Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Int J Pharm ; 585: 119535, 2020 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-32534162

RESUMO

Induction of apoptosis in tumor cells specifically within the complex tumor microenvironment is highly desirable to kill them efficiently and to enhance the effects of chemotherapy. Second mitochondria-derived activator of caspase (Smac) is a key pro-apoptotic pathway which can be activated with a Smac mimetic peptide. However, in vivo application of peptides is hampered by several limitations such as poor pharmacokinetics, rapid elimination, enzymatic degradation, and insufficient intracellular delivery. In this study, we developed a nanosystem to deliver a Smac peptide to tumor by passive targeting. We first synthesized a chimeric peptide that consists of the 8-mer Smac peptide and a 14-mer cell penetrating peptide (CPP) and then encapsulated the Smac-CPP into polymeric nanoparticles (Smac-CPP-NPs). In vitro, Smac-CPP-NPs were rapidly internalized by 4T1 mammary tumor cells and subsequently released Smac-CPP into the cells, as shown with fluorescence microscopy. Furthermore, Smac-CPP-NPs induced apoptosis in tumor cells, as confirmed with cell viability and caspase 3/7 assays. Interestingly, combination of Smac-CPP-NPs with doxorubicin (dox), a clinically used cytostatic drug, showed combined effects in vitro in 4T1 cells. The effect was significantly better than that of SMAC-CPP-NPs alone as well as empty nanoparticles and dox. In vivo, co-treatment with Smac-CPP-NPs and free dox reduced the tumor growth to 85%. Furthermore, the combination of Smac-CPP-NPs and free dox showed reduced proliferating tumor cells (Ki-67 staining) and increased apoptotic cells (cleaved caspase-3 staining) in tumors. In conclusion, the present study demonstrates that the intracellular delivery of Smac-mimetic peptide using nanoparticle system can be an interesting strategy to attenuate the tumor growth and to potentiate the therapeutic efficacy of chemotherapy in vivo.


Assuntos
Proteínas Reguladoras de Apoptose/farmacologia , Apoptose/efeitos dos fármacos , Peptídeos Penetradores de Células/farmacologia , Doxorrubicina/farmacologia , Proteínas Mitocondriais/farmacologia , Nanopartículas/química , Animais , Proteínas Reguladoras de Apoptose/administração & dosagem , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/administração & dosagem , Doxorrubicina/administração & dosagem , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/farmacologia , Concentração de Íons de Hidrogênio , Camundongos , Proteínas Mitocondriais/administração & dosagem , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química
2.
Reprod Sci ; 27(4): 1030-1036, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32124395

RESUMO

HSP60 is a mitochondrial chaperone protein that is associated with decreased overall survival of ovarian cancer patients. We determined whether targeting HSP60 with its monoclonal antibody would induce cytotoxicity in sensitive and chemoresistant ovarian cancer cells and whether it is synergistic when combined with chemotherapeutic drugs. Epithelial ovarian cancer (EOC) cells and their docetaxel- or cisplatin-resistant counterparts were utilized. HSP60 mRNA levels were determined by real-time RT-PCR. Cytotoxicity of HSP60 antibody (0.5 or 1.5 µg/ml) alone and in combination with chemotherapy were assessed by MTT Cell Proliferation Assay. Unpaired t tests were used to compare groups for real-time RT-PCR. One-way ANOVA followed by Tukey's post hoc tests with Bonferroni correction was performed for cytotoxicity comparisons. Significant synergistic effects of the antibody combined with chemotherapy were determined by the CompuSyn Software. Basal HSP60 mRNA levels were increased in chemoresistant EOC cells as compared with their sensitive counterparts (p < 0.05). There was no significant difference in cytotoxicity between EOC cell types; however, treatment with the HSP60 antibody for 24 h showed a dose response (0.5 and 1.5 µg/ml) cytotoxic effect to both sensitive and chemoresistant EOC cells as compared with the isotype control (p < 0.05). Importantly, treatment with both doses of HSP60 antibody was not cytotoxic to normal macrophages. Combination of the HSP60 antibody with docetaxel or cisplatin was significantly synergistic in both sensitive and chemoresistant EOC cells. Here, we identify a novel target that may serve not only for ovarian cancer treatment but also for sensitization of patients to chemotherapy. The cytotoxic effect of HSP60 monoclonal antibody and its synergism with chemotherapeutic agents highlight HSP60 as a promising target for therapy and chemosensitization in ovarian cancer treatment.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Carcinoma Epitelial do Ovário/tratamento farmacológico , Chaperonina 60/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteínas Mitocondriais/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Chaperonina 60/imunologia , Chaperonina 60/metabolismo , Cisplatino/administração & dosagem , Docetaxel/administração & dosagem , Quimioterapia Combinada/métodos , Feminino , Humanos , Proteínas Mitocondriais/imunologia , Proteínas Mitocondriais/metabolismo , RNA Mensageiro/metabolismo
3.
Eur J Pharmacol ; 870: 172909, 2020 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-31926126

