Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mol Cell ; 47(6): 839-50, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22885005

RESUMO

Both mitochondria, which are metabolic powerhouses, and telomeres, which help maintain genomic stability, have been implicated in cancer and aging. However, the signaling events that connect these two cellular structures remain poorly understood. Here, we report that the canonical telomeric protein TIN2 is also a regulator of metabolism. TIN2 is recruited to telomeres and associates with multiple telomere regulators including TPP1. TPP1 interacts with TIN2 N terminus, which contains overlapping mitochondrial and telomeric targeting sequences, and controls TIN2 localization. We have found that TIN2 is posttranslationally processed in mitochondria and regulates mitochondrial oxidative phosphorylation. Reducing TIN2 expression by RNAi knockdown inhibited glycolysis and reactive oxygen species (ROS) production and enhanced ATP levels and oxygen consumption in cancer cells. These results suggest a link between telomeric proteins and metabolic control, providing an additional mechanism by which telomeric proteins regulate cancer and aging.


Assuntos
Mitocôndrias/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Telômero/metabolismo , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Glicólise/genética , Humanos , Fosforilação Oxidativa , Consumo de Oxigênio , Ligação Proteica , Processamento de Proteína Pós-Traducional , Sinais Direcionadores de Proteínas , Interferência de RNA , RNA Citoplasmático Pequeno , Espécies Reativas de Oxigênio/metabolismo , Complexo Shelterina , Proteínas de Ligação a Telômeros/química , Proteínas de Ligação a Telômeros/genética
2.
Proc Natl Acad Sci U S A ; 112(12): 3764-9, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25775519

RESUMO

The dynamic mutability of microsatellite repeats is implicated in the modification of gene function and disease phenotype. Studies of the enhanced instability of long trinucleotide repeats (TNRs)-the cause of multiple human diseases-have revealed a remarkable complexity of mutagenic mechanisms. Here, we show that cold, heat, hypoxic, and oxidative stresses induce mutagenesis of a long CAG repeat tract in human cells. We show that stress-response factors mediate the stress-induced mutagenesis (SIM) of CAG repeats. We show further that SIM of CAG repeats does not involve mismatch repair, nucleotide excision repair, or transcription, processes that are known to promote TNR mutagenesis in other pathways of instability. Instead, we find that these stresses stimulate DNA rereplication, increasing the proportion of cells with >4 C-value (C) DNA content. Knockdown of the replication origin-licensing factor CDT1 eliminates both stress-induced rereplication and CAG repeat mutagenesis. In addition, direct induction of rereplication in the absence of stress also increases the proportion of cells with >4C DNA content and promotes repeat mutagenesis. Thus, environmental stress triggers a unique pathway for TNR mutagenesis that likely is mediated by DNA rereplication. This pathway may impact normal cells as they encounter stresses in their environment or during development or abnormal cells as they evolve metastatic potential.


Assuntos
Mutagênese , Estresse Fisiológico , Repetições de Trinucleotídeos , Temperatura Baixa , DNA/química , DNA/genética , Reparo do DNA , Replicação do DNA , Redes Reguladoras de Genes , Instabilidade Genômica , Proteínas de Fluorescência Verde/química , Células HEK293 , Temperatura Alta , Humanos , Repetições de Microssatélites , Estresse Oxidativo , Fenótipo , Expansão das Repetições de Trinucleotídeos
3.
J Biol Chem ; 287(6): 4014-22, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22074915

RESUMO

Fibroblast-like synoviocytes (FLS) play important roles in the pathogenesis of rheumatoid arthritis (RA). Potassium channels have regulatory roles in many cell functions. We have identified the calcium- and voltage-gated KCa1.1 channel (BK, Maxi-K, Slo1, KCNMA1) as the major potassium channel expressed at the plasma membrane of FLS isolated from patients with RA (RA-FLS). We further show that blocking this channel perturbs the calcium homeostasis of the cells and inhibits the proliferation, production of VEGF, IL-8, and pro-MMP-2, and migration and invasion of RA-FLS. Our findings indicate a regulatory role of KCa1.1 channels in RA-FLS function and suggest this channel as a potential target for the treatment of RA.


