Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Ther Oncol ; 32(1): 200771, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38596309

RESUMO

The high rates of protein synthesis and processing render multiple myeloma (MM) cells vulnerable to perturbations in protein homeostasis. The induction of proteotoxic stress by targeting protein degradation with proteasome inhibitors (PIs) has revolutionized the treatment of MM. However, resistance to PIs is inevitable and represents an ongoing clinical challenge. Our first-in-human study of the selective inhibitor of RNA polymerase I transcription of ribosomal RNA genes, CX-5461, has demonstrated a potential signal for anti-tumor activity in three of six heavily pre-treated MM patients. Here, we show that CX-5461 has potent anti-myeloma activity in PI-resistant MM preclinical models in vitro and in vivo. In addition to inhibiting ribosome biogenesis, CX-5461 causes topoisomerase II trapping and replication-dependent DNA damage, leading to G2/M cell-cycle arrest and apoptotic cell death. Combining CX-5461 with PI does not further enhance the anti-myeloma activity of CX-5461 in vivo. In contrast, CX-5461 shows synergistic interaction with the histone deacetylase inhibitor panobinostat in both the Vk∗MYC and the 5T33-KaLwRij mouse models of MM by targeting ribosome biogenesis and protein synthesis through distinct mechanisms. Our findings thus provide strong evidence to facilitate the clinical development of targeting the ribosome to treat relapsed and refractory MM.

2.
Leukemia ; 36(12): 2802-2816, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36229595

RESUMO

ETP-ALL (Early T cell Progenitor Acute Lymphoblastic Leukemia) represents a high-risk subtype of T cell acute lymphocytic leukemia (T-ALL). Therapeutically, ETP-ALL patients frequently relapse after conventional chemotherapy highlighting the need for alternative therapeutic approaches. Using our ZEB2Tg ETP-ALL mouse model we previously documented the potential utility of the catalytic LSD1 inhibitor (GSK2879552) for treating mouse/human ETP-ALL. However, this approach proved to be inefficient, especially in killing human LOUCY cell ETP-ALL xenografts in vivo. Here we have revealed the novel involvement of ZEB2/LSD1 complexes in repressing the intrinsic apoptosis pathway by inhibiting the expression of several pro-apoptotic proteins such as BIM (BCL2L11) as a major driver for ETP-ALL survival. Treatment with LSD1i (particularly with the steric inhibitor SP2509) restored the expression of ZEB2/LSD1 pro-apoptotic BIM (BCL2L11) target. In combination with a JAK/STAT pathway inhibitor (JAKi, Ruxolitinib) or with a direct inhibitor of the anti-apoptotic BCL2 protein (BCL2i, ABT-199) resistance of human and mouse ETP-ALL to LSD1i was reversed. This new combination approach efficiently inhibited the growth of human and mouse ETP-ALL cells in vivo by enhancing their differentiation and triggering an apoptotic response. These results set the stage for novel combination therapies to be used in clinical trials to treat ETP-ALL patients.


Assuntos
Inibidores de Janus Quinases , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Humanos , Camundongos , Animais , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Proteína 11 Semelhante a Bcl-2/metabolismo , Inibidores de Janus Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Histona Desmetilases/metabolismo
3.
Autophagy ; 18(6): 1274-1296, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34530675

RESUMO

Cancer cell growth is dependent upon the sustainability of proliferative signaling and resisting cell death. Macroautophagy/autophagy promotes cancer cell growth by providing nutrients to cells and preventing cell death. This is in contrast to autophagy promoting cell death under some conditions. The mechanism regulating autophagy-mediated cancer cell growth remains unclear. Herein, we demonstrate that TSSC4 (tumor suppressing subtransferable candidate 4) is a novel tumor suppressor that suppresses cancer cell growth and tumor growth and prevents cell death induction during excessive growth by inhibiting autophagy. The oncogenic proteins ERBB2 (erb-b2 receptor tyrosine kinase 2) and the activation EGFR mutant (EGFRvIII, epidermal growth factor receptor variant III) promote cell growth and TSSC4 expression in breast cancer and glioblastoma multiforme (GBM) cells, respectively. In EGFRvIII-expressing GBM cells, TSSC4 knockout shifted the function of autophagy from a pro-cell survival role to a pro-cell death role during prolonged cell growth. Furthermore, the interaction of TSSC4 with MAP1LC3/LC3 (microtubule associated protein 1 light chain 3) via its conserved LC3-interacting region (LIR) contributes to its inhibition of autophagy. Finally, TSSC4 suppresses tumorsphere formation and tumor growth by inhibiting autophagy and maintaining cell survival in tumorspheres. Taken together, sustainable cancer cell growth can be achieved by autophagy inhibition via TSSC4 expression.Abbreviations: 3-MA: 3-methyladenine; ACTB: actin beta; CQ: chloroquine; EGFRvIII: epidermal growth factor receptor variant III; ERBB2: erb-b2 receptor tyrosine kinase 2; GBM: glioblastoma multiforme; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule Associated protein 1 light chain 3; TSSC4: tumor suppressing subtransferable candidate 4.


Assuntos
Autofagia , Glioblastoma , Transformação Celular Neoplásica , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Receptor ErbB-2 , Proteínas Supressoras de Tumor
4.
J Immigr Minor Health ; 20(6): 1429-1437, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29290019

RESUMO

Nationally, racial and ethnic disparities in childhood asthma plateaued from 2005 to 2013. We assessed trends in childhood asthma in Miami, Florida using Youth Risk Behavior Surveillance System (YRBSS) data and emergency department (ED) utilization and hospitalization rates by zip code population characteristics. Asthma prevalence in Miami did not vary significantly by race/ethnicity in YRBSS respondents in 2005 (16.2-17.2%, all groups), but rose in African-Americans and Hispanics and declined in Whites by 2013 to 27.9, 20.9 and 12.6%, respectively (P = 0.02). Median asthma ED visit rates rose from 106.8 (2006-2008) to 138.2 (2011-2013; P = 0.004) per 10,000 children. High-poverty and majority African-American zip codes were 6.3 and 7.3 times more likely to have asthma ED visit rates > 200 than others (P < 0.001). In high-poverty zip codes, majority African-American population was not associated with significantly higher ED utilization. In low-poverty zip codes, the association became stronger. Greater poverty explains much, but not all of Miami African-Americans' higher asthma risk.


Assuntos
Asma/etnologia , Serviço Hospitalar de Emergência/estatística & dados numéricos , Etnicidade/estatística & dados numéricos , Pobreza/estatística & dados numéricos , Grupos Raciais/estatística & dados numéricos , Adolescente , Negro ou Afro-Americano/estatística & dados numéricos , Criança , Pré-Escolar , Feminino , Florida/epidemiologia , Inquéritos Epidemiológicos , Hispânico ou Latino/estatística & dados numéricos , Humanos , Masculino , Prevalência , Características de Residência/estatística & dados numéricos , Fatores de Risco , Fatores Socioeconômicos , População Branca/estatística & dados numéricos
5.
Int J Mol Sci ; 18(7)2017 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-28654000

RESUMO

Non-melanomatous skin cancers (NMSCs), which include basal and squamous cell carcinoma (BCC and SCC respectively), represent a significant burden on the population, as well as an economic load to the health care system; yet treatments of these preventable cancers remain ineffective. Studies estimate that there has been a 2-fold increase in the incidence of NMSCs between the 1960s and 1980s. The increase in cases of NMSCs, as well as the lack of effective treatments, makes the need for novel therapeutic approaches all the more necessary. To rationally develop more targeted treatments for NMSCs, a better understanding of the cell of origin, in addition to the underlying pathophysiological mechanisms that govern the development of these cancers, is urgently required. Research over the past few years has provided data supporting both a "bottom up" and "top down" mechanism of tumourigenesis. The "bottom up" concept involves a cancer stem cell originating in the basal compartment of the skin, which ordinarily houses the progenitor cells that contribute towards wound healing and normal cell turnover of overlying epidermal skin layers. The "top down" concept involves a more differentiated cell undergoing genetic modifications leading to dedifferentiation, giving rise to cancer initiating cells (CICs). This review explores both concepts, to paint a picture of the skin SCC cell of origin, the underlying biology, and also how this knowledge might be exploited to develop novel therapies.


Assuntos
Carcinoma Basocelular/patologia , Carcinoma de Células Escamosas/patologia , Epiderme/patologia , Células-Tronco Neoplásicas/patologia , Neoplasias Cutâneas/patologia , Pele/patologia , Animais , Carcinogênese/genética , Carcinogênese/patologia , Carcinoma Basocelular/genética , Carcinoma de Células Escamosas/genética , Desdiferenciação Celular , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Epiderme/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Células-Tronco Neoplásicas/metabolismo , Pele/metabolismo , Neoplasias Cutâneas/genética , Fatores de Transcrição/análise , Fatores de Transcrição/genética
6.
Mol Cancer Ther ; 16(9): 1765-1778, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28500235