RESUMO

The activation of the AMP activated protein kinase (AMPK) exerts antinociceptive effects in acute and neuropathic pain models. Mitochondrial open-reading-frame of the twelve S rRNA-c (MOTS-c), a mitochondrial-derived peptide, regulates many biological activities via activating AMPK. However, the role of MOTS-c in the formalin-induced inflammatory nociception remains unclear. In this study, we investigated the role of MOTS-c in the formalin-induced inflammatory nociception. The antinociceptive effect of MOTS-c was assessed by recording the time spent licking paw. The anti-inflammatory effect of MOTS-c was evaluated by detecting the inflammatory cytokine level changes in the mouse serum. Western blot was used to detect the changes of protein phosphorylation levels in the mouse spinal cord. Changes of c-fos expression in the spinal cord were assessed by immunohistochemistry. Our results showed that the intraperitoneal administration of MOTS-c reduced the time spent on licking in phase 2 in a dose-dependent manner in the formalin test. The antinociceptive effects of MOTS-c (50 mg/kg, i.p.) were attenuated by the AMPK antagonist compound C (10 mg/kg, i.p.). MOTS-c (50 mg/kg, i.p.) significantly reduced pro-inflammatory cytokine levels and elevated the level of anti-inflammatory cytokine in mouse serum. In addition, MOTS-c treatment significantly increased AMPKα phosphorylation level and suppressed formalin-induced extracellular signal-regulated kinase (ERK), c-Jun amino-terminal kinases (JNK), and P38 activation and c-fos expression in the mouse spinal cord. These results suggest that systemic administration of MOTS-c exerts antinociceptive and anti-inflammatory effects, at least partially, through activating AMPK pathway and inhibiting MAP kinases-c-fos signaling pathway in the mouse formalin test.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Analgésicos/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Proteínas Mitocondriais/administração & dosagem , Neuralgia/metabolismo , Animais , Citocinas/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Formaldeído/metabolismo , Regulação Enzimológica da Expressão Gênica , Humanos , Injeções Intraperitoneais , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos ICR , Nociceptividade/efeitos dos fármacos , Medição da Dor , Fosforilação , Proteínas Proto-Oncogênicas c-fos/metabolismo , Medula Espinal/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Cardiorenal Med ; 10(1): 42-50, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31694019

RESUMO

INTRODUCTION: Vascular calcification (VC) is a complex, regulated process involved in many disease entities. So far, there are no treatments to reverse it. Exploring novel strategies to prevent VC is important and necessary for VC-related disease intervention. OBJECTIVE: In this study, we evaluated whether MOTS-c, a novel mitochondria-related 16-aa peptide, can reduce vitamin D3 and nicotine-induced VC in rats. METHODS: Vitamin D3 plus nicotine-treated rats were injected with MOTS-c at a dose of 5 mg/kg once a day for 4 weeks. Blood pressure, heart rate, and body weight were measured, and echocardiography was performed. The expression of phosphorylated adenosine monophosphate-activated protein kinase (AMPK) and the angiotensin II type 1 (AT-1) and endothelin B (ET-B) receptors was determined by Western blot analysis. RESULTS: Our results showed that MOTS-c treatment significantly attenuated VC. Furthermore, we found that the level of phosphorylated AMPK was increased and the expression levels of the AT-1 and ET-B receptors were decreased after MOTS-c treatment. CONCLUSIONS: Our findings provide evidence that MOTS-c may act as an inhibitor of VC by activating the AMPK signaling pathway and suppressing the expression of the AT-1 and ET-B receptors.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Mitocondriais/metabolismo , Calcificação Vascular/metabolismo , Animais , Colecalciferol/administração & dosagem , Colecalciferol/efeitos adversos , Colecalciferol/metabolismo , Masculino , Proteínas Mitocondriais/administração & dosagem , Proteínas Mitocondriais/efeitos adversos , Proteínas Mitocondriais/farmacologia , Modelos Animais , Nicotina/administração & dosagem , Nicotina/efeitos adversos , Nicotina/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor de Endotelina B/efeitos dos fármacos , Receptor de Endotelina B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Calcificação Vascular/induzido quimicamente , Remodelação Ventricular/efeitos dos fármacos
5.
J Control Release ; 299: 107-120, 2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30797866

RESUMO

The tumor microenvironment of numerous prevalent cancer types is abundantly infiltrated with tumor-associated macrophages (TAMs). Macrophage mannose receptor (MMR or CD206) expressing TAMs have been shown to be key promoters of tumor progression and major opponents of successful cancer therapy. Therefore, depleting MMR+ TAMs is an interesting approach to synergize with current antitumor therapies. We studied the potential of single-domain antibodies (sdAbs) specific for MMR to target proteins to MMR+ TAMs. Anti-MMR sdAbs were genetically coupled to a reporter protein, mWasabi (wasabi green, WG), generating sdAb "drug" fusion proteins (SFPs), referred to as WG-SFPs. The resulting WG-SFPs were highly efficient in targeting MMR+ macrophages both in vitro and in vivo. As we showed that second mitochondria-derived activator of caspase (SMAC) mimetics modulate MMR+ macrophages, we further coupled the anti-MMR sdAb to an active form of SMAC, referred to as tSMAC. The resulting tSMAC-SFPs were able to bind and upregulate caspase3/7 activity in MMR+ macrophages in vitro. In conclusion, we report the proof-of-concept of an elegant approach to conjugate anti-MMR sdAbs to proteins, which opens new avenues for targeted manipulation of MMR+ tumor-promoting TAMs.