Assuntos
Membrana Celular/metabolismo , Regulação da Expressão Gênica , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/biossíntese , Febre Reumática/metabolismo , Líquido Sinovial/metabolismo , Membrana Sinovial/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Cálcio/metabolismo , Membrana Celular/patologia , Proliferação de Células , Precursores Enzimáticos/biossíntese , Feminino , Gelatinases/biossíntese , Células HEK293 , Homeostase , Humanos , Interleucina-8/biossíntese , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/antagonistas & inibidores , Masculino , Pessoa de Meia-Idade , Febre Reumática/patologia , Membrana Sinovial/patologia , Fator A de Crescimento do Endotélio Vascular/biossíntese
4.
Mol Ther ; 16(3): 599-606, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18227840

RESUMO

Hypoxia occurs in many tumors and reduces the effectiveness of radio- and chemotherapy. Hypoxia also impedes immune responses to tumors, reducing T lymphocyte production of cytokines such as interleukin-2 (IL-2) and interferon gamma, as well as the survival and proliferation of these cells. We constructed a lentiviral vector encoding a bidirectional hypoxia-inducible responsive element (HRE) derived from human vascular endothelial growth factor, which drives the hIL-2 gene and a marker gene. We used a model of human B cell lymphoma to show that tumor-specific T cells modified with this vector upregulate hIL-2 expression when oxygen tension is low in vitro and in vivo. The consequence of this effect is to increase T-cell survival and proliferation whilst sustaining effector function, even in O(2) concentrations as low as 1%. The phenotype of the transduced cells is unchanged, as is their ability to migrate to tumor. HRE-IL-2-modified cytotoxic T lymphocytes (CTLs) produce faster and more complete tumor regression than parental CTLs and increase overall survival. Hypoxia-resistant T cells may thus be of value in the treatment of human tumors in which areas of hypoxia may otherwise account for resistance to this therapeutic strategy.


Assuntos
Engenharia Genética/métodos , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/fisiologia , Apoptose/genética , Apoptose/fisiologia , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Quimiotaxia/genética , Quimiotaxia/fisiologia , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Humanos , Imuno-Histoquímica , Interleucina-2/genética , Interleucina-2/metabolismo , Lentivirus/genética , Luciferases/genética , Luciferases/metabolismo , Neoplasias/genética , Neoplasias/patologia , Neoplasias/fisiopatologia , Fenótipo , Elementos de Resposta/genética , Linfócitos T Citotóxicos/citologia , Transdução Genética , Transgenes/genética , Fator A de Crescimento do Endotélio Vascular/genética
6.
Clin Immunol ; 128(3): 329-39, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18524682

RESUMO

Hepatitis C virus (HCV)-specific cytotoxic T lymphocytes (CTLs) play an important role in HCV clearance. The frequency of HCV-specific T(CD8) in peripheral blood of HCV-infected donors is very low and HCV cannot be cultivated for reinfection of antigen presenting cells, making it difficult to detect T(CD8) of broad HCV specificities from peripheral blood mononuclear cells (PBMCs). We have developed a recombinant adenoviral system that efficiently reactivates and expands HCV-specific CTLs from PBMCs of HCV-infected donors. Replication-incompetent adenoviruses expressing individual HCV proteins (core and NS3) were produced and PBMCs from HCV-infected donors were transduced with these recombinant adeno-HCV constructs to stimulate HCV-specific CTL populations. T cells expanded from adeno-HCV stimulated cultures were potent producers of HCV-specific IFN-gamma and TNF-alpha and efficiently lysed target cells pulsed with HCV peptides. These constructs could stimulate T(CD8) directed towards multiple HCV peptides while preserving the determinant hierarchy. This approach therefore overcomes some of the shortcomings of the selective expansion of CTLs with peptide-based vaccine strategies. These findings provide an effective approach for the expansion of HCV-specific CTLs from PBMCs of HCV-infected patients and have potential for immunotherapeutic/vaccine development.


Assuntos
Adenoviridae/genética , Citocinas/imunologia , Hepacivirus/imunologia , Antígenos da Hepatite C/imunologia , Hepatite C Crônica/imunologia , Linfócitos T Citotóxicos/imunologia , Adulto , Proteínas do Capsídeo/genética , Células Cultivadas , Citocinas/análise , Citotoxicidade Imunológica , Vetores Genéticos , Hepatite C Crônica/virologia , Humanos , Ativação Linfocitária , Pessoa de Meia-Idade , Transdução Genética , Vacinas contra Hepatite Viral/imunologia
7.
Cancer Res ; 64(4): 1483-9, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14973049

RESUMO

The EBV-encoded latent membrane proteins (LMP1 and LMP2), which are expressed in various EBV-associated malignancies have been proposed as a potential target for CTL-based therapy. However, the precursor frequency for LMP-specific CTL is generally low, and immunotherapy based on these antigens is often compromised by the poor immunogenicity and potential threat from their oncogenic potential. Here we have developed a replication- incompetent adenoviral vaccine that encodes multiple HLA class I-restricted CTL epitopes from LMP1 and LMP2 as a polyepitope. Immunization with this polyepitope vaccine consistently generated strong LMP-specific CTL responses in HLA A2/K(b) mice, which can be readily detected by both ex vivo and in vivo T-cell assays. Furthermore, a human CTL response to LMP antigens can be rapidly expanded after stimulation with this recombinant polyepitope vector. These expanded T cells displayed strong lysis of autologous target cells sensitized with LMP1 and/or LMP2 CTL epitopes. More importantly, this adenoviral vaccine was also successfully used to reverse the outgrowth of LMP1-expressing tumors in HLA A2/K(b) mice. These studies demonstrate that a replication-incompetent adenovirus polyepitope vaccine is an excellent tool for the induction of a protective CTL response directed toward multiple LMP CTL epitopes restricted through common HLA class I alleles prevalent in different ethnic groups where EBV-associated malignancies are endemic.


Assuntos
Doença de Hodgkin/terapia , Neoplasias Nasofaríngeas/terapia , Linfócitos T Citotóxicos/imunologia , Vacinas Sintéticas/uso terapêutico , Proteínas da Matriz Viral/imunologia , Vacinas Virais/uso terapêutico , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Epitopos de Linfócito T , Antígenos H-2/imunologia , Humanos , Imunização , Camundongos , Dados de Sequência Molecular , Proteínas da Matriz Viral/genética , Replicação Viral
8.
J Mol Biol ; 428(15): 2978-80, 2016 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-27318194

RESUMO

Multiple pathways modulate the dynamic mutability of trinucleotide repeats (TNRs), which are implicated in neurodegenerative disease and evolution. Recently, we reported that environmental stresses induce TNR mutagenesis via stress responses and rereplication, with more than 50% of mutants carrying deletions or insertions-molecular signatures of DNA double-strand break repair. We now show that knockdown of alt-nonhomologous end joining (alt-NHEJ) components-XRCC1, LIG3, and PARP1-suppresses stress-induced TNR mutagenesis, in contrast to the components of homologous recombination and NHEJ, which have no effect. Thus, alt-NHEJ, which contributes to genetic mutability in cancer cells, also plays a novel role in environmental stress-induced TNR mutagenesis.


Assuntos
Reparo do DNA por Junção de Extremidades/genética , Mutagênese/genética , Estresse Fisiológico/genética , Repetições de Trinucleotídeos/genética , Quebras de DNA de Cadeia Dupla , Replicação do DNA/genética , Recombinação Homóloga/genética , Humanos
9.
Cell Rep ; 14(9): 2154-2165, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26923594

RESUMO

Transmitochondrial cybrids and multiple OMICs approaches were used to understand mitochondrial reprogramming and mitochondria-regulated cancer pathways in triple-negative breast cancer (TNBC). Analysis of cybrids and established breast cancer (BC) cell lines showed that metastatic TNBC maintains high levels of ATP through fatty acid ß oxidation (FAO) and activates Src oncoprotein through autophosphorylation at Y419. Manipulation of FAO including the knocking down of carnitine palmitoyltransferase-1A (CPT1) and 2 (CPT2), the rate-limiting proteins of FAO, and analysis of patient-derived xenograft models confirmed the role of mitochondrial FAO in Src activation and metastasis. Analysis of TCGA and other independent BC clinical data further reaffirmed the role of mitochondrial FAO and CPT genes in Src regulation and their significance in BC metastasis.


