Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Transplant Direct ; 7(7): e709, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34124345

RESUMO

Renal dysfunction is associated with poor long-term outcomes after liver transplantation. We examined the renal sparing effect of everolimus (EVR) compared to standard calcineurin inhibitor (CNI) immunosuppression with direct measurements of renal function over 24 months. METHODS: This was a prospective, randomized, open-label trial comparing EVR and mycophenolic acid (MPA) with CNI and MPA immunosuppression. An Investigational New Drug Application (IND # 113882) was obtained with the Food and Drug Administration as EVR is only approved for use with low-dose tacrolimus. Serum creatinine, 24-hour urine creatinine clearance, iothalamate clearance, Cockcroft-Gault creatinine clearance (CrCl), and Modification of Diet in Renal Disease estimated glomerular filtration rate were prospectively measured at 4 study visits. Nonparametric statistical tests were used for analyses, including the Mann-Whitney U test for continuous outcomes and Pearson's chi-square test for binary outcomes. Effect size was measured using Cohen's d. Patients also completed quality of life surveys using the FACT-Hep instrument at each study visit. Comparison between the 2 groups was performed using the Student t test. RESULTS: Each arm had 12 subjects; 4 patients dropped out in the EVR arm and 1 in the CNI arm by 24 months. Serum creatinine (P = 0.015), Modification of Diet in Renal Disease estimated glomerular filtration rate (P = 0.013), and 24-hour urine CrCL (P = 0.032) were significantly better at 24 months with EVR. Iothalamate clearance showed significant improvement at 12 months (P = 0.049) and a trend toward better renal function (P = 0.099) at 24 months. There was no statistical significance with Cockcroft-Gault CrCl. Adverse events were not significantly different between the 2 arms. The EVR group also showed significantly better physical, functional, and overall self-reported quality of life (P = 0.01) at 24 months. CONCLUSIONS: EVR with MPA resulted in significant long-term improvement in renal function and quality of life at 24 months after liver transplantation compared with standard CNI with MPA immunosuppression.

2.
Exp Clin Transplant ; 18(3): 345-352, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-30295586

RESUMO

OBJECTIVES: Newly developed, direct-acting antiviral therapy is effective in over 90% of cases to eradicate hepatitis C virus infection. Direct-acting antiviral therapy is also effective in liver transplant recipients with recurrent hepatitis C virus infection. However, hepatic function after sustained virologic response in transplant recipients is unknown. Here, we aimed to uncover the incidence of hepatic dysfunction in this patient group at our center. MATERIALS AND METHODS: Our study included 40 consecutive (January 2014 to February 2016) and compliant posttransplant recipients who achieved sustained viral response from direct-acting antiviral therapy. Patients were investigated for incidence and causes of hepatic dysfunction. RESULTS: In our patient group, 4 (10%) experienced hepatic dysfunction with stable baseline immunosuppression, with 2 having drastic increases in alanine aminotransferase at 15 and 32 weeks after direct-acting antiviral therapy. Biopsies showed hepatitis, and both patients were treated with hydrocortisone, which increased their baseline immunosuppression. The 3rd patient had an increase in bilirubin at 21 weeks posttherapy, with biopsy showing macrovascular steatosis. The 4th patient had a rapid increase in bilirubin at 7 weeks after direct-acting antiviral therapy, with biopsy showing significant duct loss. CONCLUSIONS: During the study period, 10% of patients experienced hepatic dysfunction after sustained viral response. Presumed causative factors included partial immune reconstitution and nonalcoholic fatty liver disease.


Assuntos
Antivirais/uso terapêutico , Hepatite C Crônica/terapia , Transplante de Fígado/efeitos adversos , Complicações Pós-Operatórias/epidemiologia , Adulto , Idoso , Feminino , Hepatite C Crônica/epidemiologia , Hepatite C Crônica/imunologia , Hepatite C Crônica/virologia , Humanos , Hospedeiro Imunocomprometido , Imunossupressores/efeitos adversos , Incidência , Testes de Função Hepática , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/diagnóstico , Complicações Pós-Operatórias/imunologia , Recidiva , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Resposta Viral Sustentada , Fatores de Tempo , Resultado do Tratamento
3.
Hepatol Int ; 13(2): 190-198, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30680672

RESUMO

BACKGROUND: Recently, a controversy has emerged: is the rate of recurrence of hepatocellular carcinoma (HCC) higher following treatment of hepatitis C virus (HCV) with direct-acting antiviral (DAA) therapy? However, the risk of HCC recurrence has not been studied in liver transplant (LTx) recipients who received DAA therapy. The aim of the present study is to compare the rate of HCC recurrence in LTx recipients who did or did not receive DAA therapy. PATIENTS AND METHODS: Sixty-three patients received LTx with HCC. Twenty-seven (42.9%) with HCV received DAA therapy (Group A), 20 (31.7%) with HCV did not receive DAA therapy (Group B), and 16 (25.4%) did not have HCV (Group C). RESULTS: In group A, three (11%), in group B, one (5%), and in group C, none had recurrence of HCC. Actuarial 4-year recurrence-free survival was 88.9, 95, and 100% in group A, B, and C, respectively (p = 0.37). Group A was subdivided into two groups for comparison with Group B: A1 included five patients who had end of treatment response (ETR) without sustained virological response (SVR), and A2 included 20 patients who achieved SVR. Three patients from A1 had HCC recurrence and no patients from A2 had HCC recurrence. (p = 0.0038; group A1, A2, and B). CONCLUSIONS: The rate of HCC recurrence in LTx patients with DAA therapy was significantly higher with ETR, without SVR, after DAA therapy compared to patients with SVR or patients who did not receive DAA therapy. LTx recipients with HCC receiving DAA therapy requires further studies.


Assuntos
Antivirais/uso terapêutico , Carcinoma Hepatocelular/secundário , Carcinoma Hepatocelular/cirurgia , Hepatite C Crônica/tratamento farmacológico , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/cirurgia , Transplante de Fígado , Recidiva Local de Neoplasia/virologia , Idoso , Benzimidazóis/uso terapêutico , Carcinoma Hepatocelular/virologia , Intervalo Livre de Doença , Quimioterapia Combinada , Feminino , Fluorenos/uso terapêutico , Hepatite C Crônica/complicações , Humanos , Neoplasias Hepáticas/virologia , Masculino , Pessoa de Meia-Idade , Ribavirina/uso terapêutico , Simeprevir/uso terapêutico , Sofosbuvir/uso terapêutico , Resposta Viral Sustentada
4.
Clin Transplant ; 28(7): 797-801, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24779669

RESUMO

The efficacy and safety of everolimus (EVR) in simultaneous pancreas and kidney transplantation (SPKT) is unclear. We retrospectively evaluated 25 consecutive SPKT recipients at our center from November 2011 to March 2013. All patients received dual induction (Thymoglobulin/basiliximab) and low-dose tacrolimus plus corticosteroids. Nine patients who received EVR were compared with 14 patients who received enteric-coated mycophenolate sodium (EC-MPS); two patients who received sirolimus were excluded from the analysis. With a median follow-up of 14 months, the pancreas graft survival rate was 100% in both groups, and the kidney graft survival rate was 100% and 93% in EVR and EC-MPS patients, respectively. One EC-MPS patient lost her kidney graft from proteinuric kidney disease. Another EC-MPS patient received treatment for clinically diagnosed pancreas and kidney graft rejection. No rejection was observed in EVR patients. Serum creatinine and HbA1c levels were similar between the groups. There was no significant difference of surgical or medical complications. In conclusion, EVR seems to provide comparable short-term outcome to EC-MPS when combined with low-dose tacrolimus/steroids and dual induction therapy. A larger study with a longer follow-up is required to further assess this combination.


Assuntos
Rejeição de Enxerto/prevenção & controle , Nefropatias/cirurgia , Transplante de Rim , Transplante de Pâncreas , Pancreatopatias/cirurgia , Sirolimo/análogos & derivados , Tacrolimo/administração & dosagem , Adulto , Relação Dose-Resposta a Droga , Everolimo , Feminino , Seguimentos , Taxa de Filtração Glomerular , Rejeição de Enxerto/mortalidade , Sobrevivência de Enxerto , Humanos , Imunossupressores/administração & dosagem , Nefropatias/mortalidade , Testes de Função Renal , Masculino , Pessoa de Meia-Idade , Pancreatopatias/mortalidade , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Sirolimo/administração & dosagem , Taxa de Sobrevida , Adulto Jovem
5.
Transpl Immunol ; 30(2-3): 88-92, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24518158

RESUMO

The Cylex ImmuKnow assay measures the amount of stimulated ATP production by CD4+ T-cells, and has been used clinically, trying to predict rejection and infection episodes. However, predictive values of this assay after induction therapy with steroid-sparing maintenance protocols are unclear. In this single-center cohort study, we analyzed renal transplant recipients who received T-cell depleting+/-anti-IL2 receptor antibodies and tacrolimus/mycophenolate maintenance without steroids. A total of 4224 ImmuKnow levels in 306 patients were available for analysis. ImmuKnow levels (Mean ± SE) changed over time after induction therapy with a paradoxical initial increase: 419 ± 23, 461 ± 32, 519 ± 14, 411 ± 10, 344 ± 6, and 405 ± 3 for pre-transplant, 0-1 wk, 1 wk-1 mo, 1-3 mos, 3 mos-1 yr, and thereafter. This change was parallel to the evolution of peripheral WBC counts and ImmuKnow levels had weak but significant correlation with WBC counts (R(2)=0.264, P<0.0001). The levels for biopsy-proven rejection (389 ± 56) and borderline/clinical rejection (254 ± 41) were not significantly higher than the levels of quiescent patients. The levels for opportunistic infection (349 ± 48) and other infections (345 ± 27) were not significantly lower than the levels of quiescent patients. The longitudinal changes in ImmuKnow levels were not predictive of rejection or infection. In conclusion, ImmuKnow levels can vary after T-cell depleting induction therapies at various time points, even without significant clinical events. Since ImmuKnow levels seem to be affected by WBC counts, ImmuKnow results need to be interpreted with caution. The effects of leukocytosis or leukopenia caused by immunosuppressive medication on the ImmuKnow assay need further investigation.


Assuntos
Rejeição de Enxerto , Terapia de Imunossupressão/métodos , Imunossupressores/administração & dosagem , Transplante de Rim , Depleção Linfocítica , Adulto , Feminino , Rejeição de Enxerto/sangue , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Humanos , Contagem de Leucócitos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
6.
Cancer Cell ; 22(3): 331-44, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22975376

RESUMO

Tumors successfully adapt to constantly changing intra- and extracellular environments, but the wirings of this process are still largely elusive. Here, we show that heat-shock-protein-90-directed protein folding in mitochondria, but not cytosol, maintains energy production in tumor cells. Interference with this process activates a signaling network that involves phosphorylation of nutrient-sensing AMP-activated kinase, inhibition of rapamycin-sensitive mTOR complex 1, induction of autophagy, and expression of an endoplasmic reticulum unfolded protein response. This signaling network confers a survival and proliferative advantage to genetically disparate tumors, and correlates with worse outcome in lung cancer patients. Therefore, mitochondrial heat shock protein 90s are adaptive regulators of tumor bioenergetics and tractable targets for cancer therapy.


Assuntos
Metabolismo Energético , Proteínas de Choque Térmico HSP90/metabolismo , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Resposta a Proteínas não Dobradas , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Autofagia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Guanidinas/farmacologia , Proteínas de Choque Térmico/genética , Humanos , Cinesinas/genética , Lactamas Macrocíclicas/farmacologia , Neoplasias Pulmonares , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos , Neoplasias/patologia , Fosforilação , Dobramento de Proteína , Proteínas Serina-Treonina Quinases/genética , Proteínas/antagonistas & inibidores , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Serina-Treonina Quinases TOR
7.
Cancer Res ; 72(13): 3251-9, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22586065

RESUMO

Survivin is an oncogene that functions in cancer cell cytoprotection and mitosis. Here we report that differential expression in cancer cells of a C-terminal splice variant of survivin, termed survivin-ΔEx3, is tightly associated with aggressive disease and markers of unfavorable prognosis. In contrast to other survivin variants, survivin-ΔEx3 localized exclusively to nuclei in tumor cells and was phosphorylated at multiple residues by the checkpoint kinase Chk2 during DNA damage. Mutagenesis of the Chk2 phosphorylation sites enhanced the stability of survivin-ΔEx3 in tumor cells, inhibited the expression of phosphorylated H2AX (γH2AX) in response to double-strand DNA breaks, and impaired growth after DNA damage. DNA damage induced Chk2 phosphorylation, stabilization of p53, induction of the cyclin-dependent kinase inhibitor p21, and homologous recombination-induced repair were not affected. In vivo, active Chk2 was detected at the earliest stages of the colorectal adenoma-to-carcinoma transition, persisted in advanced tumors, and correlated with increased survivin expression. Together, our findings suggest that Chk2-mediated phosphorylation of survivin-ΔEx3 contributes to a DNA damage-sensing checkpoint that may affect cancer cell sensitivity to genotoxic therapies.


Assuntos
Dano ao DNA , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Quinase do Ponto de Checagem 2 , Primers do DNA , Humanos , Mutagênese , Neoplasias/patologia , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Frações Subcelulares/metabolismo , Survivina
8.
PLoS One ; 6(11): e27879, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22114719

RESUMO

The small GTPase Ran orchestrates pleiotropic cellular responses of nucleo-cytoplasmic shuttling, mitosis and subcellular trafficking, but whether deregulation of these pathways contributes to disease pathogenesis has remained elusive. Here, we generated transgenic mice expressing wild type (WT) Ran, loss-of-function Ran T24N mutant or constitutively active Ran G19V mutant in pancreatic islet ß cells under the control of the rat insulin promoter. Embryonic pancreas and islet development, including emergence of insulin(+) ß cells, was indistinguishable in control or transgenic mice. However, by one month after birth, transgenic mice expressing any of the three Ran variants exhibited overt diabetes, with hyperglycemia, reduced insulin production, and nearly complete loss of islet number and islet mass, in vivo. Deregulated Ran signaling in transgenic mice, adenoviral over-expression of WT or mutant Ran in isolated islets, or short hairpin RNA (shRNA) silencing of endogenous Ran in model insulinoma INS-1 cells, all resulted in decreased expression of the pancreatic and duodenal homeobox transcription factor, PDX-1, and reduced ß cell proliferation, in vivo. These data demonstrate that a finely-tuned balance of Ran GTPase signaling is essential for postnatal pancreatic islet development and glucose homeostasis, in vivo.


Assuntos
Diabetes Mellitus/etiologia , Embrião de Mamíferos/citologia , Hiperglicemia/etiologia , Hiperinsulinismo/etiologia , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Proteína ran de Ligação ao GTP/fisiologia , Animais , Células Cultivadas , DNA/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Embrião de Mamíferos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Glucose/metabolismo , Humanos , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patologia , Técnicas Imunoenzimáticas , Insulina/sangue , Insulina/genética , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Plasmídeos/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/genética , Ratos , Proteína ran de Ligação ao GTP/antagonistas & inibidores
9.
Am J Pathol ; 178(6): 2478-83, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21549346

RESUMO

Human Scribble (Scrib) is an evolutionary-conserved cell polarity protein, but its potential role in human cancer is controversial. Herein, we show that Scrib is nearly universally overexpressed in cultured tumor cell lines and genetically disparate cancer patient series compared with matched normal tissues in vivo. Instead of a membrane association seen in normal epithelia, tumor-associated Scrib is mislocalized and found predominantly in the cytosol. Small-interfering RNA silencing of Scrib in model lung adenocarcinoma A549 cells inhibited cell migration in wound-healing assays, suppressed tumor cell invasion across Matrigel-coated inserts, and down-regulated the expression of cell motility markers and mediators of epithelial-mesenchymal transition. These data uncover a previously unrecognized exploitation of Scrib for aberrant tumor cell motility and invasion, thus potentially contributing to disease progression in humans.


Assuntos
Polaridade Celular , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Supressoras de Tumor/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Polaridade Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Membrana/genética , Neoplasias/genética , Análise Serial de Tecidos , Proteínas Supressoras de Tumor/genética
10.
J Biol Chem ; 286(19): 16758-67, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21454573

RESUMO

Survivin is a multifunctional protein with essential roles in cell division and inhibition of apoptosis, but the molecular underpinnings of its cytoprotective properties are poorly understood. Here we show that homozygous deletion of the aryl hydrocarbon receptor-interacting protein (AIP), a survivin-associated immunophilin, causes embryonic lethality in mice by embryonic day 13.5-14, increased apoptosis of Ter119(-)/CD71(-) early erythropoietic progenitors, and loss of survivin expression in its cytosolic and mitochondrial compartments in vivo. In import assays using recombinant proteins, AIP directly mediated the import of survivin to mitochondria, thus enabling its anti-apoptotic function, whereas a survivin 1-141 mutant that does not bind AIP was not imported to mitochondria and failed to inhibit apoptosis. AIP-directed mitochondrial import of survivin did not affect cell division, was independent of the organelle transmembrane potential, did not require the chaperone Heat Shock Protein 90 (Hsp90), and was inhibited by cytosolic factor(s) present in normal cells. shRNA knockdown of the mitochondrial import receptor Tom20 abolished mitochondrial import of survivin and sensitized tumor cells to apoptosis, whereas silencing of Tom70 had no effect. Therefore, an AIP-Tom20 recognition contributes to cell survival in development and cancer by mediating the mitochondrial import of survivin.


Assuntos
Apoptose , Proteínas Inibidoras de Apoptose/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Mitocôndrias/metabolismo , Proteínas Repressoras/metabolismo , Animais , Transporte Biológico , Citosol/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Proteínas de Choque Térmico HSP90/metabolismo , Células HeLa , Homozigoto , Humanos , Camundongos , Camundongos Knockout , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Survivina , Fatores de Tempo
11.
J Clin Invest ; 121(4): 1349-60, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21364280

RESUMO

Fine tuning of the protein folding environment in subcellular organelles, such as mitochondria, is important for adaptive homeostasis and may participate in human diseases, but the regulators of this process are still largely elusive. Here, we have shown that selective targeting of heat shock protein-90 (Hsp90) chaperones in mitochondria of human tumor cells triggered compensatory autophagy and an organelle unfolded protein response (UPR) centered on upregulation of CCAAT enhancer binding protein (C/EBP) transcription factors. In turn, this transcriptional UPR repressed NF-κB-dependent gene expression, enhanced tumor cell apoptosis initiated by death receptor ligation, and inhibited intracranial glioblastoma growth in mice without detectable toxicity. These data reveal what we believe to be a novel role of Hsp90 chaperones in the regulation of the protein-folding environment in mitochondria of tumor cells. Disabling this general adaptive pathway could potentially be used in treatment of genetically heterogeneous human tumors.


Assuntos
Mitocôndrias/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Resposta a Proteínas não Dobradas , Animais , Apoptose , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular Tumoral , Expressão Gênica , Técnicas de Silenciamento de Genes , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Guanidinas/farmacologia , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Lactamas Macrocíclicas/farmacologia , Camundongos , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , NF-kappa B/metabolismo , Neoplasias/genética , RNA Interferente Pequeno/genética , Transdução de Sinais , Estresse Fisiológico , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
12.
Cancer Res ; 70(22): 8988-93, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20978188

RESUMO

Mitochondrial apoptosis plays a critical role in tumor maintenance and dictates the response to therapy in vivo; however, the regulators of this process are still largely elusive. Here, we show that the molecular chaperone heat shock protein 60 (Hsp60) directly associates with cyclophilin D (CypD), a component of the mitochondrial permeability transition pore. This interaction occurs in a multichaperone complex comprising Hsp60, Hsp90, and tumor necrosis factor receptor-associated protein-1, selectively assembled in tumor but not in normal mitochondria. Genetic targeting of Hsp60 by siRNA triggers CypD-dependent mitochondrial permeability transition, caspase-dependent apoptosis, and suppression of intracranial glioblastoma growth in vivo. Therefore, Hsp60 is a novel regulator of mitochondrial permeability transition, contributing to a cytoprotective chaperone network that antagonizes CypD-dependent cell death in tumors.


Assuntos
Chaperonina 60/metabolismo , Ciclofilinas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Neoplasias/metabolismo , Animais , Apoptose , Western Blotting , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Chaperonina 60/genética , Peptidil-Prolil Isomerase F , Citometria de Fluxo , Células HCT116 , Células HEK293 , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Nus , Poro de Transição de Permeabilidade Mitocondrial , Neoplasias/genética , Neoplasias/patologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Ligação Proteica , Interferência de RNA , Transplante Heterólogo
13.
Clin Cancer Res ; 16(19): 4779-88, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20876793

RESUMO

PURPOSE: This study aimed to characterize the preclinical activity of the first class of combinatorial, mitochondria-targeted, small molecule heat shock protein-90 (Hsp90) inhibitors, gamitrinibs, in models of hormone-refractory, drug-resistant, localized, and bone metastatic prostate cancer in vivo. EXPERIMENTAL DESIGN: Mitochondrial permeability transition, apoptosis, and changes in metabolic activity were examined by time-lapse videomicroscopy, multiparametric flow cytometry, MTT, and analysis of isolated mitochondria. Drug-resistant prostate cancer cells were generated by chronic exposure of hormone-refractory PC3 cells to the Hsp90 inhibitor 17-allylaminogeldanamycin (17-AAG). The effect of gamitrinibs on s.c. or intratibial prostate cancer growth was studied in xenograft models. Bone metastatic tumor growth and bone parameters were quantified by micro-computed tomography imaging. RESULTS: In the NCI 60-cell line screening, gamitrinibs were active against all tumor cell types tested, and efficiently killed metastatic, hormone-refractory, and multidrug-resistant prostate cancer cells characterized by overexpression of the ATP binding cassette transporter P-glycoprotein. Mechanistically, gamitrinibs, but not 17-AAG, induced acute mitochondrial dysfunction in prostate cancer cells with loss of organelle membrane potential, release of cytochrome c, and caspase activity, independently of proapoptotic Bcl-2 proteins Bax and Bak. Systemic administration of gamitrinibs to mice was well tolerated, and inhibited s.c. or bone metastatic prostate cancer growth in vivo. CONCLUSIONS: Gamitrinibs have preclinical activity and favorable safety in models of drug-resistant and bone metastatic prostate cancer in vivo.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Lactamas/farmacologia , Mitocôndrias/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Pirimidinas/farmacologia , Quinonas/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Benzoquinonas/farmacologia , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Lactamas/síntese química , Lactamas/química , Lactamas Macrocíclicas/farmacologia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos SCID , Peso Molecular , Neoplasias da Próstata/metabolismo , Pirimidinas/síntese química , Pirimidinas/química , Quinonas/síntese química , Quinonas/química , Células Tumorais Cultivadas , Microtomografia por Raio-X
14.
Cancer Cell ; 17(1): 53-64, 2010 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-20129247

RESUMO

Inhibitor-of-Apoptosis (IAP) proteins contribute to tumor progression, but the requirements of this pathway are not understood. Here, we show that intermolecular cooperation between XIAP and survivin stimulates tumor cell invasion and promotes metastasis. This pathway is independent of IAP inhibition of cell death. Instead, a survivin-XIAP complex activates NF-kappaB, which in turn leads to increased fibronectin gene expression, signaling by beta1 integrins, and activation of cell motility kinases FAK and Src. Therefore, IAPs are direct metastasis genes, and their antagonists could provide antimetastatic therapies in patients with cancer.


Assuntos
Proteínas Inibidoras de Apoptose/metabolismo , Invasividade Neoplásica , Transdução de Sinais/fisiologia , Animais , Western Blotting , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , RNA Interferente Pequeno , Survivina
15.
Diabetes ; 57(10): 2718-27, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18599523

RESUMO

OBJECTIVE: Diabetes results from a deficiency of functional beta-cells due to both an increase in beta-cell death and an inhibition of beta-cell replication. The molecular mechanisms responsible for these effects in susceptible individuals are mostly unknown. The objective of this study was to determine whether a gene critical for cell division and cell survival in cancer cells, survivin, might also be important for beta-cells. RESEARCH DESIGN AND METHODS: We generated mice harboring a conditional deletion of survivin in pancreatic endocrine cells using mice with a Pax-6-Cre transgene promoter construct driving tissue-specific expression of Cre-recombinase in these cells. We performed metabolic studies and immunohistochemical analyses to determine the effects of a mono- and biallelic deletion of survivin. RESULTS: Selective deletion of survivin in pancreatic endocrine cells in the mouse had no discernible effects during embryogenesis but was associated with striking decreases in beta-cell number after birth, leading to hyperglycemia and early-onset diabetes by 4 weeks of age. Serum insulin levels were significantly decreased in animals lacking endocrine cell survivin, with relative stability of other hormones. Exogenous expression of survivin in mature beta-cells lacking endogenous survivin completely rescued the hyperglycemic phenotype and the decrease in beta-cell mass, confirming the specificity of the survivin effect in these cells. CONCLUSIONS: Our findings implicate survivin in the maintenance of beta-cell mass through both replication and antiapoptotic mechanisms. Given the widespread involvement of survivin in cancer, a novel role for survivin may well be exploited in beta-cell regulation in diseased states, such as diabetes.


Assuntos
Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Animais , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Feminino , Deleção de Genes , Glucagon/metabolismo , Imuno-Histoquímica , Proteínas Inibidoras de Apoptose , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Repressoras , Somatostatina/metabolismo , Survivina
16.
J Biol Chem ; 283(8): 5188-94, 2008 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-18086682

RESUMO

Molecular chaperones may promote cell survival, but how this process is regulated, especially in cancer, is not well understood. Using high throughput proteomics screening, we identified the cell cycle regulator and apoptosis inhibitor survivin as a novel protein associated with the molecular chaperone Hsp60. Acute ablation of Hsp60 by small interfering RNA destabilizes the mitochondrial pool of survivin, induces mitochondrial dysfunction, and activates caspase-dependent apoptosis. This response involves disruption of an Hsp60-p53 complex, which results in p53 stabilization, increased expression of pro-apoptotic Bax, and Bax-dependent apoptosis. In vivo, Hsp60 is abundantly expressed in primary human tumors, as compared with matched normal tissues, and small interfering RNA ablation of Hsp60 in normal cells is well tolerated and does not cause apoptosis. Therefore, Hsp60 orchestrates a broad cell survival program centered on stabilization of mitochondrial survivin and restraining of p53 function, and this process is selectively exploited in cancer. Hsp60 inhibitors may function as attractive anticancer agents by differentially inducing apoptosis in tumor cells.


Assuntos
Apoptose , Ciclo Celular , Chaperonina 60/metabolismo , Neoplasias/metabolismo , Apoptose/genética , Caspases/genética , Caspases/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Chaperonina 60/antagonistas & inibidores , Chaperonina 60/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Inibidoras de Apoptose , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Proteômica , RNA Interferente Pequeno/genética , Survivina , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
17.
Cell ; 131(2): 257-70, 2007 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17956728

RESUMO

Molecular chaperones, especially members of the heat shock protein 90 (Hsp90) family, are thought to promote tumor cell survival, but this function is not well understood. Here, we show that mitochondria of tumor cells, but not most normal tissues, contain Hsp90 and its related molecule, TRAP-1. These chaperones interact with Cyclophilin D, an immunophilin that induces mitochondrial cell death, and antagonize its function via protein folding/refolding mechanisms. Disabling this pathway using novel Hsp90 ATPase antagonists directed to mitochondria causes sudden collapse of mitochondrial function and selective tumor cell death. Therefore, Hsp90-directed chaperones are regulators of mitochondrial integrity, and their organelle-specific antagonists may provide a previously undescribed class of potent anticancer agents.


Assuntos
Proteínas de Choque Térmico HSP90/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Mitocôndrias/fisiologia , Organelas/fisiologia , Animais , Apoptose , Benzoquinonas/síntese química , Benzoquinonas/farmacologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Peptidil-Prolil Isomerase F , Ciclofilinas/metabolismo , Desenho de Fármacos , Homeostase , Humanos , Imunoglobulina G , Lactamas Macrocíclicas/síntese química , Lactamas Macrocíclicas/farmacologia , Melfalan , Potencial da Membrana Mitocondrial , Camundongos , Especificidade de Órgãos , Fragmentos de Peptídeos/farmacologia , Dobramento de Proteína
18.
Mol Cell ; 27(1): 17-28, 2007 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-17612487

RESUMO

Cell death pathways are likely regulated in specialized subcellular microdomains, but how this occurs is not understood. Here, we show that cyclic AMP-dependent protein kinase A (PKA) phosphorylates the inhibitor of apoptosis (IAP) protein survivin on Ser20 in the cytosol, but not in mitochondria. This phosphorylation event disrupts the binding interface between survivin and its antiapoptotic cofactor, XIAP. Conversely, mitochondrial survivin or a non-PKA phosphorylatable survivin mutant binds XIAP avidly, enhances XIAP stability, synergistically inhibits apoptosis, and accelerates tumor growth, in vivo. Therefore, differential phosphorylation of survivin by PKA in subcellular microdomains regulates tumor cell apoptosis via its interaction with XIAP.


Assuntos
Compartimento Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citoproteção , Proteínas Inibidoras de Apoptose/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Humanos , Proteínas Inibidoras de Apoptose/química , Camundongos , Proteínas Associadas aos Microtúbulos/química , Mitocôndrias/metabolismo , Neoplasias/patologia , Fosforilação , Estrutura Secundária de Proteína , Serina/metabolismo , Transplante Heterólogo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
19.
Cancer Res ; 66(24): 11576-9, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17178848

RESUMO

Tumor cells often become resistant to DNA damage-based therapy; however, the underlying mechanisms are not yet understood. Here, we show that tumor cells exposed to DNA damage counteract cell death by releasing the antiapoptotic protein, survivin, from mitochondria. This is independent of p53, and requires activated checkpoint kinase 2 (Chk2), a putative tumor suppressor. Molecular or genetic targeting of Chk2 prevents the release of survivin from mitochondria, enhances DNA damage-induced tumor cell apoptosis, and inhibits the growth of resistant in vivo tumors. Therefore, activated Chk2 circumvents its own tumor-suppressive functions by promoting tumor cell survival. Inhibiting Chk2 in combination with DNA-damaging agents may provide a rational approach for treating resistant tumors.


Assuntos
Sobrevivência Celular/fisiologia , Dano ao DNA , Proteínas Serina-Treonina Quinases/metabolismo , Adenocarcinoma , Animais , Apoptose , Neoplasias da Mama , Linhagem Celular Tumoral , Quinase do Ponto de Checagem 2 , Neoplasias do Colo , Ativação Enzimática , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos SCID , Neoplasias da Próstata , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/genética , Transfecção , Transplante Heterólogo
20.
Int J Oncol ; 28(4): 793-8, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16525626

RESUMO

Cell-specific, replicating viruses are being developed as a new class of oncolytic agents. A novel approach to viral gene therapy with the use of replication-competent herpes simplex virus has been described; G92A is a replication-competent, multimutant oncolytic herpes simplex virus (HSV) that has been evaluated for anticancer effects and selectivity in the treatment of subcutaneous tumors. G92A replicates efficiently in albumin-producing tumor cell lines but not in non-albumin-producing tumor cell lines, whereas both types are equally susceptible to a non-tissue-specific recombinant HSV, hrR3. In this study, we analyzed the antitumoral efficacy of a single intrasplenic G92A or hrR3 injection in nude mice. In vivo, G92A replicated well in liver xenografts of human albumin-producing hepatoma cells (Hep3B) but not in liver xenografts of a non-albumin-producing malignant colon tumor cell line (HT29), whereas hrR3 replicated well in both tumor types. G92A effectively and selectively replicated throughout liver tumors without apparent hepatotoxicity and inhibited tumor growth, leading to a significantly increased survival time. By monitoring lacZ histochemical staining, we determined the oncolytic potential of recombinant HSV against liver tumors. Our results indicate that G92A warrants further investigation as a clinical therapy against malignant liver tumors.


Assuntos
Neoplasias Hepáticas Experimentais/terapia , Simplexvirus/fisiologia , Animais , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Chlorocebus aethiops , DNA Recombinante/administração & dosagem , DNA Recombinante/genética , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Vírus Defeituosos/fisiologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Células HT29 , Humanos , Fígado/patologia , Fígado/virologia , Neoplasias Hepáticas Experimentais/mortalidade , Neoplasias Hepáticas Experimentais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Simplexvirus/genética , Simplexvirus/imunologia , Taxa de Sobrevida , Células Vero , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...