Assuntos
Sistemas de Liberação de Medicamentos , Lectinas Tipo C/metabolismo , Macrófagos/efeitos dos fármacos , Lectinas de Ligação a Manose/metabolismo , Receptores de Superfície Celular/metabolismo , Anticorpos de Domínio Único/administração & dosagem , Animais , Proteínas Reguladoras de Apoptose/administração & dosagem , Proteínas Reguladoras de Apoptose/farmacologia , Feminino , Células HEK293 , Humanos , Macrófagos/metabolismo , Receptor de Manose , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/administração & dosagem , Proteínas Mitocondriais/farmacologia , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos de Domínio Único/farmacologia , Microambiente Tumoral/efeitos dos fármacos
6.
Eur Rev Med Pharmacol Sci ; 22(21): 7156-7163, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30468456

RESUMO

OBJECTIVE: To explore whether MOTS-c could improve osteoporosis by promoting osteogenic differentiation of rat bone mesenchymal stem cells (BMSCs) via transforming growth factor-ß (TGF-ß)/Smad pathway. MATERIALS AND METHODS: Rat BMSCs were isolated and cultured, followed by osteogenic and lipid differentiation. CCK-8 (cell counting kit-8) assay was performed to detect the highest treatment dose of MOTS-c that did not affect cell proliferation. Expressions of osteogenesis-related genes (ALP, Bglap, and Runx2) were detected by qRT-PCR (quantitative Real-Time Polymerase Chain Reaction) and Western blot, respectively. Alizarin red staining and alkaline phosphatase (ALP) cytochemical staining were carried out to evaluate the effect of MOTS-c on BMSCs osteogenesis. TGF-ß/Smad pathway-related genes (TGF-ß1, TGF-ß2, and Smad7) in BMSCs treated with MOTS-c were detected. Finally, TGF-ß1 was knocked down to investigate the regulatory effect of MOTS-c on BMSCs osteogenesis. RESULTS: BMSCs exhibited an elongated morphology and was identified with a high purity by flow cytometry. After osteogenic differentiation, alizarin red staining and ALP staining were all positive. MOTS-c treatment could remarkably stimulate the formation of calcified nodules in BMSCs. Besides, TGF-ß/Smad pathway-related genes were significantly upregulated after BMSCs were treated with MOTS-c. Promoted osteogenesis by MOTS-c treatment was reversed by the TGF-ß1 knockdown. CONCLUSIONS: MOTS-c promotes cell differentiation of BMSCs to osteoblasts via TGF-ß/Smad pathway.


Assuntos
Células-Tronco Mesenquimais/citologia , Proteínas Mitocondriais/administração & dosagem , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Animais , Células da Medula Óssea , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Osteoblastos/citologia , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/genética
7.
Zhongguo Fei Ai Za Zhi ; 19(5): 241-6, 2016 May 20.
Artigo em Chinês | MEDLINE | ID: mdl-27215450

RESUMO

BACKGROUND: The curative effect of radiotherapy may be limited by the radioresistance of tumor. Mimetic compounds of Second mitochondria-derived activator of caspase (Smac) were hopeful to become new drugs of radiosensitization for tumor because they can increase radiation induced apoptosis in tumor cells. The aim of present study is to observe the radiosensitization effect of a new Smac mimetic ANTP-SmacN7 fusion peptide in H460 cell line. METHODS: In order to observe if the fusion peptide can enter into tumor cell, ANTP-SmacN7 fusion peptide was synthesized and linked by FITC. H460 cell was divided into control, radiation only, ANTP-SmacN7 only and ANTP-SmacN7 combined with radiation group. The cells were exposed by 0, 2, 4 and 6 Gy and the concentration of ANTP-SmacN7 was 20 µmol/L. Proliferation of H460 tumor cell was detected by WST-1 assay. There are four groups in the present study: control group, radiation group, ANTP-SmacN7 group and ANTP-SmacN7 combined with radiation group. Apoptosis was detected by flow cytometry at 24 and 48 hours after the treatment of all the groups. The level of caspase3 and cleaved caspase3 were detected by Western blot assay. RESULTS: ANTP-SmacN7 can enter into cells and promote the radiosensitization of H460 cell obviously (F=25.1, P<0.01, sensitivity enhancement ratio was 1.86). The treatment of ANTP-SmacN7 combined with radiation decreased the cloning forming efficiency (χ2=45.2, P<0.01; χ2=40.3, P<0.01), activated caspase3 by promoting the expression of cleaved caspase3 and increased the apoptosis of H460 cell line. CONCLUSIONS: ANTP-SmacN7 fusion peptide had remarkably radiosensitization effect on H460 cell line. ANTP-SmacN7 fusion peptide might be hopeful to be applied in radiosensitization therapy as a new Smac mimetic polypeptide in the future.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Neoplasias Pulmonares/radioterapia , Proteínas Mitocondriais/administração & dosagem , Peptídeos/administração & dosagem , Radiossensibilizantes/administração & dosagem , Apoptose/efeitos da radiação , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/fisiopatologia , Proteínas Mitocondriais/metabolismo , Peptídeos/metabolismo , Radiossensibilizantes/metabolismo
8.
ACS Appl Mater Interfaces ; 8(14): 8918-27, 2016 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-26890991

RESUMO

Tumor stromal cells have been increasingly recognized to interact with tumor parenchyma cells and promote tumor growth. Therefore, we speculated that therapeutics delivery to both parenchyma cells and stromal cells simultaneously might treat a tumor more effectively. Tissue factor (TF) was shown to be extensively located in a tumor and was abundantly sited in both tumor parenchyma cells and stromal cells including neo-vascular cells, tumor-associated fibroblasts, and tumor-associated macrophages, indicating it might function as a favorable target for drug delivery to multiple cell types simultaneously. EGFP-EGF1 is a fusion protein derived from factor VII, the natural ligand of TF. It retains the specific TF binding capability but does not cause coagulation. In the present study, a nanoparticle modified with EGFP-EGF1 (ENP) was constructed as a multitargeting drug delivery system. The protein binding experiment showed EGFP-EGF1 could bind well to A549 tumor cells and other stromal cells including neo-vascular cells, tumor-associated fibroblasts, and tumor-associated macrophages. Compared with unmodified nanoparticles (NP), ENP uptake by A549 cells and those stromal cells was significantly enhanced but inhibited by excessive free EGFP-EGF1. In addition, ENP induced more A549 tumor cell apoptosis than Taxol and NP when paclitaxel (PTX) was loaded. In vivo, ENP accumulated more specially in TF-overexpressed A549 tumors by in vivo imaging, mainly regions unoccupied by factor VII and targeted tumor parenchyma cells as well as different types of stromal cells by immunofluorescence staining. Treatment with PTX-loaded ENP (ENP-PTX) significantly reduced the A549 tumor growth in nude mice while NP-PTX- and Taxol-treated mice had lower response to the therapy. Furthermore, H&E and TUNEL staining revealed that ENP-PTX induced more severe tumor necrosis and more extensive cell apoptosis. Altogether, the present study demonstrated that ENP could target multiple key cell types in tumors through TF, which could be utilized to improve the therapeutic effect of anticancer drugs.


Assuntos
Sistemas de Liberação de Medicamentos , Proteínas de Fluorescência Verde/genética , Proteínas Mitocondriais/genética , Nanopartículas/administração & dosagem , Neoplasias/tratamento farmacológico , Fator G para Elongação de Peptídeos/genética , Células A549 , Animais , Apoptose/efeitos dos fármacos , Fibroblastos Associados a Câncer/efeitos dos fármacos , Proteínas de Fluorescência Verde/administração & dosagem , Proteínas de Fluorescência Verde/química , Humanos , Macrófagos/efeitos dos fármacos , Camundongos , Proteínas Mitocondriais/administração & dosagem , Proteínas Mitocondriais/química , Nanopartículas/química , Neoplasias/patologia , Proteínas de Fusão Oncogênica/administração & dosagem , Proteínas de Fusão Oncogênica/química , Paclitaxel/administração & dosagem , Paclitaxel/química , Fator G para Elongação de Peptídeos/administração & dosagem , Fator G para Elongação de Peptídeos/química , Ligação Proteica , Células Estromais/efeitos dos fármacos , Células Estromais/patologia
9.
Chembiochem ; 17(7): 570-5, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26895508

RESUMO

Cell surface p32, the target of LyP-1 homing peptide, is upregulated in tumors and atherosclerotic plaques and has been widely used as a receptor for systemic delivery of payloads. Here, we identified an improved LyP-1-mimicking peptide (TT1, CKRGARSTC). We used this peptide in a fluorescence polarization-based high-throughput screening of a 50,000-compound chemical library and identified a panel of compounds that bind p32 with low micromolar affinity. Among the hits identified in the screen, two compounds were shown to specifically bind to p32 in multiple assays. One of these compounds was chosen for an in vivo study. Nanoparticles surface-functionalized with this compound specifically adhered to surfaces coated with recombinant p32 and, when injected intravenously, homed to p32-expressing breast tumors in mice. This compound provides a lead for the development of p32-targeted affinity ligands that circumvent some of the limitations of peptide-based probes in guided drug delivery.


Assuntos
Aminopiridinas/química , Neoplasias da Mama/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Etilenodiaminas/química , Proteínas Mitocondriais/administração & dosagem , Peptídeos Cíclicos/administração & dosagem , Aminopiridinas/farmacologia , Animais , Antineoplásicos/administração & dosagem , Proteínas de Transporte , Linhagem Celular Tumoral , Etilenodiaminas/farmacologia , Feminino , Humanos , Ligantes , Camundongos , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Nanopartículas/química
10.
Biochem J ; 473(1): 99-107, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26508734

RESUMO

Cancer cells are capable of sphere formation (transformation) through reactive oxygen species (ROS) and glycolysis shift. Transformation is linked to tumorigenesis and therapy resistance, hence targeting regulators of ROS and glycolysis is important for cancer therapeutic candidates. Here, we demonstrate that Smac mimetic AZ58 in combination with tumour necrosis factor-α (TNF-α) was able to inhibit the production of ROS, inhibit glycolysis through Pim-1 kinase-mediated Ser-112 phosphorylation of BAD, and increase depolarization of mitochondria. We also identified mitochondrial isoforms of Pim-1 kinase that were targeted for degradation by AZ58 in combination with TNF-α or AZ58 in combination with Fas ligand (FasL) plus cycloheximide (CHX) through caspase-3 to block transformation. Our study demonstrates that Smac mimetic in combination with TNF-α is an ideal candidate to target Pim-1 expression, inhibit ROS production and to block transformation from blebbishields.


Assuntos
Materiais Biomiméticos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transformação Genética/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Reguladoras de Apoptose , Materiais Biomiméticos/administração & dosagem , Linhagem Celular Tumoral , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Proteínas Mitocondriais/administração & dosagem , Isoformas de Proteínas/metabolismo , Transformação Genética/efeitos dos fármacos , Fator de Necrose Tumoral alfa/administração & dosagem
11.
Invest Ophthalmol Vis Sci ; 56(2): 1129-40, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25613946

RESUMO

PURPOSE: To address the permanent disability induced by mitochondrial dysfunction in experimental autoimmune encephalomyelitis (EAE). METHODS: Mice sensitized for EAE were rescued by intravitreal injection of adeno-associated viral vector serotype 2 with the complex I subunit gene scAAV-NDUFA6Flag. Controls were injected with a mitochondrially targeted red fluorescent protein (scAAV-COX8-cherry). Another group received scAAV-COX8-cherry, but was not sensitized for EAE. Serial pattern electroretinograms (PERGs) and optical coherent tomography (OCT) evaluated visual function and structure of the retina at 1, 3, and 6 months post injection (MPI). Treated mice were killed 6 MPI for histopathology. Immunodetection of cleaved caspase 3 gauged apoptosis. Complex I activity was assessed spectrophotometrically. Expression of NDUFA6Flag in the retina and optic nerve were evaluated between 1 week to 1 month post injection by RT-PCR, immunofluorescence and immunoblotting. RESULTS: Reverse transcription-PCR and immunoblotting confirmed NDUFA6Flag overexpression with immunoprecipitation and blue native PAGE showing integration into murine complex I. Overexpression of NDUFA6Flag in the visual system of EAE mice rescued retinal complex I activity completely, axonal loss by 73%, and retinal ganglion cell (RGC) loss by 88%, RGC apoptosis by 66%, and restored the 33% loss of complex I activity in EAE to normal levels; thereby, preventing loss of vision indicated by the 43% reduction in the PERG amplitudes of EAE mice. CONCLUSIONS: NDUFA6 gene therapy provided long-term suppression of neurodegeneration in the EAE animal model suggesting that it may also ameliorate the mitochondrial dysfunction associated with permanent disability in optic neuritis and MS patients.


Assuntos
Encefalomielite Autoimune Experimental/terapia , Terapia Genética/métodos , Proteínas Mitocondriais/administração & dosagem , Animais , Apoptose , DNA/genética , Eletrorretinografia , Encefalomielite Autoimune Experimental/diagnóstico , Encefalomielite Autoimune Experimental/genética , Feminino , Regulação da Expressão Gênica , Immunoblotting , Injeções Intravítreas , Camundongos , Camundongos Endogâmicos DBA , Microscopia Eletrônica de Transmissão , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/genética , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrofotometria , Tomografia de Coerência Óptica , Resultado do Tratamento
12.
Am J Respir Crit Care Med ; 187(8): 865-78, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23449689

RESUMO

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal, female-predominant, vascular disease. Pathologic changes in PA smooth muscle cells (PASMC) include excessive proliferation, apoptosis-resistance, and mitochondrial fragmentation. Activation of dynamin-related protein increases mitotic fission and promotes this proliferation-apoptosis imbalance. The contribution of decreased fusion and reduced mitofusin-2 (MFN2) expression to PAH is unknown. OBJECTIVES: We hypothesize that decreased MFN2 expression promotes mitochondrial fragmentation, increases proliferation, and impairs apoptosis. The role of MFN2's transcriptional coactivator, peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α), was assessed. MFN2 therapy was tested in PAH PASMC and in models of PAH. METHODS: Fusion and fission mediators were measured in lungs and PASMC from patients with PAH and female rats with monocrotaline or chronic hypoxia+Sugen-5416 (CH+SU) PAH. The effects of adenoviral mitofusin-2 (Ad-MFN2) overexpression were measured in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: In normal PASMC, siMFN2 reduced expression of MFN2 and PGC1α; conversely, siPGC1α reduced PGC1α and MFN2 expression. Both interventions caused mitochondrial fragmentation. siMFN2 increased proliferation. In rodent and human PAH PASMC, MFN2 and PGC1α were decreased and mitochondria were fragmented. Ad-MFN2 increased fusion, reduced proliferation, and increased apoptosis in human PAH and CH+SU. In CH+SU, Ad-MFN2 improved walking distance (381 ± 35 vs. 245 ± 39 m; P < 0.05); decreased pulmonary vascular resistance (0.18 ± 0.02 vs. 0.38 ± 0.14 mm Hg/ml/min; P < 0.05); and decreased PA medial thickness (14.5 ± 0.8 vs. 19 ± 1.7%; P < 0.05). Lung vascularity was increased by MFN2. CONCLUSIONS: Decreased expression of MFN2 and PGC1α contribute to mitochondrial fragmentation and a proliferation-apoptosis imbalance in human and experimental PAH. Augmenting MFN2 has therapeutic benefit in human and experimental PAH.