Assuntos
Metabolismo Energético , Ácidos Graxos/metabolismo , Mitocôndrias/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Quinases da Família src/metabolismo , Animais , Carcinogênese , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos SCID , Transplante de Neoplasias , Oxirredução , Fosforilação , Processamento de Proteína Pós-Traducional , Neoplasias de Mama Triplo Negativas/patologia
10.
Oncotarget ; 6(35): 37385-97, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26460949

RESUMO

Several studies have demonstrated that the antitumor activities of both T and natural killer (NK) effector populations are limited by the immunosuppressive strategies of tumors. In several malignant transformations, the expression of HLA-G by tumor cells rises dramatically, rendering them strongly immunosuppressive. In this study, we postulated that the absence of HLA-G receptors would prevent the immunosuppressive effects of both soluble and membrane-bound HLA-G. Thus, we investigated the therapeutic potential of effector NK cells genetically modified to downregulate the expression of ILT2 (HLA-G receptor) on their cell surfaces. We have shown that the proliferation of modified NK is still dependent on stimulation signals (no malignant transformation). ILT2- NK cells proliferate, migrate, and eliminate HLA-G negative targets cells to the same extent parental NK cells do. However, in the presence of HLA-G positive tumors, ILT2- NK cells exhibit superior proliferation, conjugate formation, degranulation, and killing activities compared to parent NK cells. We tested the effectiveness of ILT2- NK cells in vivo using a xenograft cancer model and found that silencing ILT2 rescued their anti-tumor activity.We believe that combining ILT2- NK cells with existing therapeutic strategies will strengthen the antitumor response in cancer patients.


Assuntos
Antígenos CD/metabolismo , Antígenos HLA-G/metabolismo , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/transplante , Leucemia/terapia , Ativação Linfocitária , Linfócitos do Interstício Tumoral/metabolismo , Receptores Imunológicos/metabolismo , Microambiente Tumoral , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Degranulação Celular , Proliferação de Células , Quimiotaxia , Técnicas de Cocultura , Citotoxicidade Imunológica , Células HEK293 , Antígenos HLA-G/genética , Antígenos HLA-G/imunologia , Humanos , Células K562 , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Leucemia/genética , Leucemia/imunologia , Leucemia/metabolismo , Leucemia/patologia , Receptor B1 de Leucócitos Semelhante a Imunoglobulina , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Camundongos Endogâmicos NOD , Camundongos SCID , Fenótipo , Interferência de RNA , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Transdução de Sinais , Fatores de Tempo , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Hum Gene Ther ; 13(11): 1337-47, 2002 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-12162816

RESUMO

Recombinant adenoviral vectors have potential for the treatment of a variety of musculoskeletal defects and such gene therapy systems have been a recent research focus in orthopedic surgery. In studies reported here, two different adenovirus vectors have been compared for their ability to transduce human bone marrow mesenchymal stem cells (hBM-MSCs) and elicit bone formation in vivo. Vectors consisted either of standard adenovirus type 5 (Ad5) vector or a chimeric adenovirus type 5 vector that contains an adenovirus type 35 fiber (Ad5F35), which has been recently demonstrated to bestow a different cellular tropism, and a complete cDNA encoding human bone morphogenetic 2 (BMP2). Studies were also conducted to compare the transduction efficiency of these vectors using enhanced green fluorescent protein (GFP). hBM-MSCs transduced with Ad5F35 vectors had higher levels of transgene expression than those transduced with Ad5 vectors. The results also demonstrate that hBM-MSCs lack the coxsackie-adenovirus receptor (CAR), which is responsible for cellular adsorption of Ad5. Therefore, the data suggest that Ad5 virus adsorption to hBM-MSCs is inefficient. Ad5BMP2- or Ad5F35BMP2-transduced hBM-MSCs were also compared in an in vivo heterotopic bone formation assay. Mineralized bone was radiologically identified only in muscle that received the Ad5F35BMP2 transduced hBM-MSCs. In summary, Ad5F35BMP2 can efficiently transduce hBM-MSCs leading to enhanced bone formation in vivo.


Assuntos
Adenoviridae/genética , Proteínas Morfogenéticas Ósseas/genética , Vetores Genéticos , Osteogênese , Fator de Crescimento Transformador beta , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/biossíntese , Calcificação Fisiológica , Carcinoma/patologia , Linhagem Celular , Transformação Celular Viral , Células Cultivadas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , DNA Recombinante/genética , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde , Humanos , Injeções Intramusculares , Proteínas Luminescentes/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes/metabolismo , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Transdução Genética , Transgenes , Células Tumorais Cultivadas
12.
J Cereb Blood Flow Metab ; 22(8): 959-70, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12172381

RESUMO

Traumatic brain injury (TBI) causes delayed neuronal deficits that in principle could be prevented by timely intervention with therapeutic genes. However, appropriate vectors for gene transfer to the brain with TBI remain to be developed. First-generation adenoviruses (fgAd) are usually associated with inflammatory and toxic effects when inoculated into brains, despite their high efficiency of gene transfer to these tissues. In this study the authors attempted to determine whether a less immunogenic gene-transfer protocol can be established in the traumatically injured rat brain using helper-dependent adenoviruses (hdAd), a novel adenoviral construct with full deletion of viral coding sequences. Their results show that transgene expression from intrahippocampally inoculated hdAd is maintained for at least 2 months after TBI, in contrast to the much shorter duration of fgAd-mediated gene expression. There was only minimal secretion of proinflammatory IL-1beta and TNF-alpha after inoculation of hdAd. Furthermore, the hdAd-mediated gene expression was associated with less microglial proliferation, astrocytic activation, and macrophage infiltration than observed in fgAd-inoculated brains. There was no additional tissue loss after hdAd inoculation compared with PBS injection. Although both anti-adenoviral and neutralizing antibodies were found in serum after brain inoculation of hdAd, they did not appear to affect transgene expression. The results suggest that hdAd are less immunogenic vectors than conventional adenoviral vectors, and offer improved vehicles for long-term therapeutic transgene transfer to traumatically injured brains.


Assuntos
Adenoviridae/genética , Lesões Encefálicas/fisiopatologia , Técnicas de Transferência de Genes , Vetores Genéticos , Hipocampo/fisiopatologia , Inflamação , Adenoviridae/imunologia , Animais , Anticorpos Antivirais/sangue , Lesões Encefálicas/genética , Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Vírus Auxiliares , Hipocampo/imunologia , Hipocampo/patologia , Interleucina-1/metabolismo , Macrófagos/fisiologia , Masculino , Neuroglia/fisiologia , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Transgenes , Fator de Necrose Tumoral alfa/metabolismo , beta-Galactosidase/metabolismo
13.
PLoS One ; 8(10): e76740, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24146918

RESUMO

Natural killer (NK) cells are large granular lymphocytes that participate in both innate and adaptive immune responses against tumors and pathogens. They are also involved in other conditions, including organ rejection, graft-versus-host disease, recurrent spontaneous abortions, and autoimmune diseases such as multiple sclerosis. We demonstrate that human NK cells express the potassium channels Kv1.3 and KCa3.1. Expression of these channels does not vary with expression levels of maturation markers but varies between adherent and non-adherent NK cell subpopulations. Upon activation by mitogens or tumor cells, adherent NK (A-NK) cells preferentially up-regulate KCa3.1 and non-adherent (NA-NK) cells preferentially up-regulate Kv1.3. Consistent with this different phenotype, A-NK and NA-NK do not display the same sensitivity to the selective KCa3.1 blockers TRAM-34 and NS6180 and to the selective Kv1.3 blockers ShK-186 and PAP-1 in functional assays. Kv1.3 block inhibits the proliferation and degranulation of NA-NK cells with minimal effects on A-NK cells. In contrast, blocking KCa3.1 increases the degranulation and cytotoxicity of A-NK cells, but not of NA-NK cells. TRAM-34, however, does not affect their ability to form conjugates with target tumor cells, to migrate, or to express chemokine receptors. TRAM-34 and NS6180 also increase the proliferation of both A-NK and NA-NK cells. This results in a TRAM-34-induced increased ability of A-NK cells to reduce in vivo tumor growth. Taken together, our results suggest that targeting KCa3.1 on NK cells with selective blockers may be beneficial in cancer immunotherapy.


Assuntos
Citotoxicidade Imunológica , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Células Matadoras Naturais/imunologia , Animais , Receptor 1 de Quimiocina CX3C , Adesão Celular/efeitos dos fármacos , Degranulação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Interleucina-12/farmacologia , Interleucina-15/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Células K562 , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/fisiologia , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/metabolismo , Camundongos , Proteínas Associadas a Pancreatite , Fenótipo , Bloqueadores dos Canais de Potássio/farmacologia , Pirazóis/metabolismo , Receptores de Quimiocinas/metabolismo , Proteínas Recombinantes/farmacologia , Regulação para Cima/efeitos dos fármacos
14.
PLoS One ; 8(5): e61747, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23671572

RESUMO

Mitochondrial-nucleus cross talks and mitochondrial retrograde regulation can play a significant role in cellular properties. Transmitochondrial cybrid systems (cybrids) are an excellent tool to study specific effects of altered mitochondria under a defined nuclear background. The majority of the studies using the cybrid model focused on the significance of specific mitochondrial DNA variations in mitochondrial function or tumor properties. However, most of these variants are benign polymorphisms without known functional significance. From an objective of rectifying mitochondrial defects in cancer cells and to establish mitochondria as a potential anticancer drug target, understanding the role of functional mitochondria in reversing oncogenic properties under a cancer nuclear background is very important. Here we analyzed the potential reversal of oncogenic properties of a highly metastatic cell line with the introduction of non-cancerous mitochondria. Cybrids were established by fusing the mitochondria DNA depleted 143B TK- ρ0 cells from an aggressive osteosarcoma cell line with mitochondria from benign breast epithelial cell line MCF10A, moderately metastatic breast cancer cell line MDA-MB-468 and 143B cells. In spite of the uniform cancerous nuclear background, as observed with the mitochondria donor cells, cybrids with benign mitochondria showed high mitochondrial functional properties including increased ATP synthesis, oxygen consumption and respiratory chain activities compared to cybrids with cancerous mitochondria. Interestingly, benign mitochondria could reverse different oncogenic characteristics of 143B TK(-) cell including cell proliferation, viability under hypoxic condition, anti-apoptotic properties, resistance to anti-cancer drug, invasion, and colony formation in soft agar, and in vivo tumor growth in nude mice. Microarray analysis suggested that several oncogenic pathways observed in cybrids with cancer mitochondria are inhibited in cybrids with non-cancerous mitochondria. These results suggest the critical oncogenic regulation by mitochondrial-nuclear cross talk and highlights rectifying mitochondrial functional properties as a promising target in cancer therapy.


Assuntos
Núcleo Celular/genética , DNA Mitocondrial/genética , Neoplasias Mamárias Experimentais/genética , Mitocôndrias/genética , Trifosfato de Adenosina/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Hipóxia Celular , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , DNA Mitocondrial/metabolismo , Doxorrubicina/farmacologia , Feminino , Xenoenxertos , Humanos , Células Híbridas/efeitos dos fármacos , Células Híbridas/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Metástase Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética
15.
J Vis Exp ; (54)2011 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-21860378

RESUMO

Hypoxia is defined as the reduction or lack of oxygen in organs, tissues, or cells. This decrease of oxygen tension can be due to a reduced supply in oxygen (causes include insufficient blood vessel network, defective blood vessel, and anemia) or to an increased consumption of oxygen relative to the supply (caused by a sudden higher cell proliferation rate). Hypoxia can be physiologic or pathologic such as in solid cancers, rheumatoid arthritis, atherosclerosis etc… Each tissues and cells have a different ability to adapt to this new condition. During hypoxia, hypoxia inducible factor alpha (HIF) is stabilized and regulates various genes such as those involved in angiogenesis or transport of oxygen. The stabilization of this protein is a hallmark of hypoxia, therefore detecting HIF is routinely used to screen for hypoxia. In this article, we propose two simple methods to induce hypoxia in mammalian cell cultures and simple tests to evaluate the hypoxic status of these cells.


Assuntos
Hipóxia Celular/fisiologia , Técnicas Citológicas/métodos , Linhagem Celular , Células Cultivadas , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células K562 , Oxigênio/metabolismo
16.
J Vis Exp ; (57)2011 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-22127014

RESUMO

Reactive oxygen species include a number of molecules that damage DNA and RNA and oxidize proteins and lipids (lipid peroxydation). These reactive molecules contain an oxygen and include H(2;)O(2;) (hydrogen peroxide), NO (nitric oxide), O(2;)(-) (oxide anion), peroxynitrite (ONOO(-)), hydrochlorous acid (HOCl), and hydroxyl radical (OH(-)). Oxidative species are produced not only under pathological situations (cancers, ischemic/reperfusion, neurologic and cardiovascular pathologies, infectious diseases, inflammatory diseases, autoimmune diseases , etc…) but also during physiological (non-pathological) situations such as cellular metabolism. Indeed, ROS play important roles in many cellular signaling pathways (proliferation, cell activation, migration etc..). ROS can be detrimental (it is then referred to as "oxidative and nitrosative stress") when produced in high amounts in the intracellular compartments and cells generally respond to ROS by upregulating antioxidants such as superoxide dismutase (SOD) and catalase (CAT), glutathione peroxidase (GPx) and glutathione (GSH) that protects them by converting dangerous free radicals to harmless molecules (i.e. water). Vitamins C and E have also been described as ROS scavengers (antioxidants). Free radicals are beneficial in low amounts. Macrophage and neutrophils-mediated immune responses involve the production and release of NO, which inhibits viruses, pathogens and tumor proliferation. NO also reacts with other ROS and thus, also has a role as a detoxifier (ROS scavenger). Finally NO acts on vessels to regulate blood flow which is important for the adaptation of muscle to prolonged exercise. Several publications have also demonstrated that ROS are involved in insulin sensitivity. Numerous methods to evaluate ROS production are available. In this article we propose several simple, fast, and affordable assays; these assays have been validated by many publications and are routinely used to detect ROS or its effects in mammalian cells. While some of these assays detect multiple ROS, others detect only a single ROS.


Assuntos
Leucemia de Células T/metabolismo , Neoplasias Pulmonares/química , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Fluoresceínas/química , Corantes Fluorescentes/química , Humanos , Células Jurkat , Neoplasias Pulmonares/metabolismo
17.
Methods Mol Biol ; 651: 1-29, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20686957

RESUMO

The abnormal decrease or the lack of oxygen supply to cells and tissues is called hypoxia. This condition is commonly seen in various diseases such as rheumatoid arthritis and atherosclerosis, also in solid cancers. Pre-clinical and clinical studies have shown that hypoxic cancers are extremely aggressive, resistant to standard therapies (chemotherapy and radiotherapy), and thus very difficult to eradicate. Hypoxia affects both the tumor and the immune cells via various pathways. This review summarizes the most common effects of hypoxia on immune cells that play a key role in the anti-tumor response, the limitation of current therapies, and the potential solutions that were developed for hypoxic malignancies.


Assuntos
Leucócitos/imunologia , Neoplasias/patologia , Neoplasias/terapia , Animais , Hipóxia Celular , Humanos , Neoplasias/imunologia
18.
Blood ; 105(11): 4247-54, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15728125

RESUMO

The efficacy of adoptive T-cell therapy as treatment for malignancies may be enhanced by genetic modification of infused cells. However, oncogenic events due to vector/transgene integration, and toxicities due to the infused cells themselves, have tempered enthusiasm. A safe and efficient means of removing aberrant cells in vivo would ameliorate these concerns. We describe a "safety switch" that can be stably and efficiently expressed in human T cells without impairing phenotype, function, or antigen specificity. This reagent is based on a modified human caspase 9 fused to a human FK506 binding protein (FKBP) to allow conditional dimerization using a small molecule pharmaceutical. A single 10-nM dose of synthetic dimerizer drug induces apoptosis in 99% of transduced cells selected for high transgene expression in vitro and in vivo. This system has several advantages over currently available suicide genes. First, it consists of human gene products with low potential immunogenicity. Second, administration of dimerizer drug has no effects other than the selective elimination of transduced T cells. Third, inducible caspase 9 maintains function in T cells overexpressing antiapoptotic molecules. These characteristics favor incorporation of inducible caspase 9 as a safety feature in human T-cell therapies.


Assuntos
Caspases/genética , Indução Enzimática , Genes Transgênicos Suicidas , Terapia Genética/métodos , Imunoterapia Adotiva/métodos , Animais , Apoptose/efeitos dos fármacos , Caspase 9 , Caspases/uso terapêutico , Dimerização , Humanos , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T/metabolismo , Linfócitos T/transplante , Proteínas de Ligação a Tacrolimo/uso terapêutico
19.
J Biol Chem ; 280(18): 17807-14, 2005 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-15757909

RESUMO

By confining germline transcription as a byproduct of the mechanisms inherent to genetic rearrangements, the translation of respective mRNAs and their biological relevance might have been overlooked. Here we report the identification, cloning, and biochemical characterization of a human Vkappa-like protein that is encoded by a germline transcript. This surrogate protein assembles with the immunoglobulin mu heavy chain at the surface of B cell progenitors and precursors to form a kappa-like antigen receptor. These findings support the notion that germline transcription is not futile and stress the flexibility in eukaryotic gene usage and expression. In addition, the present study confirms the co-existence of surrogate lambda and kappa receptors that are proposed to work in concert to promote B lymphocyte maturation.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Mutação em Linhagem Germinativa/genética , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/fisiologia , Transcrição Gênica/genética , Sequência de Aminoácidos , Linhagem Celular , Humanos , Região Variável de Imunoglobulina/genética , Cadeias kappa de Imunoglobulina/genética , Células Jurkat , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Receptores de Células Precursoras de Linfócitos B , Receptores de Antígenos de Linfócitos B
20.
Blood ; 104(8): 2272-80, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15161664

RESUMO

Effective targeting of vectors to tumor cells that have metastasized to multiple different tissue sites remains a major challenge for gene therapy. Tumor-specific cytotoxic T lymphocytes (CTLs) have been shown in animal models and in humans to be able to cross tissue barriers and traffic to tumor cells. However, their capacity to eliminate malignancy has been limited by tumor immune evasion strategies. We now use a model of Epstein-Barr virus-mediated malignancy to show that human CTLs themselves may be modified to release therapeutic vectors following engagement of their antigen-specific receptors and that these vectors will effectively transduce and destroy tumor targets. We generated EBV-specific CTLs that were transgenic for the adenoviral E1 gene under the control of the cell activation-dependent CD40 ligand (CD40L) promoter. Following transduction with E1-deficient adenoviral vectors, these CTLs produced infectious virus when exposed to HLA-matched EBV-expressing targets, but not on exposure to major histocompatibility complex (MHC)-mismatched or otherwise irrelevant cells. This approach provides a means of delivering oncolytic/therapeutic vectors not only to locally accessible macroscopic tumors as is presently the case, but also to disseminated metastatic disease, while avoiding the risks associated with systemic administration of large doses of adenoviral vectors.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Neoplasias/genética , Neoplasias/terapia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Adenoviridae/fisiologia , Linhagem Celular , Expressão Gênica , Terapia Genética/instrumentação , Vetores Genéticos/fisiologia , Herpesvirus Humano 4/fisiologia , Humanos , Cinética , Neoplasias/imunologia , Neoplasias/patologia , Linfócitos T Citotóxicos/virologia , Fatores de Tempo , Transdução Genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA