Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Ther Adv Med Oncol ; 15: 17588359231151840, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36756142

RESUMO

Background: Abemaciclib is the first and only cyclin-dependent kinases 4 and 6 inhibitor approved for adjuvant treatment of hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-), node-positive, and high-risk early breast cancer (EBC), with indications varying by geography. Premenopausal patients with HR+, HER2- tumors may have different tumor biology and treatment response compared to postmenopausal patients. Objectives: We describe the efficacy and safety of abemaciclib plus endocrine therapy (ET) for the large subgroup of premenopausal patients with HR+, HER2- EBC in monarchE. Design: Randomized patients (1:1) received adjuvant ET with or without abemaciclib for 2 years plus at least 3 additional years of ET as clinically indicated. Methods: Patients were stratified by menopausal status (premenopausal versus postmenopausal) at diagnosis. Standard ET (tamoxifen or aromatase inhibitor) with or without gonadotropin-releasing hormone agonist was determined by physician's choice. Invasive disease-free survival (IDFS) and distant relapse-free survival (DRFS) by menopausal status were assessed at data cutoff on 1 April 2021 (median follow-up of 27 months). Results: Among randomized patients, 2451 (43.5%) were premenopausal and 3181 (56.4%) were postmenopausal. The choice of ET for premenopausal patients varied considerably between countries. Treatment benefit was consistent across menopausal status, with a numerically greater effect size in premenopausal patients. For premenopausal patients, abemaciclib with ET resulted in a 42.2% and 40.3% reduction in the risk of developing IDFS and DRFS events, respectively. Absolute improvement at 3 years was 5.7% for IDFS and 4.4% for DRFS rates. Safety profile for premenopausal patients was consistent with the overall safety population. Conclusion: Abemaciclib with ET demonstrated clinically meaningful treatment benefit for IDFS and DRFS versus ET alone regardless of menopausal status and first ET, with a numerically greater benefit in the premenopausal compared to the postmenopausal population. Safety data in premenopausal patients are consistent with the overall safety profile of abemaciclib.

2.
Oncologist ; 28(1): e77-e81, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36342342

RESUMO

The monarchE Cohort 1 patient population was enrolled based on high-risk clinicopathological features that can easily be identified as part of routine clinical breast cancer evaluation. Efficacy data from Cohort 1 demonstrate substantial evidence of benefit for adjuvant abemaciclib+ET in patients with HR+, HER2- early breast cancer at high risk of recurrence (ClinicalTrials.gov: NCT03155997 [monarchE]).


Assuntos
Neoplasias da Mama , Receptor ErbB-2 , Feminino , Humanos , Aminopiridinas/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzimidazóis/uso terapêutico , Neoplasias da Mama/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Receptor ErbB-2/uso terapêutico
4.
Nat Commun ; 10(1): 5649, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31827082

RESUMO

Clonal hematopoiesis of indeterminate potential (CHIP) increases with age and is associated with increased risks of hematological malignancies. While TP53 mutations have been identified in CHIP, the molecular mechanisms by which mutant p53 promotes hematopoietic stem and progenitor cell (HSPC) expansion are largely unknown. Here we discover that mutant p53 confers a competitive advantage to HSPCs following transplantation and promotes HSPC expansion after radiation-induced stress. Mechanistically, mutant p53 interacts with EZH2 and enhances its association with the chromatin, thereby increasing the levels of H3K27me3 in genes regulating HSPC self-renewal and differentiation. Furthermore, genetic and pharmacological inhibition of EZH2 decreases the repopulating potential of p53 mutant HSPCs. Thus, we uncover an epigenetic mechanism by which mutant p53 drives clonal hematopoiesis. Our work will likely establish epigenetic regulator EZH2 as a novel therapeutic target for preventing CHIP progression and treating hematological malignancies with TP53 mutations.


Assuntos
Epigênese Genética , Doenças Hematológicas/metabolismo , Hematopoese , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Doenças Hematológicas/genética , Doenças Hematológicas/fisiopatologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Metilação , Camundongos Endogâmicos C57BL , Mutação , Ligação Proteica
5.
Sci Rep ; 9(1): 15803, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31676795

RESUMO

Colorectal cancer (CRC) is the fourth leading cause of cancer-related deaths worldwide. Liver metastasis is the major cause of CRC patient mortality, occurring in 60% patients with no effective therapies. Although studies have indicated the role of miRNAs in CRC, an in-depth miRNA expression analysis is essential to identify clinically relevant miRNAs and understand their potential in targeting liver metastasis. Here we analyzed miRNA expressions in 405 patient tumors from publicly available colorectal cancer genome sequencing project database. Our analyses showed miR-132, miR-378f, miR-605 and miR-1976 to be the most significantly downregulated miRNAs in primary and CRC liver metastatic tissues, and CRC cell lines. Observations in CRC cell lines indicated that ectopic expressions of miR-378f, -605 and -1976 suppress CRC cell proliferation, anchorage independent growth, metastatic potential, and enhance apoptosis. Consistently, CRC patients with higher miR-378f and miR-1976 levels exhibited better survival. Together, our data suggests an anti-tumorigenic role of these miRNAs in CRC and warrant future in vivo evaluation of the molecules for developing biomarkers or novel therapeutic strategies.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Hepáticas/secundário , MicroRNAs/metabolismo , Neoplasias Colorretais/patologia , Feminino , Humanos , Neoplasias Hepáticas/genética , Masculino
7.
J Surg Oncol ; 118(3): 431-439, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30076741

RESUMO

BACKGROUND AND OBJECTIVES: Hepatocellular carcinoma (HCC) in patients with hepatitis B virus (HBV) exhibit lower tumor microRNA-26a (miR-26a) expression which is associated with worse outcomes. It is unknown if similar miR-26a loss occurs in HCC developed in other liver diseases. We examined tumor miR-26a expression and its impact on recurrence and mortality in a North American HCC cohort. METHODS: MiR-26a levels from tumor and surrounding nontumor liver tissue in 186 subjects were collected. We defined lower tumor expression of miR-26a as <1-fold that of the adjacent nontumor liver tissue. RESULTS: Viral hepatitis (42%; 40% hepatitis C and 2% HBV), alcohol (19%), and nonalcoholic fatty liver disease (NAFLD) (18%) were the most common causes of liver disease. The prevalence of lower tumor miR-26a expression was 68%, and it was evident in HCCs arising in all etiologies (viral hepatitis 60%, alcohol 61%, and NAFLD 76%). Subjects with lower tumor miR-26a expression had significantly higher tumor recurrence (hazard ratio [HR], 2.45; 95% confidence interval [CI], 1.18 to 5.1; P = 0.016) and higher mortality of borderline significance (HR, 1.51; 95% CI, 0.94 to 2.41; P = 0.086). CONCLUSION: Reduced miR-26a expression is a common phenomenon in HCC arising in North American patients with different underlying liver diseases and may increase recurrence and mortality after surgery.


Assuntos
Biomarcadores Tumorais/sangue , Carcinoma Hepatocelular/cirurgia , Regulação Neoplásica da Expressão Gênica , Hepatectomia/métodos , Neoplasias Hepáticas/cirurgia , MicroRNAs/sangue , Recidiva Local de Neoplasia/sangue , Idoso , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/patologia , Feminino , Seguimentos , Humanos , Neoplasias Hepáticas/patologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/genética , Prognóstico , Transdução de Sinais , Taxa de Sobrevida
8.
Oncotarget ; 8(50): 87607-87622, 2017 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-29152105

RESUMO

Acute myeloid leukemia (AML) is a devastating illness which carries a very poor prognosis, with most patients living less than 18 months. Leukemia relapse may occur because current therapies eliminate proliferating leukemia cells but fail to eradicate quiescent leukemia-initiating cells (LICs) that can reinitiate the disease after a period of latency. While we demonstrated that p53 target gene Necdin maintains hematopoietic stem cell (HSC) quiescence, its roles in LIC quiescence and response to chemotherapy are unclear. In this study, we utilized two well-established murine models of human AML induced by MLL-AF9 or AML1-ETO9a to determine the role of Necdin in leukemogenesis. We found that loss of Necdin decreased the number of functional LICs and enhanced myeloid differentiation in vivo, leading to delayed development of leukemia induced by MLL-AF9. Importantly, Necdin null LICs expressing MLL-AF9 were less quiescent than wild-type LICs. Further, loss of Necdin enhanced the response of MLL-AF9+ leukemia cells to chemotherapy treatment, manifested by decreased viability and enhanced apoptosis. We observed decreased expression of Bcl2 and increased expression of p53 and its target gene Bax in Necdin null leukemia cells following chemotherapy treatment, indicating that p53-dependent apoptotic pathways may be activated in the absence of Necdin. In addition, we found that loss of Necdin decreased the engraftment of AML1-ETO9a+ hematopoietic stem and progenitor cells in transplantation assays. However, Necdin-deficiency did not affect the response of AML1-ETO9a+ hematopoietic cells to chemotherapy treatment. Thus, Necdin regulates leukemia-initiating cell quiescence and chemotherapy response in a context-dependent manner. Our findings suggest that pharmacological inhibition of Necdin may hold potential as a novel therapy for leukemia patients with MLL translocations.