Assuntos
GTP Fosfo-Hidrolases/deficiência , Proteínas de Choque Térmico/deficiência , Hipertensão Pulmonar/fisiopatologia , Dinâmica Mitocondrial/fisiologia , Proteínas Mitocondriais/deficiência , Fatores de Transcrição/deficiência , Animais , Apoptose/fisiologia , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Tolerância ao Exercício/efeitos dos fármacos , Hipertensão Pulmonar Primária Familiar , Feminino , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Pulmão/citologia , Pulmão/patologia , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/deficiência , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/administração & dosagem , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/fisiologia , Atrofia Óptica Autossômica Dominante/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Ratos Sprague-Dawley
13.
PLoS One ; 7(12): e51461, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23240027

RESUMO

BACKGROUND: The members of inhibitor of apoptosis proteins (IAPs) family are key negative regulators of apoptosis. Overexpression of IAPs are found in hepatocellular carcinoma (HCC), and can contribute to chemotherapy resistance and recurrence of HCC. Small-molecule Second mitochondria-derived activator of caspases (Smac) mimetics have recently emerged as novel anticancer drugs through targeting IAPs. The specific aims of this study were to 1) examine the anticancer activity of Smac mimetics as a single agent and in combination with chemotherapy in HCC cells, and 2) investigate the mechanism of anticancer action of Smac mimetics. METHODS: Four HCC cell lines, including SMMC-7721, BEL-7402, HepG2 and Hep3B, and 12 primary HCC cells were used in this study. Smac mimetic SM-164 was used to treat HCC cells. Cell viability, cell death induction and clonal formation assays were used to evaluate the anticancer activity. Western blotting analysis and a pancaspase inhibitor were used to investigate the mechanisms. RESULTS: Although SM-164 induced complete cIAP-1 degradation, it displayed weak inhibitory effects on the viability of HCC cells. Nevertheless, SM-164 considerably potentiated Apo2 ligand or TNF-related apoptosis-inducing ligand (APO2L/TRAIL)- and Doxorubicin-mediated anticancer activity in HCC cells. Mechanistic studies demonstrated that SM-164 in combination with chemotherapeutic agents resulted in enhanced activation of caspases-9, -3 and cleavage of poly ADP-ribose polymerase (PARP), and also led to decreased AKT activation. CONCLUSIONS: Smac mimetics can enhance chemotherapeutic-mediated anticancer activity by enhancing apoptosis signaling and suppressing survival signaling in HCC cells. This study suggests Smac mimetics are potential therapeutic agents for HCC.


Assuntos
Antineoplásicos/administração & dosagem , Biomimética , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Mitocondriais , Triazóis/administração & dosagem , Desaminases APOBEC , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citidina Desaminase/metabolismo , Doxorrubicina/administração & dosagem , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular/síntese química , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Proteínas Mitocondriais/administração & dosagem , Proteínas Mitocondriais/síntese química , Proteínas Musculares/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
14.
Int J Mol Med ; 30(1): 199-203, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22469910

RESUMO

Endogenous molecules released by dying cells [i.e., damage-associated molecular patterns (DAMPs)] after trauma and severe blood loss can activate pattern recognition receptors, leading to a cascade of inflammatory responses and organ injury. Mitochondrial transcription factor A (TFAM) is a transcription factor for mitochondrial DNA. TFAM is structurally related to high mobility group box 1 (HMGB1), an important member of DAMPs. We, therefore, hypothesized that TFAM can be released into the circulation after hemorrhage to initiate inflammatory responses. In order to examine this hypothesis, male Sprague-Dawley rats were bled to and maintained at a mean arterial pressure of 40 mmHg for 90 min. They were then resuscitated with an equal volume of shed blood in the form of Ringer's lactate (i.e., low-volume resuscitation) over 60 min. TFAM levels in the serum were measured at 4 h after hemorrhage and resuscitation. Our results showed that serum levels of TFAM were more than doubled after hemorrhage and resuscitation. To further characterize TFAM's biological activity, we expressed recombinant rat TFAM with a GST-tag (GST-TFAM) in an E. coli expression system. The purity of GST-TFAM was over 99% and it was immunoreactive for specific anti-TFAM antibodies. Using RAW 264.7 cells and primary rat peritoneal macrophages, we showed that GST-TFAM dose-dependently increased TNF-α release. To determine the biological activity of GST-TFAM in vivo, GST-TFAM was intravenously injected in healthy male adult rats. Our results demonstrated that intravenous injection of GST-TFAM, not GST alone, upregulated circulating levels of pro-inflammatory cytokines, increased neutrophil infiltration to the lungs and caused organ injury in healthy animals. Thus, TFAM can act as a DAMP and may contribute to the initiation of inflammatory responses in hemorrhagic shock.


Assuntos
Proteínas de Ligação a DNA/sangue , Proteínas de Ligação a DNA/imunologia , Mediadores da Inflamação/imunologia , Proteínas Mitocondriais/sangue , Proteínas Mitocondriais/imunologia , Choque Hemorrágico/imunologia , Fatores de Transcrição/sangue , Fatores de Transcrição/imunologia , Animais , Linhagem Celular , Proteínas de Ligação a DNA/administração & dosagem , Proteínas de Ligação a DNA/genética , Interleucina-6/sangue , Macrófagos/metabolismo , Masculino , Camundongos , Proteínas Mitocondriais/administração & dosagem , Proteínas Mitocondriais/genética , Infiltração de Neutrófilos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/administração & dosagem , Choque Hemorrágico/metabolismo , Choque Hemorrágico/patologia , Fatores de Transcrição/administração & dosagem , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/sangue
15.
Mitochondrion ; 11(1): 108-18, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20727424

RESUMO

Recombinant human mitochondrial transcription factor A protein (rhTFAM) was evaluated for its acute effects on cultured cells and chronic effects in mice. Fibroblasts incubated with rhTFAM acutely increased respiration in a chloramphenicol-sensitive manner. SH-SY5Y cells showed rhTFAM concentration-dependent reduction of methylpyridinium (MPP(+))-induced oxidative stress and increases in lowered ATP levels and viability. Mice treated with weekly i.v. rhTFAM showed increased mitochondrial gene copy number, complex I protein levels and ATP production rates; oxidative damage to proteins was decreased ~50%. rhTFAM treatment improved motor recovery rate after treatment with MPTP and dose-dependently improved survival in the lipopolysaccharide model of endotoxin sepsis.