RESUMO

Currently available treatment options are unlikely to be curative for the majority of multiple myeloma patients, emphasizing a continuing role for the introduction of investigational agents that can overcome drug resistance. The canonical Wnt/ß-catenin signaling pathway, essential for self-renewal, growth, and survival, has been found to be dysregulated in multiple myeloma, particularly in advanced stages of disease. This provides the rationale for evaluating the novel ß-catenin inhibitor BC2059 as monotherapy and in combination with proteasome inhibitors in vitro and in vivo Here, we show nuclear localization of ß-catenin in human myeloma cell lines (HMCL), consistent with activation of the canonical Wnt pathway. BC2059 attenuates ß-catenin levels, in both the cytoplasm and the nucleus, reducing the transcriptional activity of the TCF4/LEF complex and the expression of its target gene axin 2. Treatment of HMCL with BC2059 inhibits proliferation and induces apoptosis in a dose-dependent manner. This is also observed in HMCL-stromal cell cocultures, mitigating the protective effect afforded by the stroma. Similarly, BC2059 induces apoptosis in primary multiple myeloma samples in vitro, causing minimal apoptosis on healthy peripheral blood mononuclear cells. Furthermore, it synergizes with the proteasome inhibitor bortezomib both in HMCL and primary multiple myeloma samples. Finally, in xenograft models of human myelomatosis, BC2059 delays tumor growth and prolongs survival with minor on-target side effects. Collectively, these results demonstrate the efficacy of targeting the Wnt/ß-catenin pathway with BC2059 both in vitro and in vivo, at clinically achievable doses. These findings support further clinical evaluation of BC2059 for the treatment of multiple myeloma. Mol Cancer Ther; 16(9); 1765-78. ©2017 AACR.


Assuntos
Antineoplásicos/farmacologia , Bortezomib/farmacologia , Mieloma Múltiplo/metabolismo , Inibidores de Proteassoma/farmacologia , beta Catenina/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Transgênicos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Ativação Transcricional/efeitos dos fármacos , Carga Tumoral , beta Catenina/antagonistas & inibidores , beta Catenina/genética
7.
Mater Sci Eng C Mater Biol Appl ; 71: 584-593, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27987748

RESUMO

Engineered nanoparticles with multiple complementary imaging modalities are of great benefit to the rapid treatment and diagnosis of disease in various organs. Herein, we report the formulation of cubosomes and hexosomes that carry multiple amphiphilic imaging contrast agents in their self-assembled lipid bilayers. This is the first report of the use of both near infrared fluorescent (NIRF) imaging and gadolinium lipid based magnetic resonance (MR) imaging modalities in cubosomes and hexosomes. High-throughput screening was used to rapidly optimize formulations with desirable nano-architectures and low in vitro cytotoxicity. The dual-modal imaging nanoparticles in vivo biodistribution and organ specific contrast enhancement were then studied. The NIRF in vivo imaging results indicated accumulation of both cubosomes and hexosomes in the liver and spleen of mice up to 20h post-injection. Remarkably, the biodistribution of the nanoparticle formulations was affected by the mesophase (i.e. cubic or hexagonal), a finding of significant importance for the future use of these compounds, with hexosomes showing higher accumulation in the spleen than the liver compared to cubosomes. Furthermore, in vivo MRI data of animals injected with either type of lyotropic liquid crystal nanoparticle displayed enhanced contrast in the liver and spleen.


Assuntos
Meios de Contraste , Imageamento por Ressonância Magnética , Nanopartículas/química , Imagem Óptica , Animais , Células CHO , Meios de Contraste/química , Meios de Contraste/farmacocinética , Meios de Contraste/farmacologia , Cricetulus , Humanos , Masculino , Camundongos , Células U937
8.
Br J Haematol ; 169(3): 333-43, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25653020

RESUMO

MDX-1097 is an antibody specific for a unique B cell antigen called kappa myeloma antigen (KMA) that consists of cell membrane-associated free kappa light chain (κFLC). KMA was detected on kappa human multiple myeloma cell lines (κHMCLs), on plasma cells (PCs) from kappa multiple myeloma (κMM) patients and on κPC dyscrasia tissue cryosections. In primary κMM samples, KMA was present on CD38+ cells that were CD138 and CD45 positive and/or negative. MDX-1097 exhibited a higher affinity for KMA compared to κFLC and the latter did not abrogate binding to KMA. MDX-1097-mediated antibody-dependent cellular cytotoxicity (ADCC) and in vitro exposure of target cells to the immunomodulatory drug lenalidomide resulted in increased KMA expression and ADCC. Also, in vitro exposure of peripheral blood mononuclear cells (PBMCs) to lenalidomide enhanced MDX-1097-mediated ADCC. PBMCs obtained from myeloma patients after lenalidomide therapy elicited significantly higher levels of MDX-1097-mediated ADCC than cells obtained prior to lenalidomide treatment. These data establish KMA as a relevant cell surface antigen on MM cells that can be targeted by MDX-1097. The ADCC-inducing capacity of MDX-1097 and its potentiation by lenalidomide provide a powerful rationale for clinical evaluation of MDX-1097 alone and in combination with lenalidomide.


Assuntos
Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antineoplásicos/farmacologia , Cadeias kappa de Imunoglobulina/metabolismo , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/metabolismo , Talidomida/análogos & derivados , Anticorpos Monoclonais/metabolismo , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Antineoplásicos/metabolismo , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Imunofenotipagem , Lenalidomida , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Mieloma Múltiplo/diagnóstico , Fenótipo , Plasmócitos/metabolismo , Ligação Proteica , Talidomida/farmacologia
9.
J Asthma ; 50(5): 480-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23544421

RESUMO

OBJECTIVE: Asthma is the most common chronic condition in childhood and disproportionately impacts the poorer and ethnic minorities. The objectives of the study were to estimate the prevalence of asthma in Miami-Dade County (MDC) schoolchildren to aid case-finding and linkage to care. METHODS: We used the CDC Youth Risk Behavior Surveillance System (YRBSS) definition of possible asthma ("Ever told by a doctor or nurse that they had asthma and still had asthma") and analyzed data from four sources. These included the: (1)-MDC YRBSS 2009, (2)-MDC Health Connect Asthma Survey of school nurses (>2008), (3)-parents' survey in the five HealthConnect survey elementary schools with highest asthma prevalence, and (4)-focus group with parents of asthmatic children. RESULTS: (1)-MDC YRBSS data showed that 21.3% of high- and 21.4% of middle-school students had been diagnosed with possible asthma. Prevalence was the highest in African-American middle-school girls (26.9%). (2)-HealthConnect survey responders reported that 4.9% of the students in 131 MDC schools had possible asthma. Asthma prevalence was higher in elementary schools (median = 7.1%) and in low-income MDC zip codes. (3)-Of the parent survey responders, 24.9% indicated that their child had possible asthma, and 19.2% reported that their children had no usual source of care. (4)-Focus group participants reported frequent loss of Medicaid coverage for their children, landlords' indifference to the role of poorly maintained housing in asthma, and unmet needs regarding knowledge of health system navigation. CONCLUSIONS: Asthma may be common in MDC schoolchildren, particularly in poor communities. Formidable structural factors limit the caregivers' abilities to manage childhood asthma.


Assuntos
Asma/epidemiologia , Avaliação das Necessidades , Adolescente , Criança , Feminino , Florida/epidemiologia , Grupos Focais , Humanos , Masculino , Pais , Prevalência , Estudantes
10.
Biol Blood Marrow Transplant ; 18(5): 683-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22281302

RESUMO

Age-related differences in thymic function influence the rapidity of T cell reconstitution following hematopoietic stem cell transplantation (HSCT). In adults, thymic reconstitution is delayed until after marrow engraftment is established, and is significantly improved by approaches that increase marrow chimerism, such as pretransplantation irradiation. In contrast, we show that neonatal mice undergo more rapid and efficient thymic reconstitution than adults, even when bone marrow (BM) engraftment is minimal and in the absence of pretransplantation radiation. We have previously shown that the neonatal thymus produces high levels of vascular endothelial growth factor (VEGF) that drives angiogenesis locally. In this report, we show that inhibition of VEGF prior to HSCT prevents rapid thymic reconstitution in neonates, but has no effect on thymic reconstitution in adults. These data suggest that the early radiation-independent thymic reconstitution unique to the neonatal host is mediated through VEGF, and reveals a novel pathway that might be targeted to improve immune reconstitution post-HSCT.


Assuntos
Transplante de Medula Óssea , Linfócitos T/imunologia , Timócitos/imunologia , Timo/imunologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fatores Etários , Animais , Animais Recém-Nascidos , Sobrevivência de Enxerto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Microscopia Confocal , Neovascularização Fisiológica , Receptores de Fatores de Crescimento do Endotélio Vascular/administração & dosagem , Receptores de Fatores de Crescimento do Endotélio Vascular/efeitos adversos , Receptores de Fatores de Crescimento do Endotélio Vascular/imunologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/efeitos da radiação , Timócitos/efeitos dos fármacos , Timócitos/efeitos da radiação , Timo/irrigação sanguínea , Timo/efeitos dos fármacos , Timo/efeitos da radiação , Quimeras de Transplante , Fator A de Crescimento do Endotélio Vascular/imunologia , Irradiação Corporal Total
11.
Exp Hematol ; 40(1): 3-13.e3, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22001673

RESUMO

Purine analogs such as 6-thioguanine (6TG) cause myelotoxicity upon conversion into nucleotides by hypoxanthine-guanine phosphoribosyltransferase (HPRT). Here we have developed a novel and highly efficient strategy employing 6TG as a single agent for both conditioning and in vivo chemoselection of HPRT-deficient hematopoietic stem cells. The dose-response and time course of 6TG myelotoxicity were first compared in HPRT wild-type mice and HPRT-deficient transgenic mice. Dosage and schedule parameters were optimized to employ 6TG for myelosuppressive conditioning, immediately followed by in vivo chemoselection of HPRT-deficient transgenic donor bone marrow (BM) transplanted into syngeneic HPRT wild-type recipients. At appropriate doses, 6TG induced selective myelotoxicity without any adverse effects on extrahematopoietic tissues in HPRT wild-type mice, while hematopoietic stem cells deficient in HPRT activity were highly resistant to its cytotoxic effects. Combined 6TG conditioning and post-transplantation chemoselection consistently achieved ∼95% engraftment of HPRT-deficient donor BM, with low overall toxicity. Long-term reconstitution of immunophenotypically normal BM was achieved in both primary and secondary recipients. Our results provide proof-of-concept that single-agent 6TG can be used for both myelosuppressive conditioning without requiring irradiation and for in vivo chemoselection of HPRT-deficient donor cells. Our results show that by applying the myelosuppressive effects of 6TG both before (as conditioning) and after transplantation (as chemoselection), highly efficient engraftment of HPRT-deficient hematopoietic stem cells can be achieved.


Assuntos
Transplante de Medula Óssea , Medula Óssea/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Hipoxantina Fosforribosiltransferase/deficiência , Tioguanina/farmacologia , Condicionamento Pré-Transplante , Animais , Medula Óssea/enzimologia , Relação Dose-Resposta a Droga , Células-Tronco Hematopoéticas/metabolismo , Hipoxantina Fosforribosiltransferase/metabolismo , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Tioguanina/administração & dosagem , Tioguanina/efeitos adversos , Fatores de Tempo
12.
Blood ; 113(12): 2723-31, 2009 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-19088378

RESUMO

Although the mechanisms of cross-talk that regulate the hematopoietic and epithelial compartments of the thymus are well established, the interactions of these compartments with the thymic endothelium have been largely ignored. Current understanding of the thymic vasculature is based on studies of adult thymus. We show that the neonatal period represents a unique phase of thymic growth and differentiation, marked by endothelium that is organized as primitive, dense networks of capillaries dependent on vascular endothelial growth factor (VEGF). VEGF dependence in neonates is mediated by significantly higher levels of both VEGF production and endothelial VEGF receptor 2 (VEGF-R2) expression than in the adult thymus. VEGF is expressed locally in the neonatal thymus by immature, CD4(-)CD8(-) "double negative" (DN) thymocytes and thymic epithelium. Relative to adult thymus, the neonatal thymus has greater thymocyte proliferation, and a predominance of immature thymocytes and cortical thymic epithelial cells (cTECs). Inhibition of VEGF signaling during the neonatal period results in rapid loss of the dense capillaries in the thymus and a marked reduction in the number of thymocytes. These data demonstrate that, during the early postnatal period, VEGF mediates cross-talk between the thymocyte and endothelial compartments of the thymus.


Assuntos
Animais Recém-Nascidos/fisiologia , Endotélio Vascular/metabolismo , Células Epiteliais/metabolismo , Timo/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Capilares/crescimento & desenvolvimento , Contagem de Células , Endotélio Vascular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Linfócitos Nulos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/fisiologia , Pericitos/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Organismos Livres de Patógenos Específicos , Timo/irrigação sanguínea , Timo/citologia , Timo/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
13.
Mol Cancer Ther ; 7(4): 980-92, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18413811

RESUMO

New molecular cancer treatment strategies aim to reconstitute wild-type p53 (WTp53) function in mutant p53 (MTp53)-expressing tumors as a means of resensitizing cells to chemotherapy or radiotherapy. The success of this approach may depend on whether MTp53 proteins are acting in a dominant-negative or independent gain-of-function mode. Herein, we describe an isogenic, temperature-sensitive p53 model (p53(A138V)) in p53-null human H1299 lung cancer cells in which WTp53 can be selectively coexpressed with a temperature-sensitive MTp53 allele (A138V) during initial DNA damage and subsequent DNA repair. Cells expressing MTp53 alone or coexpressing induced WTp53 and MTp53 were tested for p53 transcription, G(1) and G(2) cell cycle checkpoints, apoptosis, and long-term clonogenic survival following DNA damage. Transient transfection of WTp53 into H1299 cells, or shift-down of H1299-p53(A138V) stable transfectants to 32 degrees C to induce WTp53, led to increased p21(WAF1) expression and G(1) and G(2) arrests following DNA damage but did not increase BAX expression or apoptosis. In contrast, both transient and stable expression of the p53(A138V) mutant in p53-null H1299 cells (e.g. testing gain-of-function) at 37 degrees C blocked p21(WAF1) induction following DNA damage. Cell death was secondary to mitotic catastrophe and/or tumor cell senescence. Overexpression of WTp53 did not resensitize resistant MTp53-expressing cells to ionizing radiation, cisplatinum, or mitomycin C. Our results suggest that human MTp53 proteins can lead to resistant phenotypes independent of WTp53-mediated transcription and checkpoint control. This should be considered when using p53 as a prognostic factor and therapeutic target.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Tolerância a Radiação/genética , Proteína Supressora de Tumor p53/genética , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Senescência Celular , Cisplatino/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Citometria de Fluxo , Imunofluorescência , Fase G1/efeitos dos fármacos , Fase G1/efeitos da radiação , Fase G2/efeitos dos fármacos , Fase G2/efeitos da radiação , Raios gama , Humanos , Immunoblotting , Imunoprecipitação , Mitomicina/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Radiação Ionizante , Radiossensibilizantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/efeitos da radiação , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos da radiação , Ensaio Tumoral de Célula-Tronco , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/metabolismo , beta-Galactosidase/metabolismo
14.
Semin Radiat Oncol ; 16(1): 51-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16378907

RESUMO

In response to DNA breaks, human cells delay their progression through the G1, S, and G2 phases of the cell cycle. This response requires the coordinated effort of the ATM-CHK2-p53 and ATR-CHK1 DNA damage-sensing pathways and DNA repair (eg, DNA-PK and RAD51 complexes). The turnover of many of these DNA damage-associated proteins is controlled by the 26S proteasome. In this article, we review molecular strategies that target each of these pathways using silencing RNA (siRNA), antisense, or small-molecule inhibition. Although these agents can radiosensitize tumor cells, little data are available regarding potential effects on normal tissues to determine the potential therapeutic ratio of these strategies after fractionated radiotherapy. Clinical trials using such agents will require novel correlative science endpoints to track DNA repair and cell-cycle arrest and will need careful assessment of normal tissue toxicity and stability.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Genes cdc/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Radiossensibilizantes/metabolismo , Radioterapia , Proteínas Supressoras de Tumor/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular/genética , Ciclo Celular/fisiologia , Ciclo Celular/efeitos da radiação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/efeitos da radiação , Reparo do DNA/genética , Reparo do DNA/efeitos da radiação , Proteínas de Ligação a DNA/genética , Genes cdc/efeitos da radiação , Humanos , Biologia Molecular/métodos , Proteínas Serina-Treonina Quinases/genética , Proteínas Supressoras de Tumor/genética
15.
Cancer Res ; 65(23): 10810-21, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16322227

RESUMO

Despite a clear link between ataxia-telangiectasia mutated (ATM)-dependent phosphorylation of p53 and cell cycle checkpoint control, the intracellular biology and subcellular localization of p53 phosphoforms during the initial sensing of DNA damage is poorly understood. Using G0-G1 confluent primary human diploid fibroblast cultures, we show that endogenous p53, phosphorylated at Ser15 (p53Ser15), accumulates as discrete, dose-dependent and chromatin-bound foci within 30 minutes following induction of DNA breaks or DNA base damage. This biologically distinct subpool of p53Ser15 is ATM dependent and resistant to 26S-proteasomal degradation. p53Ser15 colocalizes and coimmunoprecipitates with gamma-H2AX with kinetics similar to that of biochemical DNA double-strand break (DNA-dsb) rejoining. Subnuclear microbeam irradiation studies confirm p53Ser15 is recruited to sites of DNA damage containing gamma-H2AX, ATM(Ser1981), and DNA-PKcs(Thr2609) in vivo. Furthermore, studies using isogenic human and murine cells, which express Ser15 or Ser18 phosphomutant proteins, respectively, show defective nuclear foci formation, decreased induction of p21WAF, decreased gamma-H2AX association, and altered DNA-dsb kinetics following DNA damage. Our results suggest a unique biology for this p53 phosphoform in the initial steps of DNA damage signaling and implicates ATM-p53 chromatin-based interactions as mediators of cell cycle checkpoint control and DNA repair to prevent carcinogenesis.


Assuntos
Dano ao DNA , DNA/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Hidrolases Anidrido Ácido , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Cromatina/metabolismo , Reparo do DNA , Enzimas Reparadoras do DNA/metabolismo , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Células HCT116 , Histonas/metabolismo , Humanos , Imunoprecipitação , Camundongos , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo
16.
Radiother Oncol ; 76(2): 168-76, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16026872

RESUMO

BACKGROUND AND PURPOSE: Intratumoral hypoxia has been correlated with poor clinical outcome in prostate cancer. Prostate cancer cells can be genetically unstable and have altered DNA repair. We, therefore, hypothesized that the expression of DNA double-strand break (DNA-dsb) repair genes in normal and malignant prostate cultures can be altered under hypoxic conditions. METHODS AND MATERIALS: The expression of homologous recombination (HR) and non-homologous recombination (NHEJ) genes following gas hypoxia (0.2%) or exposure to HIF1alpha-inducing agent, CoCl2 (100 microM), was determined for normal diploid fibroblasts (GM05757) and the pre-malignant and malignant prostate cell lines, BPH-1, 22RV-1, DU145 and PC3. RNA and protein levels were determined using RT-PCR and Western blotting. Additionally, p53 genotype and function, the level of hypoxia-induced apoptosis, and cell cycle distribution, were determined to correlate to changes in DNA-dsb gene expression. RESULTS: Induction of hypoxia was confirmed using HIF1alpha and VEGF expression in gas- and CoCl2-treated cultures. Hypoxia (48-72 h of 0.2% O2) decreased RNA expression of a number of HR-related genes (e.g. Rad51, Rad52, Rad54, BRCA1, BRCA2) in both normal and malignant cultures. Similar decreases in RNA pertaining to the NHEJ-related genes (e.g. Ku70, DNA-PKcs, DNA Ligase IV, Xrcc4) were observed. In selected cases, hypoxia-mediated decreases in RNA expression led to decreased DNA-dsb protein expression. CoCl2-treated cultures did not show decreased DNA-dsb protein expression. The ability of hypoxia to down-regulate Rad51 and other HR-associated genes under hypoxia was not correlated to c-Abl or c-Myc gene expression, p53 genotype or function, propensity for hypoxia-mediated apoptosis, or specific changes in cell cycle distribution. CONCLUSIONS: Hypoxia can down-regulate expression of DNA-dsb repair genes in both normal and cancer cells. If associated with a functional decrease in DNA-dsb repair, this observation could provide a potential basis for the observed genetic instability within tumor cells exposed to hypoxia.


Assuntos
Hipóxia Celular/fisiologia , Reparo do DNA/genética , Regulação para Baixo , Expressão Gênica , Neoplasias da Próstata/genética , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Genes p53 , Humanos , Masculino , Neoplasias da Próstata/patologia , Análise de Sequência de DNA , Células Tumorais Cultivadas
17.
Cancer Metastasis Rev ; 23(3-4): 237-57, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15197326

RESUMO

The p53 tumor suppressor protein is a key mediator of an ATM-dependent DNA damage response cascade following cellular exposure to ionizing radiation. The p53-family members, p63 and p73, are highly similar to p53, yet are differentially activated by IR, UV and cis-platinum via ATM and c-abl/ATR signaling pathways. Loss of function of p53 can occur by mutation or degradation; giving rise to alterations in G(1) and G(2) cell cycle checkpoint control, cell death, DNA repair and genetic stability. The end result of these alterations can be the generation of radioresistant mutant tumor cells. Indeed, in isogenic systems, loss of p53 or p73 function has been associated with decreased chemosensitivity and radiosensitivity, in vitro. However, clinical data supporting a role for p53 genotype as an independent predictive factor for radiotherapy outcome continues to be controversial due to variable endpoints in clinical trial design and in methodology in detecting p53 function. Nonetheless, in carefully controlled radiotherapy studies where mutations in p53 have been detected using DNA sequencing or functional assays, the presence of mutant p53 can be associated with decreased local control following radiotherapy. This suggests that novel molecular treatment strategies specifically designed to re-institute normal p53 function within resistant tumors can be used as combined modality protocols to improve local control and maintain a therapeutic ratio. A future challenge lies in the pre-therapy determination of a 'molecular therapeutic ratio' for individual patients which could allow for specific prognostication based on p53 functional status and subsequent individualized therapy.


Assuntos
Tolerância a Radiação/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Dano ao DNA , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/radioterapia , Transdução de Sinais , Ativação Transcricional/genética , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/classificação , Proteína Supressora de Tumor p53/genética
18.
Mol Cell Biol ; 24(2): 662-74, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14701739

RESUMO

The detection of DNA damage activates DNA repair pathways and checkpoints to allow time for repair. Ultimately, these responses must be coordinated to ensure that cell cycle progression is halted until repair is completed. Several multiprotein complexes containing members of the structural maintenance of chromosomes family of proteins have been described, including the condensin and cohesin complexes, that are critical for chromosomal organization. Here we show that the Smc5/Smc6 (Smc5/6) complex is required for a coordinated response to DNA damage and normal chromosome integrity. Fission yeast cells lacking functional Smc6 initiate a normal checkpoint response to DNA damage, culminating in the phosphorylation and activation of the Chk1 protein kinase. Despite this, cells enter a lethal mitosis, presumably without completion of DNA repair. Another subunit of the complex, Nse1, is a conserved member of this complex and is also required for this response. We propose that the failure to maintain a checkpoint response stems from the lack of ongoing DNA repair or from defective chromosomal organization, which is the signal to maintain a checkpoint arrest. The Smc5/6 complex is fundamental to genome integrity and may function with the condensin and cohesin complexes in a coordinated manner.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA/fisiologia , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Ciclo Celular , Proteínas de Ciclo Celular/genética , Dano ao DNA/genética , Reparo do DNA/genética , Reparo do DNA/fisiologia , DNA Fúngico/genética , DNA Fúngico/metabolismo , Dados de Sequência Molecular , Mutação , Schizosaccharomyces/citologia , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/genética , Homologia de Sequência de Aminoácidos
19.
Oncogene ; 22(24): 3721-33, 2003 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-12802279

RESUMO

2-Aminopurine (2-AP) is an adenine analog shown to cause cells to bypass chemical- and radiation-induced cell cycle arrest through as-yet unidentified mechanisms. 2-AP has also been shown to act as a kinase inhibitor. Tumor suppressor p53 plays an important role in the control of cell cycle and apoptosis in response to genotoxic stress. We were interested in examining the effect of 2-AP on p53 phosphorylation and its possible consequences on checkpoint control in cells subjected to various forms of DNA damage. Here, we show that 2-AP suppresses p53 phosphorylation in response to gamma radiation, adriamycin, or ultraviolet treatment. This is partly explained by the ability of the kinase inhibitor to inhibit ATM or ATR activities in vitro and impair ATM- or ATR-dependent p53 phosphorylation in vivo. However, 2-AP is also capable of inhibiting p53 phosphorylation in cells deficient in ATM, DNA-PK, or ATR suggesting the existence of multiple pathways by which this kinase inhibitor modulates p53 activation. Biologically, the 2-AP-mediated inhibition of p53 stabilization enables wild-type p53-containing cells to bypass adriamycin-induced G(2)/M arrest. In the long term, however, 2-AP facilitates cells to resist DNA damage-induced cell death independently of p53.


Assuntos
2-Aminopurina/farmacologia , Apoptose , Proteínas de Ciclo Celular , Dano ao DNA , Proteínas de Ligação a DNA , Inibidores Enzimáticos/farmacologia , Inibidores de Proteínas Quinases , Células 3T3 , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteína Quinase Ativada por DNA , Camundongos , Fosforilação , Proteínas Serina-Treonina Quinases/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor
20.
Int Rev Cytol ; 222: 99-140, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12503848

RESUMO

Cellular reproduction, at its basic level, is simply the passing of genetic information from a single parent cell into two daughter cells. As the cellular genome encodes all the information that defines a cell, it is crucial that the genome be accurately replicated. Furthermore, the duplicated genome must be properly segregated so that each daughter cell contains the exact same information as the parent cell. The processes by which this occurs is known as the cell cycle. The failure of either duplication or segregation of the genome can have disastrous consequences for an organism, including cancer and death. This article discusses what is known about checkpoints, the surveillance mechanisms that monitor both the fidelity and accuracy of DNA replication and segregation. Specifically, we will focus on the G2 checkpoint that is responsible for ensuring proper segregation of the duplicated genome into the daughter cells and how this checkpoint functions to arrest entry into mitosis in response to DNA damage.


Assuntos
Dano ao DNA , Proteínas de Ligação a DNA , Fase G2 , Proteínas de Schizosaccharomyces pombe , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Enzimas Reparadoras do DNA , Endonucleases/genética , Endonucleases/metabolismo , Células Eucarióticas/citologia , Fase G2/genética , Fase G2/fisiologia , Genes cdc , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Proteínas de Saccharomyces cerevisiae , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...