9.
Stem Cells ; 35(4): 1053-1064, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28009085

RESUMO

The molecular pathways regulating lymphoid priming, fate, and development of multipotent bone marrow hematopoietic stem and progenitor cells (HSPCs) that continuously feed thymic progenitors remain largely unknown. While Notch signal is indispensable for T cell specification and differentiation, the downstream effectors are not well understood. PRL2, a protein tyrosine phosphatase that regulates hematopoietic stem cell proliferation and self-renewal, is highly expressed in murine thymocyte progenitors. Here we demonstrate that protein tyrosine phosphatase PRL2 and receptor tyrosine kinase c-Kit are critical downstream targets and effectors of the canonical Notch/RBPJ pathway in early T cell progenitors. While PRL2 deficiency resulted in moderate defects of thymopoiesis in the steady state, de novo generation of T cells from Prl2 null hematopoietic stem cells was significantly reduced following transplantation. Prl2 null HSPCs also showed impaired T cell differentiation in vitro. We found that Notch/RBPJ signaling upregulated PRL2 as well as c-Kit expression in T cell progenitors. Further, PRL2 sustains Notch-mediated c-Kit expression and enhances stem cell factor/c-Kit signaling in T cell progenitors, promoting effective DN1-DN2 transition. Thus, we have identified a critical role for PRL2 phosphatase in mediating Notch and c-Kit signals in early T cell progenitors. Stem Cells 2017;35:1053-1064.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores Notch/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Linfócitos T/citologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Camundongos Endogâmicos C57BL , Modelos Biológicos , Transdução de Sinais , Timo/metabolismo , Regulação para Cima
10.
Sci Rep ; 5: 11450, 2015 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-26095125

RESUMO

Dense fibrotic stroma associated with pancreatic ductal adenocarcinoma (PDAC) is a major obstacle for drug delivery to the tumor bed and plays a crucial role in pancreatic cancer progression. Current, anti-stromal therapies have failed to improve tumor response to chemotherapy and patient survival. Furthermore, recent studies show that stroma impedes tumor progression, and its complete ablation accelerates PDAC progression. In an effort to understand the molecular mechanisms associated with tumor-stromal interactions, using in vitro and in vivo models and PDAC patient biopsies, we show that the loss of miR-29 is a common phenomenon of activated pancreatic stellate cells (PSCs)/fibroblasts, the major stromal cells responsible for fibrotic stromal reaction. Loss of miR-29 is correlated with a significant increase in extracellular matrix (ECM) deposition, a major component in PDAC stroma. Our in vitro miR-29 gain/loss-of-function studies document the role of miR-29 in PSC-mediated ECM stromal protein accumulation. Overexpression of miR-29 in activated stellate cells reduced stromal deposition, cancer cell viability, and cancer growth in co-culture. Furthermore, the loss of miR-29 in TGF-ß1 activated PSCs is SMAD3 dependent. These results provide insights into the mechanistic role of miR-29 in PDAC stroma and its potential use as a therapeutic agent to target PDAC.


Assuntos
Carcinoma Ductal Pancreático/genética , Matriz Extracelular/metabolismo , Fibrose/patologia , MicroRNAs/genética , Neoplasias Pancreáticas/genética , Animais , Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Ativação Enzimática/genética , Matriz Extracelular/genética , Fibroblastos/citologia , Fibrose/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/tratamento farmacológico , Células Estreladas do Pâncreas/citologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral
11.
J Biol Chem ; 290(7): 3894-909, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25538234

RESUMO

Macrophages are vital to innate immunity and express pattern recognition receptors and integrins for the rapid detection of invading pathogens. Stimulation of Dectin-1 and complement receptor 3 (CR3) activates Erk- and Akt-dependent production of reactive oxygen species (ROS). Shp2, a protein-tyrosine phosphatase encoded by Ptpn11, promotes activation of Ras-Erk and PI3K-Akt and is crucial for hematopoietic cell function; however, no studies have examined Shp2 function in particulate-stimulated ROS production. Maximal Dectin-1-stimulated ROS production corresponded kinetically to maximal Shp2 and Erk phosphorylation. Bone marrow-derived macrophages (BMMs) from mice with a conditionally deleted allele of Ptpn11 (Shp2(flox/flox);Mx1Cre+) produced significantly lower ROS levels compared with control BMMs. Although YFP-tagged phosphatase dead Shp2-C463A was strongly recruited to the early phagosome, its expression inhibited Dectin-1- and CR3-stimulated phospho-Erk and ROS levels, placing Shp2 phosphatase function and Erk activation upstream of ROS production. Further, BMMs expressing gain of function Shp2-D61Y or Shp2-E76K and peritoneal exudate macrophages from Shp2D61Y/+;Mx1Cre+ mice produced significantly elevated levels of Dectin-1- and CR3-stimulated ROS, which was reduced by pharmacologic inhibition of Erk. SIRPα (signal regulatory protein α) is a myeloid inhibitory immunoreceptor that requires tyrosine phosphorylation to exert its inhibitory effect. YFP-Shp2C463A-expressing cells have elevated phospho-SIRPα levels and an increased Shp2-SIRPα interaction compared with YFP-WT Shp2-expressing cells. Collectively, these findings indicate that Shp2 phosphatase function positively regulates Dectin-1- and CR3-stimulated ROS production in macrophages by dephosphorylating and thus mitigating the inhibitory function of SIRPα and by promoting Erk activation.


Assuntos
Macrófagos/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Explosão Respiratória/fisiologia , Animais , Western Blotting , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Imunofluorescência , Imunoprecipitação , Integrases/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/citologia , Masculino , Camundongos , Camundongos Knockout , Fagocitose , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Tirosina/metabolismo
12.
Blood ; 120(13): 2669-78, 2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-22806893

RESUMO

Intracellular mechanism(s) that contribute to promiscuous signaling via oncogenic KIT in systemic mastocytosis and acute myelogenous leukemia are poorly understood. We show that SHP2 phosphatase is essential for oncogenic KIT-induced growth and survival in vitro and myeloproliferative disease (MPD) in vivo. Genetic disruption of SHP2 or treatment of oncogene-bearing cells with a novel SHP2 inhibitor alone or in combination with the PI3K inhibitor corrects MPD by disrupting a protein complex involving p85α, SHP2, and Gab2. Importantly, a single tyrosine at position 719 in oncogenic KIT is sufficient to develop MPD by recruiting p85α, SHP2, and Gab2 complex to oncogenic KIT. Our results demonstrate that SHP2 phosphatase is a druggable target that cooperates with lipid kinases in inducing MPD.


Assuntos
Transformação Celular Neoplásica/patologia , Proteína Adaptadora GRB2/fisiologia , Mutação/genética , Transtornos Mieloproliferativos/etiologia , Transtornos Mieloproliferativos/prevenção & controle , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Proteínas Proto-Oncogênicas c-kit/genética , Animais , Apoptose , Western Blotting , Transplante de Medula Óssea , Proliferação de Células , Transformação Celular Neoplásica/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunoprecipitação , Integrases/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Transtornos Mieloproliferativos/mortalidade , Fosforilação/efeitos dos fármacos , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais/efeitos dos fármacos , Taxa de Sobrevida , Tirosina/metabolismo
13.
Curr Opin Hematol ; 19(4): 273-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22504523

RESUMO

PURPOSE OF REVIEW: The protein tyrosine phosphatase Shp2 is encoded by PTPN11 and positively regulates physiologic hematopoiesis. Mutations of PTPN11 cause the congenital disorder Noonan syndrome and pathologically promote human leukemias. Given the high frequency of PTPN11 mutations in human disease, several animal models have been generated to investigate Shp2 in hematopoietic stem cell (HSC) function and leukemic transformation. RECENT FINDINGS: Two independent animal models bearing knockout of Shp2 in hematopoietic tissues clearly demonstrate the necessity of Shp2 in HSC repopulating capacity. Reduced HSC quiescence and increased apoptosis accounts for diminished HSC function in the absence of Shp2. The germline mutation Shp2D61G enhances HSC activity and induces myeloproliferative disease (MPD) in vivo by HSC transformation. The somatic mutation Shp2D61Y produces MPD in vivo but fails to induce acute leukemia, whereas somatic Shp2E76K produces MPD in vivo that transforms into full-blown leukemia. HSCs expressing Shp2D61Y do not generate MPD in recipient animals upon transplantation, whereas Shp2E76K-expressing HSCs yield MPD as well as acute leukemia in recipient animals. The mechanisms underlying the unique functions of Shp2D61Y and Shp2E76K in HSC transformation and leukemogenesis continue to be under investigation. SUMMARY: Further understanding of the physiologic and pathologic role of Shp2 in hematopoiesis and leukemogenesis, respectively, will yield information needed to develop therapeutic strategies targeted to Shp2 in human disease.


Assuntos
Transformação Celular Neoplásica , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Leucemia Mieloide Aguda/enzimologia , Transtornos Mieloproliferativos/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Animais , Regulação Neoplásica da Expressão Gênica/genética , Células-Tronco Hematopoéticas/citologia , Leucemia Mieloide Aguda/genética , Modelos Animais , Transtornos Mieloproliferativos/genética
14.
Cancer Cell ; 20(3): 357-69, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21907926

RESUMO

We show constitutive activation of Rho kinase (ROCK) in cells bearing oncogenic forms of KIT, FLT3, and BCR-ABL, which is dependent on PI3K and Rho GTPase. Genetic or pharmacologic inhibition of ROCK in oncogene-bearing cells impaired their growth as well as the growth of acute myeloid leukemia patient-derived blasts and prolonged the life span of mice bearing myeloproliferative disease. Downstream from ROCK, rapid dephosphorylation or loss of expression of myosin light chain resulted in enhanced apoptosis, reduced growth, and loss of actin polymerization in oncogene-bearing cells leading to significantly prolonged life span of leukemic mice. In summary we describe a pathway involving PI3K/Rho/ROCK/MLC that may contribute to myeloproliferative disease and/or acute myeloid leukemia in humans.


Assuntos
Transformação Celular Neoplásica , Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia/metabolismo , Transtornos Mieloproliferativos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fator de Células-Tronco/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Quinases Associadas a rho/metabolismo , Actinas/metabolismo , Animais , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Proteínas de Fusão bcr-abl/biossíntese , Proteínas de Fusão bcr-abl/genética , Humanos , Leucemia/mortalidade , Leucemia/patologia , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Transtornos Mieloproliferativos/mortalidade , Transtornos Mieloproliferativos/patologia , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo , Fosfatidilinositol 3-Quinases/biossíntese , Fosforilação , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/genética , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/genética , Fator de Células-Tronco/biossíntese , Fator de Células-Tronco/genética , Tirosina Quinase 3 Semelhante a fms/biossíntese , Tirosina Quinase 3 Semelhante a fms/genética , Proteínas rho de Ligação ao GTP/biossíntese , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética
15.
Leuk Res ; 35(8): 1108-10, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21420732

RESUMO

Prolonged administration of methyl transferase inhibitors may increase response rates in myelodysplastic syndromes (MDS). Fourteen MDS patients with anemia and less than 10% marrow blasts received azacitidine 50 mg/m(2) thrice weekly for 2 weeks every 4 weeks; 7 also received weekly erythropoietin. The response rate of 43% did not improve the rates reported with other azacitidine administration schedules, so the study was closed. A decreased apoptosis of primitive erythroid progenitors and increased expression of BclX(L) was observed with treatment in responding patients compared to non-responders. Azacitidine may modulate BclX(L) and improve erythropoiesis through reduction of apoptosis in primitive erythroid progenitor population in MDS.


Assuntos
Anemia/tratamento farmacológico , Antimetabólitos Antineoplásicos/uso terapêutico , Azacitidina/uso terapêutico , Células Precursoras Eritroides/efeitos dos fármacos , Eritropoese/efeitos dos fármacos , Síndromes Mielodisplásicas/complicações , Idoso , Idoso de 80 Anos ou mais , Anemia/etiologia , Apoptose/efeitos dos fármacos , Eritropoetina/uso terapêutico , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/terapia , Prognóstico , Proteína bcl-X/metabolismo
16.
Cancer Res ; 71(6): 2203-11, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21285250

RESUMO

The asymmetric organization of epithelial cells is a basic counter to cellular proliferation. However, the mechanisms whereby pro-growth pathways are modulated by intracellular factors that control cell shape are not well understood. This study demonstrates that the adaptor protein Amot, in addition to its established role in regulating cellular asymmetry, also promotes extracellular signal-regulated kinase 1 and 2 (ERK1/2)-dependent proliferation of mammary cells. Specifically, expression of Amot80, but not a mutant lacking its polarity protein interaction domain, enhances ERK1/2-dependent proliferation of MCF7 cells. Further, expression of Amot80 induces nontransformed MCF10A cells to overgrow as disorganized cellular aggregates in Matrigel. Conversely, Amot expression is required for proliferation of breast cancer cells in specific microenvironmental contexts that require ERK1/2 signaling. Thus, Amot is proposed to coordinate the dysregulation of cell polarity with the induction of neoplastic growth in mammary cells.


Assuntos
Proliferação de Células , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Angiomotinas , Linhagem Celular , Linhagem Celular Tumoral , Colágeno , Combinação de Medicamentos , Ativação Enzimática , Células Epiteliais/citologia , Feminino , Células HEK293 , Humanos , Immunoblotting , Peptídeos e Proteínas de Sinalização Intercelular/genética , Laminina , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Glândulas Mamárias Humanas/citologia , Proteínas de Membrana/genética , Proteínas dos Microfilamentos , Microscopia Confocal , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mutação , Proteoglicanas , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Microambiente Tumoral
17.
J Med Chem ; 53(6): 2482-93, 2010 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-20170098

RESUMO

The Src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2) plays a pivotal role in growth factor and cytokine signaling. Gain-of-function SHP2 mutations are associated with Noonan syndrome, various kinds of leukemias, and solid tumors. Thus, there is considerable interest in SHP2 as a potential target for anticancer and antileukemia therapy. We report a salicylic acid based combinatorial library approach aimed at binding both active site and unique nearby subpockets for enhanced affinity and selectivity. Screening of the library led to the identification of a SHP2 inhibitor II-B08 (compound 9) with highly efficacious cellular activity. Compound 9 blocks growth factor stimulated ERK1/2 activation and hematopoietic progenitor proliferation, providing supporting evidence that chemical inhibition of SHP2 may be therapeutically useful for anticancer and antileukemia treatment. X-ray crystallographic analysis of the structure of SHP2 in complex with 9 reveals molecular determinants that can be exploited for the acquisition of more potent and selective SHP2 inhibitors.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Indóis/química , Indóis/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Ácido Salicílico/química , Triazóis/química , Triazóis/farmacologia , Animais , Apoptose/efeitos dos fármacos , Área Sob a Curva , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Cristalografia por Raios X , Inibidores Enzimáticos/farmacocinética , Feminino , Humanos , Indóis/farmacocinética , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Modelos Químicos , Modelos Moleculares , Estrutura Molecular , Mutação , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/prevenção & controle , Ligação Proteica , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 11/química , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Bibliotecas de Moléculas Pequenas , Triazóis/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Mol Cell Biol ; 29(16): 4376-93, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19528235

RESUMO

Juvenile myelomonocytic leukemia (JMML) is characterized by myelomonocytic cell overproduction and commonly bears activating mutations in PTPN11. Murine hematopoietic progenitors expressing activating Shp2 undergo myelomonocytic differentiation, despite being subjected to conditions that normally support only mast cells. Evaluation of hematopoietic-specific transcription factor expression indicates reduced GATA2 and elevated c-Jun in mutant Shp2-expressing progenitors. We hypothesized that mutant Shp2-induced Ras hyperactivation promotes c-Jun phosphorylation and constitutive c-Jun expression, permitting, as a coactivator of PU.1, excessive monocytic differentiation and reduced GATA2. Hematopoietic progenitors expressing activating Shp2 demonstrate enhanced macrophage CFU (CFU-M) compared to that of wild-type Shp2-expressing cells. Treatment with the JNK inhibitor SP600125 or cotransduction with GATA2 normalizes activating Shp2-generated CFU-M. However, cotransduction of DeltaGATA2 (lacking the C-terminal zinc finger, needed to bind PU.1) fails to normalize CFU-M. NIH 3T3 cells expressing Shp2E76K produce higher levels of luciferase expression directed by the macrophage colony-stimulating factor receptor (MCSFR) promoter, which utilizes c-Jun as a coactivator of PU.1. Coimmunoprecipitation demonstrates increased c-Jun-PU.1 complexes in mutant Shp2-expressing hematopoietic progenitors, while chromatin immunoprecipitation demonstrates increased c-Jun binding to the c-Jun promoter and an increased c-Jun-PU.1 complex at the Mcsfr promoter. Furthermore, JMML progenitors express higher levels of c-JUN than healthy controls, substantiating the disease relevance of these mechanistic findings.


Assuntos
Diferenciação Celular/fisiologia , Fator de Transcrição GATA2/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Monócitos/fisiologia , Mutação , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Animais , Antracenos/metabolismo , Criança , Ativação Enzimática , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Lactente , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Leucemia Mielomonocítica Juvenil/genética , Mastócitos/citologia , Mastócitos/fisiologia , Camundongos , Monócitos/citologia , Células NIH 3T3 , Regiões Promotoras Genéticas , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
19.
J Biol Chem ; 283(4): 2120-8, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18025090

RESUMO

The use of RNA interference has proven to be an effective means to study the function of genes. Constitutive synthesis of small interfering RNA molecules can be accomplished with the use of viral vectors expressing short hairpin RNA (shRNA). Binding of shRNA to the target mRNA promotes transcript degradation. So far, little is known about the effects that shRNA induce in vivo. To determine the feasibility of using helper-dependent adenoviral vectors for expression of shRNA in liver, we have designed an shRNA construct to mouse fabp5 (fatty acid-binding protein 5). Intravenous administration of this vector resulted in approximately 75% silencing of fabp5. Increasing the dose of vector did not result in higher levels of silencing, indicating that there is a threshold for the level of knockdown that can be achieved. Synthesis of high levels of shRNA molecules did not alter the levels of cellular micro-RNA, such as miR-122 and let-7a, suggesting that the exportin-5 pathway was not affected. However, high level shRNA expression resulted in activation of the interferon response. Thus, an important consideration when using shRNA-based vectors in vivo is to closely monitor signs of interferon-stimulated gene expression, since a narrow window exists between gene silencing efficacy and nonspecific effects.


Assuntos
Dependovirus , Proteínas de Ligação a Ácido Graxo/biossíntese , Interferons/biossíntese , Fígado/metabolismo , Proteínas de Neoplasias/biossíntese , Estabilidade de RNA , RNA Antissenso/biossíntese , Animais , Proteínas de Ligação a Ácido Graxo/antagonistas & inibidores , Proteínas de Ligação a Ácido Graxo/genética , Regulação da Expressão Gênica/genética , Inativação Gênica/fisiologia , Humanos , Interferons/genética , Carioferinas/biossíntese , Carioferinas/genética , Fígado/citologia , Camundongos , MicroRNAs/biossíntese , MicroRNAs/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , RNA Antissenso/genética , Transdução Genética
20.
Diagn Microbiol Infect Dis ; 52(4): 285-93, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15893902

RESUMO

Candida glabrata is emerging as a more common and important human pathogen. It is less susceptible to azole antifungals than Candida albicans, thus, posing some unique treatment challenges. Previously undetected C. glabrata isolates were identified from clinical specimens by adding bile to the growth medium. Cholesterol was found to be the responsible ingredient in bile. Six bile-dependent isolates were characterized and were found to exhibit wild-type equivalent growth when provided human or bovine serum or free cholesterol. Sterol profiles of the 6 isolates and a C. glabrata matching wild-type strain not requiring cholesterol indicated that 2 were defective in squalene epoxidase (encoded by the ERG1 gene) activity, 3 were defective in lanosterol synthase (encoded by the ERG7 gene) activity, and the sixth was defective in heme biosynthesis. All 7 isolates produced profiles that contained cholesterol transported from the media. Because Saccharomyces cerevisiae mutants unable to synthesize heme will take up exogenous sterol under aerobic conditions, hem1 nulls of C. glabrata and C. albicans were generated and tested for growth on ergosterol media. Only the C. glabrata hem1 was able to grow indicating significant differences in exogenous sterol uptake between the 2 organisms. The ability of C. glabrata to replace ergosterol with host sterol may be responsible for its elevated azole resistance.


Assuntos
Azóis/farmacologia , Candida glabrata/efeitos dos fármacos , Candida glabrata/metabolismo , Esteróis/metabolismo , Animais , Candida albicans/crescimento & desenvolvimento , Candida albicans/metabolismo , Candida glabrata/genética , Candida glabrata/crescimento & desenvolvimento , Bovinos , Colesterol/metabolismo , Meios de Cultura , Ergosterol/metabolismo , Proteínas Fúngicas/genética , Heme/biossíntese , Humanos , Mutação , Soro/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...