Assuntos
Trifosfato de Adenosina/biossíntese , Proteínas de Ligação a DNA/farmacologia , Endotoxemia/mortalidade , Mitocôndrias/metabolismo , Proteínas Mitocondriais/farmacologia , Atividade Motora/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Fatores de Transcrição/farmacologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Animais , Linhagem Celular Tumoral , Respiração Celular/efeitos dos fármacos , Sobrevivência Celular , DNA Mitocondrial/efeitos dos fármacos , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Proteínas de Ligação a DNA/administração & dosagem , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endotoxemia/tratamento farmacológico , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/administração & dosagem , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/administração & dosagem , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Br J Cancer ; 102(12): 1707-16, 2010 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-20461078

RESUMO

BACKGROUND: XIAP (X-linked inhibitor of apoptosis protein) is an anti-apoptotic protein exerting its activity by binding and suppressing caspases. As XIAP is overexpressed in several tumours, in which it apparently contributes to chemoresistance, and because its activity in vivo is antagonised by second mitochondria-derived activator of caspase (SMAC)/direct inhibitor of apoptosis-binding protein with low pI, small molecules mimicking SMAC (so called SMAC-mimetics) can potentially overcome tumour resistance by promoting apoptosis. METHODS: Three homodimeric compounds were synthesised tethering a monomeric SMAC-mimetic with different linkers and their affinity binding for the baculoviral inhibitor repeats domains of XIAP measured by fluorescent polarisation assay. The apoptotic activity of these molecules, alone or in combination with tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and/or Bortezomib, was tested in melanoma cell lines by MTT viability assays and western blot analysis of activated caspases. RESULTS: We show that in melanoma cell lines, which are typically resistant to chemotherapeutic agents, XIAP knock-down sensitises cells to TRAIL treatment in vitro, also favouring the accumulation of cleaved caspase-8. We also describe a new series of 4-substituted azabicyclo[5.3.0]alkane monomeric and dimeric SMAC-mimetics that target various members of the IAP family and powerfully synergise at submicromolar concentrations with TRAIL in inducing cell death. Finally, we show that the simultaneous administration of newly developed SMAC-mimetics with Bortezomib potently triggers apoptosis in a melanoma cell line resistant to the combined effect of SMAC-mimetics and TRAIL. CONCLUSION: Hence, the newly developed SMAC-mimetics effectively synergise with TRAIL and Bortezomib in inducing cell death. These findings warrant further preclinical studies in vivo to verify the anticancer effectiveness of the combination of these agents.


Assuntos
Ácidos Borônicos/farmacologia , Morte Celular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Melanoma/tratamento farmacológico , Proteínas Mitocondriais/farmacologia , Pirazinas/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Proteínas Reguladoras de Apoptose , Bortezomib , Caspase 8/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Interações Medicamentosas , Sinergismo Farmacológico , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Proteínas Mitocondriais/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem
17.
Hinyokika Kiyo ; 54(1): 53-6, 2008 Jan.
Artigo em Japonês | MEDLINE | ID: mdl-18260362

RESUMO

Renal cell carcinoma (RCC) accounts for approximately 2% of all cancer cases worldwide. Metastatic disease is often present at the time of diagnosis of RCC and its poor response to chemotherapy and radiotherapy causes poor prognosis. Immunotherapy is relatively effective for RCC, but the response rate is approximately 15-20%. Therefore, new therapeutic approaches are necessary for these patients with metastatic RCC. Recently, the mechanisms responsible for the growth of RCC have been clarified, and molecular targeted therapy has been developed. In this paper, we review the new molecular targeted therapeutic agents effective for RCC.


Assuntos
Antineoplásicos/administração & dosagem , Carcinoma de Células Renais/tratamento farmacológico , Neoplasias Renais/tratamento farmacológico , Proteínas Reguladoras de Apoptose , Benzenossulfonatos/administração & dosagem , Sistemas de Liberação de Medicamentos , Humanos , Indóis/administração & dosagem , Interferon beta/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Lipossomos , Proteínas Mitocondriais/administração & dosagem , Niacinamida/análogos & derivados , Compostos de Fenilureia , Piridinas/administração & dosagem , Pirróis/administração & dosagem , Sirolimo/administração & dosagem , Sirolimo/análogos & derivados , Sorafenibe , Sunitinibe
18.
Exp Cell Res ; 312(8): 1277-88, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16466653

RESUMO

The protein transduction domain (PTD) of HIV-1 TAT has been extensively documented with regard to its membrane transduction potential, as well as its efficient delivery of biomolecules in vivo. However, the majority of PTD and PTD-conjugated molecules translocate to the nucleus rather than to the cytoplasm after transduction, due to the functional nuclear localization sequence (NLS). Here, we report a cytoplasmic transduction peptide (CTP), which was deliberately designed to ensure the efficient cytoplasmic delivery of the CTP-fused biomolecules. In comparison with PTD, CTP and its fusion partners exhibited a clear preference for cytoplasmic localization, and also markedly enhanced membrane transduction potential. Unlike the mechanism underlying PTD-mediated transduction, CTP-mediated transduction occurs independently of the lipid raft-dependent macropinocytosis pathway. The CTP-conjugated Smac/DIABLO peptide (Smac-CTP) was also shown to be much more efficient than Smac-PTD in the blockage of the antiapoptotic properties of XIAP, suggesting that cytoplasmic functional molecules can be more efficiently targeted by CTP-mediated delivery. In in vivo trafficking studies, CTP-fused beta-gal exhibited unique organ tropisms to the liver and lymph nodes when systemically injected into mice, whereas PTD-beta-gal exhibited no such tropisms. Taken together, our findings implicate CTP as a novel delivery peptide appropriate for (i) molecular targeting to cytoplasmic compartments in vitro, (ii) the development of class I-associated CTL vaccines, and (iii) special drug delivery in vivo, without causing any untoward effects on nuclear genetic material.


Assuntos
Citoplasma/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Produtos do Gene tat/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Proteínas Recombinantes de Fusão/administração & dosagem , Animais , Proteínas Reguladoras de Apoptose , Compartimento Celular/efeitos dos fármacos , Compartimento Celular/fisiologia , Citoplasma/efeitos dos fármacos , Desenho de Fármacos , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Jurkat , Fígado/efeitos dos fármacos , Fígado/metabolismo , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Mitocondriais/administração & dosagem , Proteínas Mitocondriais/metabolismo , Biologia Molecular/métodos , Peptídeos/química , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/efeitos dos fármacos , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
19.
Cancer Res ; 63(4): 831-7, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12591734

RESUMO

The inhibitor of apoptosis proteins (IAPs) plays a central role in repressing caspase-mediated cell death. However, little is known about the actual role of endogenously expressed IAPs in cancer cells. We found that the cytochrome c/apoptotic protease-activating factor-1 (apoptosome)-dependent caspase activation is deficient in human non-small cell lung cancer (NSCLC) NCI-H460 cells. This dysfunctional apoptosome activity was not correlated with any decrease of apoptosome component factors, but it was linked to an increased X-linked inhibitor of apoptosis protein (XIAP). In H460 cells, the overexpressed XIAP, but not c-IAP1, bound to the processed form of caspase-9 and suppressed the activation of downstream effector caspases. Moreover, the defect in apoptosome activity in H460 cells was dramatically restored by the IAP-targeting SmacN7 peptide, which disrupted XIAP-caspase-9 binding, indicating an essential role of the IAP in the apoptosome inhibition. However, the SmacN7 did not show any striking effect on the apoptosome activity of normal lung fibroblast cells, although these cells also expressed modest amounts of IAP. To explore the therapeutic approach, we additionally developed SmacN7(R)8, a newly designed cell permeable peptide. The SmacN7(R)8 selectively reversed the apoptosis resistance of H460 cells, and when in combination with chemotherapy, regressed the tumor growth in vivo with little toxicity to the mice. Our results indicate that IAP-dependent suppression of apoptosome predominantly occurs in IAP-overexpressing tumor, and the IAP-targeting Smac peptide is an effective molecule to increase tumor cell death induced by chemotherapy in vitro and in vivo.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proteínas de Transporte/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Mitocondriais/farmacologia , Peptídeos/farmacologia , Proteínas/antagonistas & inibidores , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Fator Apoptótico 1 Ativador de Proteases , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteínas de Transporte/administração & dosagem , Proteínas de Transporte/síntese química , Caspase 9 , Caspases/metabolismo , Cisplatino/administração & dosagem , Grupo dos Citocromos c/metabolismo , Sinergismo Farmacológico , Ativação Enzimática , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Mitocondriais/administração & dosagem , Proteínas Mitocondriais/síntese química , Peptídeos/administração & dosagem , Peptídeos/síntese química , Biossíntese de Proteínas , Proteínas/genética , Proteínas/metabolismo , Células Tumorais Cultivadas , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Cancer Res ; 62(1): 18-23, 2002 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11782351

RESUMO

Release of cytochrome c from mitochondria to cytosol has been identified as one of the central events of apoptosis. Direct injection of cytochrome c induces apoptosis in some but not in all cell types. We observed that LNCaP prostate cancer cells failed to undergo apoptosis induced by cytochrome c microinjections. Microinjection of cytochrome c with another mitochondrial protein, Smac, was sufficient to activate caspases, however. Smac is believed to function as a neutralizer of caspase inhibitors, and mass spectrometry analysis identified XIAP as a predominant Smac binding protein in LNCaP cells. These findings are consistent with a requirement for a release of Smac from mitochondria to enable caspase activation in prostate cells. Indeed, translocation of Smac from mitochondria to cytosol was observed in LNCaP cells that undergo apoptosis and was inhibited by epidermal growth factor, which is a survival factor for these cells. These results further emphasize the central role of mitochondria in the regulation of apoptosis in prostate cancer cells.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Transporte/administração & dosagem , Grupo dos Citocromos c/administração & dosagem , Proteínas Mitocondriais/administração & dosagem , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases , Sequência de Aminoácidos , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Cromonas/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Cavalos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Masculino , Espectrometria de Massas , Microinjeções , Proteínas Mitocondriais/metabolismo , Dados de Sequência Molecular , Morfolinas/farmacologia , Testes de Precipitina , Neoplasias da Próstata/tratamento farmacológico , Ligação Proteica , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA