is an autosomal recessive, neurodegenerative, multisystem disorder characterized by severe phenotypes developing in infancy. Recently, mutations in the endoplasmic reticulum (ER)-associated co-chaperone SIL1/BAP were identified to be the major cause of MSS. SIL1 acts as a nucleotide exchange factor for BiP, the ER Hsp70 orthologue, which plays an essential role in the folding and assembly of nascent polypeptide chains in the ER. SIL1 facilitates the release of BiP from unfolded protein substrates, enabling the subsequent folding and transport of the protein. Although most mutations leading to MSS result in deletion of the majority of the protein, three separate mutations have been identified that disrupt only the last five or six amino acids of the protein, which were assumed to encode a divergent ER retention motif. This study presents an in depth analysis of two of these mutants and reveals that the phenotype in the affected individuals is not likely to be due to depletion of SIL1 from the ER via secretion. Instead, our analyses show that the mutant proteins are particularly unstable and either form large aggregates in the ER or are rapidly degraded via the proteasome. In agreement with our findings, homology modeling suggests that the very C-terminal residues of SIL1 play a role in its structural integrity rather than its localization. These new insights might be a first step toward a possible pharmacological treatment of certain types of MSS by specifically stabilizing the mutant SIL1 protein.


Assuntos
Retículo Endoplasmático/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mutação , Proteólise , Degenerações Espinocerebelares/metabolismo , Motivos de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Retículo Endoplasmático/genética , Retículo Endoplasmático/patologia , Chaperona BiP do Retículo Endoplasmático , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Estabilidade Proteica , Degenerações Espinocerebelares/genética , Degenerações Espinocerebelares/patologia
12.
Ophthalmology ; 117(2): 298-302, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19969368

RESUMO

OBJECTIVE: To compare visual acuity (VA) outcomes after bevacizumab or ranibizumab treatment for AMD. DESIGN: Comparative, retrospective case series. PARTICIPANTS: We followed 452 patients in a retrospective study of exudative AMD treated with anti-vascular endothelial growth factor drugs; 324 patients were treated with bevacizumab and 128 patients with ranibizumab. METHODS: All treatment-naïve patients who received either bevacizumab or ranibizumab were followed for 1 year. Baseline characteristics and VA were recorded using standard descriptive statistics. MAIN OUTCOME MEASURES: Visual acuity. RESULTS: At 12 months, the distribution of VA improved in both groups with 22.9% of bevacizumab and 25.0% of ranibizumab attaining >or=20/40. Improvement in vision was observed in 27.3% of the bevacizumab group and 20.2% of the ranibizumab group. The mean number of injections at 12 months was 4.4 for bevacizumab and 6.2 for ranibizumab. There were 8 (2%) deaths in the bevacizumab group and 4 (3%) in the ranibizumab group. Two patients developed endophthalmitis in the bevacizumab group and the ranibizumab group. The bevacizumab group had slightly worse acuity at baseline, but both groups showed improvement and stability of vision over time. CONCLUSIONS: Both treatments seem to be effective in stabilizing VA loss. There was no difference in VA outcome between the 2 treatment groups. Because the study is a nonrandomized comparison, selection bias could mask a true treatment difference. Results from the Comparison of the Age-related Macular Degeneration Treatment Trials will provide more definitive information about the comparative effectiveness of these drugs.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Neovascularização de Coroide/tratamento farmacológico , Degeneração Macular/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais Humanizados , Bevacizumab , Neovascularização de Coroide/fisiopatologia , Feminino , Humanos , Injeções , Degeneração Macular/fisiopatologia , Masculino , Pessoa de Meia-Idade , Ranibizumab , Estudos Retrospectivos , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Acuidade Visual/fisiologia , Corpo Vítreo
13.
Diabetes ; 57(11): 3034-44, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18591394

RESUMO

OBJECTIVE: Cytokines contribute to beta-cell destruction in type 1 diabetes. Endoplasmic reticulum (ER) stress-mediated apoptosis has been proposed as a mechanism for beta-cell death. We tested whether ER stress was necessary for cytokine-induced beta-cell death and also whether ER stress gene activation was present in beta-cells of the NOD mouse model of type 1 diabetes. RESEARCH DESIGN AND METHODS: INS-1 beta-cells or rat islets were treated with the chemical chaperone phenyl butyric acid (PBA) and exposed or not to interleukin (IL)-1beta and gamma-interferon (IFN-gamma). Small interfering RNA (siRNA) was used to silence C/EBP homologous protein (CHOP) expression in INS-1 beta-cells. Additionally, the role of ER stress in lipid-induced cell death was assessed. RESULTS: Cytokines and palmitate triggered ER stress in beta-cells as evidenced by increased phosphorylation of PKR-like ER kinase (PERK), eukaryotic initiation factor (EIF)2alpha, and Jun NH(2)-terminal kinase (JNK) and increased expression of activating transcription factor (ATF)4 and CHOP. PBA treatment attenuated ER stress, but JNK phosphorylation was reduced only in response to palmitate, not in response to cytokines. PBA had no effect on cytokine-induced cell death but was associated with protection against palmitate-induced cell death. Similarly, siRNA-mediated reduction in CHOP expression protected against palmitate- but not against cytokine-induced cell death. In NOD islets, mRNA levels of several ER stress genes were reduced (ATF4, BiP [binding protein], GRP94 [glucose regulated protein 94], p58, and XBP-1 [X-box binding protein 1] splicing) or unchanged (CHOP and Edem1 [ER degradation enhancer, mannosidase alpha-like 1]). CONCLUSIONS: While both cytokines and palmitate can induce ER stress, our results suggest that, in contrast to lipoapoptosis, the PERK-ATF4-CHOP ER stress-signaling pathway is not necessary for cytokine-induced beta-cell death.


Assuntos
Citocinas/farmacologia , Retículo Endoplasmático/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator 4 Ativador da Transcrição/metabolismo , Animais , Western Blotting , Ácido Butírico/farmacologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Retículo Endoplasmático/efeitos dos fármacos , Fator de Iniciação 2 em Eucariotos/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Palmitatos/farmacologia , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , Ratos , Ratos Wistar , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/fisiologia , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , eIF-2 Quinase/metabolismo
14.
Blood ; 101(5): 1919-27, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12424201

RESUMO

Anaplastic Large Cell Lymphomas (ALCLs) carry translocations in which the anaplastic lymphoma kinase (ALK) gene is juxtaposed to various genes, the most common of which is the NPM/B23 gene. ALK fusion proteins result in the constitutive activation of ALK tyrosine kinase, thereby enhancing proliferation and increasing cell survival. A direct role for NPM-ALK in cellular transformation has been shown in vitro with immortalized cell lines and in vivo using retroviral transfer experiments. Nonetheless, there is no direct evidence of its oncogenic potential in T lymphocytes, which represent the most common target of ALK chimeras. Here, we describe a new mouse model of lymphomagenesis in which human NPM-ALK transcription was targeted to T cells. NPM-ALK transgenic (Tg) mice were born with the expected mendelian distribution, normal lymphoid organs, and a normal number and proportion of helper and suppressor T cells. However, after a short period of latency, all NPM-ALK Tg mice developed malignant lymphoproliferative disorders (mean survival, 18 weeks). NPM-ALK Tg thymic lymphomas displayed a T-cell phenotype characteristic of immature thymocytes and frequently coexpressed surface CD30. A subset of the NPM-ALK Tg mice also developed clonal B-cell plasma cell neoplasms. These tumors arose in peripheral lymphoid organs (plasmacytomas) or within the bone marrow and often led to peripheral neuropathies and limb paralysis. Our NPM-ALK Tg mice are a suitable model to dissect the molecular mechanisms of ALK-mediated transformation and to investigate the efficacy of new therapeutic approaches for the treatment of human ALCL in vivo.


Assuntos
Transformação Celular Neoplásica/genética , Linfoma de Células T/genética , Plasmocitoma/genética , Proteínas Tirosina Quinases/fisiologia , Neoplasias do Timo/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Janus Quinase 3 , Tecido Linfoide/patologia , Linfoma de Células B/genética , Linfoma de Células B/patologia , Linfoma de Células T/patologia , Transtornos Linfoproliferativos/genética , Camundongos , Camundongos Transgênicos , Fosforilação , Plasmocitoma/patologia , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Fator de Transcrição STAT3 , Transdução de Sinais , Neoplasias do Timo/patologia , Transativadores/metabolismo
15.
Oncogene ; 21(7): 1038-47, 2002 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-11850821

RESUMO

The anaplastic lymphoma kinase (ALK) gene is characteristically translocated in Anaplastic Large Cell Lymphomas (ALCL) and the juxtaposition of the ALK gene to multiple partners results in its constitutive protein tyrosine kinase activity. We show here that expression of activated ALK induces the constitutive phosphorylation of Stat3 in transfected cells as well as in primary human ALCLs. Furthermore, immunohistochemical studies demonstrate that among distinct human B and T cell lymphomas, activation of Stat3 nuclear translocation is uniquely associated with ALK expression. NPM-ALK also binds and activates Jak3; however, Jak3 is not required for Stat3 activation or for cell transformation in vitro. Moreover, src family kinases are not necessary for NPM-ALK-mediated Stat3 activation or transformation, suggesting that Stat3 may be phosphorylated directly by ALK. To evaluate relevant targets of ALK-activated Stat3, we investigated the regulation of the anti-apoptotic protein Bcl-x(L) and its role in cell survival in NPM-ALK positive cells. NPM-ALK expression caused enhanced Bcl-x(L) transcription, largely mediated by Stat3. Increased expression of Bcl-x(L) provided sufficient anti-apoptotic signals to protect cells from treatment with specific inhibitors of the Jaks/Stat pathway or the Brc-Abl kinase. These studies support a pathogenic mechanism whereby stimulation of anti-apoptotic signals through activation of Stat3 contributes to the successful outgrowth of ALK positive tumor cells.


Assuntos
Apoptose , Proteínas de Ligação a DNA/metabolismo , Linfoma Anaplásico de Células Grandes/metabolismo , Proteínas Tirosina Quinases/fisiologia , Transativadores/metabolismo , Quinase do Linfoma Anaplásico , Animais , Linhagem Celular , Proteínas de Ligação a DNA/imunologia , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imuno-Histoquímica , Janus Quinase 3 , Linfoma Anaplásico de Células Grandes/enzimologia , Linfoma Anaplásico de Células Grandes/patologia , Fosforilação , Proteínas Tirosina Quinases/análise , Proteínas Tirosina Quinases/imunologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Receptores Proteína Tirosina Quinases , Fator de Transcrição STAT3 , Transativadores/imunologia , Células Tumorais Cultivadas , Regulação para Cima , Proteína bcl-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...