Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 195: 47-57, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36566798

RESUMO

Doxorubicin (Dox), an anthracycline antibiotic, is an anticancer drug that inhibits DNA replication and cellular metabolic processes in cancer cells with high proliferative potential. However, Dox causes severe side effects, including myocardial damage and heart failure, but the molecular mechanism underlying Dox-induced myocardial injury remains uncertain. In the present study, we evaluated the effects of Dox on the mitochondrial quality control system and regulation of mitochondrial respiration and autophagy in an in vitro rat myoblast H9c2 cell culture model using western blotting, immunohistochemistry, the Seahorse XF24 system, and flow cytometry. Our results showed that Dox did not impair the initiation of autophagic flux or the functions of lysosomes; however, Dox affected the mitochondrial quality control system, leading to a fission-dominant morphology and impaired regulation of mitochondrial respiration, thereby increasing oxidative stress and inhibited progression of autophagy, particularly the fusion of autophagosomes with lysosomes. This inhibition caused a significant decrease in the formation of autolysosomes and was responsible for the accumulation of dysfunctional mitochondria and subsequent increase in oxidative stress, eventually leading to increased myocardial cell death.


Assuntos
Doxorrubicina , Miócitos Cardíacos , Ratos , Animais , Miócitos Cardíacos/metabolismo , Doxorrubicina/efeitos adversos , Autofagia , Mitocôndrias/metabolismo , Antibióticos Antineoplásicos/farmacologia , Estresse Oxidativo , Apoptose
2.
J Pharmacol Sci ; 145(1): 150-154, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33357773

RESUMO

Proteins interacting with G protein-coupled receptors (GPCRs) can modulate signal transduction of these receptors. However, the regulatory mechanisms of the interacting proteins are diverse and largely unknown. We have previously shown that Tctex-1 (or DYNLT1) can interact with the parathyroid hormone receptor (PTHR). In the present study, we investigated the role of Tctex-1 in the PTHR signaling and found that Tctex-1 augmented the PTHR-mediated Gs/adenylyl cyclase (AC) pathway by activating AC regardless of the binding to PTHR. Furthermore, Tctex-1 directly bound to AC type 6. These data demonstrate a novel mechanism underlying GPCR/Gs signaling regulated by Tctex-1.


Assuntos
Adenilil Ciclases/metabolismo , Dineínas/metabolismo , Dineínas/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células 3T3 , Animais , Células HEK293 , Humanos , Camundongos , Ligação Proteica , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/fisiologia
3.
Mol Pharmacol ; 96(4): 441-451, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31383768

RESUMO

The G protein-coupled receptor (GPCR) signaling pathways mediated by trimeric G proteins have been extensively elucidated, but their associated regulatory mechanisms remain unclear. Parathyroid hormone (PTH)/PTH-related protein receptor (PTHR) is a GPCR coupled with Gs and Gq Gs activates adenylyl cyclases (ACs), which produces cAMP to regulate various cell fates. We previously showed that cell surface expression of PTHR was increased by its direct interaction with a subcortical cytoskeletal protein, 4.1G, whereas PTHR-mediated Gs/AC/cAMP signaling was suppressed by 4.1G through an unknown mechanism in human embryonic kidney (HEK)293 cells. In the present study, we found that AC type 6 (AC6), one of the major ACs activated downstream of PTHR, interacts with 4.1G in HEK293 cells, and the N-terminus of AC6 (AC6-N) directly and selectively binds to the 4.1/ezrin/radixin/moesin (FERM) domain of 4.1G (4.1G-FERM) in vitro. AC6-N was distributed at the plasma membrane, which was disturbed by knockdown of 4.1G. An AC6-N mutant, AC6-N-3A, in which three consecutive arginine residues are mutated to alanine residues, altered both binding to 4.1G-FERM and its plasma membrane distribution in vivo. Further, we overexpressed AC6-N to competitively inhibit the interaction of endogenous AC6 and 4.1G in cells. cAMP production induced by forskolin, an adenylyl cyclase activator, and PTH-(1-34) was enhanced by AC6-N expression and 4.1G-knockdown. In contrast, AC6-N-3A had no impact on forskolin- and PTH-(1-34)-induced cAMP productions. These data provide a novel regulatory mechanism that AC6 activity is suppressed by the direct binding of 4.1G to AC6-N, resulting in attenuation of PTHR-mediated Gs/AC6/cAMP signaling.


Assuntos
Adenilil Ciclases/química , Adenilil Ciclases/metabolismo , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Adenilil Ciclases/genética , Sítios de Ligação , Membrana Celular/metabolismo , Colforsina/farmacologia , AMP Cíclico/metabolismo , Ativação Enzimática , Células HEK293 , Humanos , Mutação , Ligação Proteica , Transdução de Sinais
4.
EBioMedicine ; 20: 27-38, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28579242

RESUMO

Mitochondrial dysfunction increases oxidative stress and depletes ATP in a variety of disorders. Several antioxidant therapies and drugs affecting mitochondrial biogenesis are undergoing investigation, although not all of them have demonstrated favorable effects in the clinic. We recently reported a therapeutic mitochondrial drug mitochonic acid MA-5 (Tohoku J. Exp. Med., 2015). MA-5 increased ATP, rescued mitochondrial disease fibroblasts and prolonged the life span of the disease model "Mitomouse" (JASN, 2016). To investigate the potential of MA-5 on various mitochondrial diseases, we collected 25 cases of fibroblasts from various genetic mutations and cell protective effect of MA-5 and the ATP producing mechanism was examined. 24 out of the 25 patient fibroblasts (96%) were responded to MA-5. Under oxidative stress condition, the GDF-15 was increased and this increase was significantly abrogated by MA-5. The serum GDF-15 elevated in Mitomouse was likewise reduced by MA-5. MA-5 facilitates mitochondrial ATP production and reduces ROS independent of ETC by facilitating ATP synthase oligomerization and supercomplex formation with mitofilin/Mic60. MA-5 reduced mitochondria fragmentation, restores crista shape and dynamics. MA-5 has potential as a drug for the treatment of various mitochondrial diseases. The diagnostic use of GDF-15 will be also useful in a forthcoming MA-5 clinical trial.


Assuntos
Ácidos Indolacéticos/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Fenilbutiratos/farmacologia , Multimerização Proteica/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Biomarcadores , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , DNA Mitocondrial , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/metabolismo , Fibroblastos/metabolismo , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/genética , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/genética , Dinâmica Mitocondrial/efeitos dos fármacos , ATPases Mitocondriais Próton-Translocadoras/química , Complexos Multiproteicos/metabolismo , Mutação , Biogênese de Organelas , Prognóstico , Substâncias Protetoras , Ligação Proteica
6.
Methods Mol Biol ; 1317: 55-67, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26072401

RESUMO

Suicide gene therapy of cancer (SGTC) entails the introduction of a cDNA sequence into tumor cells whose polypeptide product is capable of either directly activating apoptotic pathways itself or facilitating the activation of pharmacologic agents that do so. The latter class of SGTC approaches is of the greater utility in cancer therapy owing to the ability of some small, activated cytotoxic compounds to diffuse from their site of activation into neighboring malignant cells, where they can also mediate destruction. This phenomenon, termed "bystander killing", can be highly advantageous in driving significant tumor regression in vivo without the requirement of transduction of each and every tumor cell with the suicide gene. We have developed a robust suicide gene therapy enzyme/prodrug system based on an engineered variant of the human thymidylate kinase (TMPK), which has been endowed with the ability to drive azidothymidine (AZT) activation. Delivery of this suicide gene sequence into tumors by means of recombinant lentivirus-mediated transduction embodies an SGTC strategy that successfully employs bystander cell killing as a mechanism to achieve significant ablation of solid tumors in vivo. Thus, this engineered TMPK/AZT suicide gene therapy axis holds great promise for clinical application in the treatment of inoperable solid tumors in the neoadjuvant setting. Here we present detailed procedures for the preparation of recombinant TMPK-based lentivirus, transduction of target cells, and various approaches for the evaluation of bystander cell killing effects in SGCT in both in vitro and in vivo models.


Assuntos
Efeito Espectador/efeitos dos fármacos , Genes Transgênicos Suicidas , Terapia Genética/métodos , Neoplasias/genética , Neoplasias/terapia , Núcleosídeo-Fosfato Quinase/genética , Pró-Fármacos/farmacologia , Zidovudina/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Colorimetria , Expressão Gênica , Engenharia Genética , Células HEK293 , Humanos , Lentivirus/metabolismo , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Núcleosídeo-Fosfato Quinase/uso terapêutico , Proteínas Recombinantes/metabolismo , Transdução Genética , Transgenes
7.
Tohoku J Exp Med ; 236(3): 225-32, 2015 07.
Artigo em Inglês | MEDLINE | ID: mdl-26118651

RESUMO

Mitochondria are key organelles implicated in a variety of processes related to energy and free radical generation, the regulation of apoptosis, and various signaling pathways. Mitochondrial dysfunction increases cellular oxidative stress and depletes ATP in a variety of inherited mitochondrial diseases and also in many other metabolic and neurodegenerative diseases. Mitochondrial diseases are characterized by the dysfunction of the mitochondrial respiratory chain, caused by mutations in the genes encoded by either nuclear DNA or mitochondrial DNA. We have hypothesized that chemicals that increase the cellular ATP levels may ameliorate the mitochondrial dysfunction seen in mitochondrial diseases. To search for the potential drugs for mitochondrial diseases, we screened an in-house chemical library of indole-3-acetic-acid analogs by measuring the cellular ATP levels in Hep3B human hepatocellular carcinoma cells. We have thus identified mitochonic acid 5 (MA-5), 4-(2,4-difluorophenyl)-2-(1H-indol-3-yl)-4-oxobutanoic acid, as a potential drug for enhancing ATP production. MA-5 is a newly synthesized derivative of the plant hormone, indole-3-acetic acid. Importantly, MA-5 improved the survival of fibroblasts established from patients with mitochondrial diseases under the stress-induced condition, including Leigh syndrome, MELAS (myopathy encephalopathy lactic acidosis and stroke-like episodes), Leber's hereditary optic neuropathy, and Kearns-Sayre syndrome. The improved survival was associated with the increased cellular ATP levels. Moreover, MA-5 increased the survival of mitochondrial disease fibroblasts even under the inhibition of the oxidative phosphorylation or the electron transport chain. These data suggest that MA-5 could be a therapeutic drug for mitochondrial diseases that exerts its effect in a manner different from anti-oxidant therapy.


Assuntos
Trifosfato de Adenosina/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Descoberta de Drogas , Fibroblastos/efeitos dos fármacos , Ácidos Indolacéticos/química , Ácidos Indolacéticos/farmacologia , Doenças Mitocondriais/tratamento farmacológico , Fenilbutiratos/farmacologia , Análise de Variância , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Fibroblastos/fisiologia , Humanos , Fosforilação Oxidativa , Fenilbutiratos/química , Bibliotecas de Moléculas Pequenas
8.
PLoS One ; 8(10): e78711, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24194950

RESUMO

We previously described a novel suicide (or 'cell fate control') gene therapy enzyme/prodrug system based on an engineered variant of human thymidylate kinase (TMPK) that potentiates azidothymidine (AZT) activation. Delivery of a suicide gene sequence into tumors by lentiviral transduction embodies a cancer gene therapy that could employ bystander cell killing as a mechanism driving significant tumor regression in vivo. Here we present evidence of a significant bystander cell killing in vitro and in vivo mediated by the TMPK/AZT suicide gene axis that is reliant on the formation of functional gap-junctional intercellular communications (GJICs). Potentiation of AZT activation by the engineered TMPK expressed in the human prostate cancer cell line, PC-3, resulted in effective bystander killing of PC-3 cells lacking TMPK expression--an effect that could be blocked by the GJIC inhibitor, carbenoxolone. Although GJICs are mainly formed by connexins, a new family of GJIC molecules designated pannexins has been recently identified. PC-3 cells expressed both connexin43 (Cx43) and Pannexin1 (Panx1), but Panx1 expression predominated at the plasma membrane, whereas Cx43 expression was primarily localized to the cytosol. The contribution of bystander effects to the reduction of solid tumor xenografts established by the PC-3 cell line was evaluated in an animal model. We demonstrate the contribution of bystander cell killing to tumor regression in a xenograft model relying on the delivery of expression of the TMPK suicide gene into tumors via direct intratumoral injection of recombinant therapeutic lentivirus. Taken together, our data underscore that the TMPK/AZT enzyme-prodrug axis can be effectively utilized in suicide gene therapy of solid tumors, wherein significant tumor regression can be achieved via bystander effects mediated by GJICs.


Assuntos
Efeito Espectador/fisiologia , Genes Transgênicos Suicidas/genética , Terapia Genética/métodos , Núcleosídeo-Fosfato Quinase/genética , Pró-Fármacos/farmacologia , Neoplasias da Próstata/terapia , Zidovudina/farmacologia , Análise de Variância , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Citometria de Fluxo , Células HEK293 , Humanos , Masculino , Microscopia Confocal , Pró-Fármacos/química , Espécies Reativas de Oxigênio/metabolismo , Zidovudina/química
10.
Mol Ther ; 20(5): 1002-13, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22273576

RESUMO

The safety of cell therapy applications can be enhanced by the introduction of Cell Fate Control (CFC) elements, which encode pharmacologically controlled cellular suicide switches. CFC Gene Therapy (CFCGT) offers the possibility of establishing control over gene-modified cells (GMCs) with regards to their proliferation, differentiation, or function. However, enzymes commonly employed in these approaches often possess poor kinetics and high immunogenicity. We describe a novel CFCGT system based on engineered variants of human deoxyCytidine Kinase (dCK) that overcomes limitations of current modalities. Mutants of dCK with rationally designed active sites that make them thymidine-activating were stably introduced into cells by recombinant lentiviral vectors (LVs). Transduced cells maintained growth kinetics and function. These dCK mutants efficiently activate bromovinyl-deoxyuridine (BVdU), L-deoxythymidine (LdT), and L-deoxyuridine (LdU), which are otherwise not toxic to wild-type cells. We show that mutant dCK-expressing Jurkat, Molt-4, and U87mg cells could be efficiently eliminated in vitro and in xenogeneic leukemia and tumor models in vivo. We also describe a fusion construct of the thymidine-activating dCK to the cytoplasmic tail-truncated LNGFR molecule and applications to in vivo eradication of primary human T cells. This novel CFCGT system offers unique plasticity with respect to the wide range of prodrugs it can potentiate, and can be used as a reliable safety switch in cell and gene therapy.


Assuntos
Desoxicitidina Quinase/genética , Terapia Genética/métodos , Neoplasias/tratamento farmacológico , Animais , Bromodesoxiuridina/administração & dosagem , Domínio Catalítico , Linhagem Celular Tumoral , Desoxicitidina Quinase/metabolismo , Desoxiuridina/administração & dosagem , Ativação Enzimática/efeitos dos fármacos , Engenharia Genética , Vetores Genéticos , Humanos , Lentivirus/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Cultura Primária de Células , Pró-Fármacos/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T/patologia , Timidina/administração & dosagem , Timidina/genética , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cell Signal ; 23(11): 1843-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21749919

RESUMO

Functional roles of putative helix 8 in the carboxy-terminal tail of the human histamine H(3) receptor were investigated using deleted and alanine-substituted mutant receptors. While the deletion of the carboxy-terminal tail did not decrease the total expression level, surface expression, or ligand binding affinity, the agonist-stimulated cAMP response, [((35))S] GTPγS binding, and MAPK activation were totally abolished. The receptor lacking the carboxy-terminal tail also failed to respond to an inverse agonist, thioperamide, suggesting that the carboxy-terminal tail is involved in the regulation of receptor activity by changing G-protein coupling with the receptor. Site-directed mutagenesis revealed that hydrophobic amino acids in the putative helix 8 such as phenylalanines at position 419 (F7.60) and 423 (F7.64) or leucines at 426 (L7.67) and 427 (L7.68) were important for the agonist-induced activation of H(3) receptor. Substitution of F7.60 also resulted in a receptor that was less responsive to inactivation by the inverse agonist, implying the existence of an intermediate conformation that can be either activated or inactivated. Our results suggest that hydrophobic interface of putative helix 8 is important for the regulation of H(3) receptor activity, presumably by stabilizing the helix to the plasma membrane.


Assuntos
Aminoácidos/química , Proteínas de Ligação ao GTP/metabolismo , Agonistas dos Receptores Histamínicos/farmacologia , Mutação , Receptores Histamínicos H3 , Transdução de Sinais/genética , Sequência de Aminoácidos , Aminoácidos/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Citometria de Fluxo , Células HEK293 , Histamina/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Piperidinas/farmacologia , Plasmídeos , Ligação Proteica , Estrutura Secundária de Proteína , Receptores Histamínicos H3/química , Receptores Histamínicos H3/genética , Receptores Histamínicos H3/metabolismo , Relação Estrutura-Atividade , Transfecção
12.
J Cardiol ; 54(2): 245-54, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19782262

RESUMO

For some applications, the success of gene therapy depends on the efficiency of gene transfer into target organs, however, delivery to many tissues is limited. Efforts have been made to improve the efficiency of gene transfer into target organs such as the brain by using mannitol or vascular endothelial growth factor (VEGF) prior to gene delivery, since these treatments have been reported to increase vascular permeability in experimental animals. Here, we investigated the effect of VEGF pretreatment of neonatal mice on the ability of injected lentivirus (LV)--engineering expression of firefly luciferase (luc)--to enhance the transduction of various organs, including the brain and heart. LV/luc was delivered to VEGF-treated neonatal mice via the temporal vein. Whole-body bioluminescence imaging (WBLI) of luciferase expression showed that VEGF pretreatment does not diminish transgene expression over time since it remained steady for up to 12 weeks. Ex vivo imaging of the organs and assessments of organ luciferase activity showed that VEGF pretreatment resulted in significantly increased luciferase expression not only in the heart, but also in the brain, lung, and kidney. This study shows that VEGF may have therapeutic importance to enhance the efficiency of viral gene delivery to the heart, as well as to other target organs.


Assuntos
Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Lentivirus/genética , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Miocárdio/enzimologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Animais Recém-Nascidos , Luciferases de Vaga-Lume/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Estimulação Química , Transdução Genética , Fator A de Crescimento do Endotélio Vascular/administração & dosagem
13.
Biochem Biophys Res Commun ; 369(2): 603-8, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18302930

RESUMO

Histamine H3 receptor (H3R), one of G protein-coupled receptors (GPCRs), has been known to regulate neurotransmitter release negatively in central and peripheral nervous systems. Recently, a variety of intracellular proteins have been identified to interact with carboxy (C)-termini of GPCRs, and control their intracellular trafficking and signal transduction efficiencies. Screening for such proteins that interact with the C-terminus of H3R resulted in identification of one of the chloride intracellular channel (CLIC) proteins, CLIC4. The association of CLIC4 with H3R was confirmed in in vitro pull-down assays, coimmunoprecipitation from rat brain lysate, and immunofluorescence microscopy of rat cerebellar neurons. The data from flowcytometric analysis, radioligand receptor binding assay, and cell-based ELISA indicated that CLIC4 enhanced cell surface expression of wild-type H3R, but not a mutant form of the receptor that failed to interact with CLIC4. These results indicate that, by binding to the C-terminus of H3R, CLIC4 plays a critical role in regulation of the receptor cell surface expression.


Assuntos
Membrana Celular/metabolismo , Cerebelo/metabolismo , Canais de Cloreto/metabolismo , Neurônios/metabolismo , Receptores Histamínicos H3/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Células PC12 , Ratos , Ratos Wistar
14.
Pharmacology ; 81(3): 251-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18253064

RESUMO

We examined the effects of beta-adrenoceptor agonists on the membrane currents of smooth muscle cells from the human urinary bladder using a whole-cell patch clamp to investigate the involvement of Ca(2+)-activated K(+) (K(Ca)) channels in relaxation by beta-adrenergic agonists. With 0.05 mmol/l EGTA in the patch pipette, depolarizing pulses evoked outward rectifying currents. Isoproterenol (1 micromol/l) significantly increased the membrane currents by 75% at +80 mV with 0.05 mmol/l EGTA pipette solution. BRL 37344 (1 micromol/l) significantly increased the membrane currents by 44% at +80 mV. Iberiotoxin (100 nmol/l) significantly decreased the membrane currents by 60% at +80 mV. In the presence of iberiotoxin, the potentiation of the outward currents by isoproterenol was greatly suppressed and, in the presence of iberiotoxin and apamin (1 micromol/l), the potentiation by isoproterenol was totally abolished. On the other hand, with 5 mmol/l EGTA pipette solution, depolarizing pulses evoked smaller outward currents. Isoproterenol (1 micromol/l) did not change the membrane currents with 5 mmol/l EGTA pipette solution. The real-time PCR analysis revealed the expression of beta(2)-adrenoceptors in the cells. These results suggest that Ca(2+)-activated and iberiotoxin- and apamin-sensitive currents via both large-conductance and small-conductance K(Ca) channels could be increased by stimulation of beta(2)-adrenoceptors.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Receptores Adrenérgicos beta/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/efeitos dos fármacos , Apamina/farmacologia , Linhagem Celular , Eletrofisiologia , Etanolaminas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Isoproterenol/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Bexiga Urinária/metabolismo
15.
Mol Ther ; 15(5): 962-70, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17375075

RESUMO

Gene therapy and stem cell transplantation safety could be enhanced by control over the fate of therapeutic cells. Suicide gene therapy uses enzymes that convert prodrugs to cytotoxic entities; however, heterologous moieties with poor kinetics are employed. We describe a novel enzyme/prodrug combination for selectively inducing apoptosis in lentiviral vector-transduced cells. Rationally designed variants of human thymidylate kinase (tmpk) that effectively phosphorylate 3'-azido-3'-deoxythymidine (AZT) were efficiently delivered. Transduced Jurkat cell lines were eliminated by AZT. We demonstrate that this schema targeted both dividing and non-dividing cells, with a novel killing mechanism involving apoptosis induction via disruption of the mitochondrial inner membrane potential and activation of caspase-3. Primary murine and human T cells were also transduced and responded to AZT. Furthermore, low-dose AZT administration to non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice injected with transduced K562 cells suppressed tumor growth. This novel suicide gene therapy approach can thus be integrated as a safety switch into therapeutic vectors.


Assuntos
Terapia Genética/métodos , Núcleosídeo-Fosfato Quinase/genética , Pró-Fármacos/farmacologia , Zidovudina/farmacologia , Animais , Antígenos CD19/metabolismo , Antimetabólitos/metabolismo , Antimetabólitos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Caspase 3/metabolismo , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Feminino , Citometria de Fluxo , Genes Transgênicos Suicidas , Vetores Genéticos/genética , Humanos , Células Jurkat , Células K562 , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Núcleosídeo-Fosfato Quinase/metabolismo , Pró-Fármacos/metabolismo , Pró-Fármacos/uso terapêutico , Linfócitos T/imunologia , Transfecção , Zidovudina/metabolismo , Zidovudina/uso terapêutico
16.
Int J Cancer ; 118(1): 62-73, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16049981

RESUMO

Our study reports a preliminary investigation into the role of human H2 relaxin in prostate tumor growth. A luciferase-expressing human prostate cancer cell line, PC-3, was generated and termed PC3-Luc. PC3-Luc cells were transduced with lentiviral vectors engineering the expression of either enhanced green fluorescent protein (eGFP) or both H2 relaxin and eGFP in a bicistronic format. These transduced cells were termed PC3-Luc-eGFP and PC3-Luc-H2/eGFP, respectively. To gauge effects, PC3-Luc-H2/eGFP and PC3-Luc-eGFP cells were injected into NOD/SCID mice and monitored over 6 weeks. PC-3 tumor xenografts overexpressing H2 relaxin exhibited greater tumor volumes compared to control tumors. Circulating H2 relaxin levels in sera increased with the relative size of the tumor, with moderately elevated H2 relaxin levels in mice bearing PC3-Luc-H2/eGFP tumors compared to PC3-Luc-eGFP tumors. Zymographic analysis demonstrated that proMMP-9 enzyme activity was significantly downregulated in H2 relaxin-overexpressing tumors. An advanced angiogenic phenotype was observed in H2 relaxin-overexpressing tumors indicated by greater intratumoral vascularization by immunohistochemical staining of endothelial cells with anti-mouse CD31. Moreover, PC3-Luc-H2/eGFP tumors exhibited increased VEGF transcript by reverse-transcription PCR, compared to basal levels in control animals. Taken together, our study provides the first account of a potential role of H2 relaxin in prostate tumor development.


Assuntos
Neovascularização Patológica , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Relaxina/biossíntese , Animais , Proliferação de Células , Marcadores Genéticos , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos SCID , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Transplante Heterólogo , Células Tumorais Cultivadas , Regulação para Cima
17.
Proc Natl Acad Sci U S A ; 101(48): 16909-14, 2004 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-15550536

RESUMO

Successful therapy for many inherited disorders could be improved if the intervention were initiated early. This is especially true for lysosomal storage disorders. Earlier intervention may allow metabolic correction to occur before lipid buildup has irreversible consequences and/or before the immune system mounts limiting responses. We have been developing gene therapy to treat lysosomal storage disorders, especially Fabry disease. We describe studies directed toward metabolic correction in neonatal animals mediated by recombinant lentiviral vectors. To develop this method, we first injected a marking lentiviral vector that engineers expression of luciferase into the temporal vein of recipient neonatal animals. The use of a cooled charged-coupled device camera allowed us to track transgene expression over time in live animals. We observed intense luciferase expression in many tissues, including the brain, that did not diminish over 24 weeks. Next, we injected neonatal Fabry mice a single time with a therapeutic lentiviral vector engineered to express human alpha-galactosidase A. The injection procedure was well tolerated. We observed increased plasma levels of alpha-galactosidase A activity starting at our first plasma collection point (4 weeks). Levels of alpha-galactosidase A activity were found to be significantly elevated in many tissues even after 28 weeks. No immune response was observed against the corrective transgene product. Increased levels of enzyme activity also led to significant reduction of globotriaosylceramide in the liver, spleen, and heart. This approach provides a method to treat lysosomal storage disorders and other disorders before destructive manifestations occur.


Assuntos
Doença de Fabry/terapia , Vetores Genéticos , Lentivirus/genética , Transgenes , Animais , Animais Recém-Nascidos , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Ensaio de Imunoadsorção Enzimática , Doença de Fabry/genética , Humanos , Imuno-Histoquímica , Camundongos
18.
Comp Hepatol ; 3 Suppl 1: S4, 2004 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-14960156

RESUMO

HSCs showed myofibroblast-like shapes when cultured on polystyrene surface or on type I collagen-coated surface, whereas HSCs cultured on type I collagen gel were induced to elongate cellular processes, suggesting that HSCs recognize 3-D structure of extracellular type I collagen fibrils and change their morphology and function. In this study we examined the differentially regulated gene expression by extracellular matrix (ECM) components by PCR-differential display (PCR-DD) analysis followed by cloning and FASTA homology search, and identified the mRNA species as a transcription factor SP1, breast cancer resistant protein (BCRP), dystonin, and KAP3B. Regulation of dystonin and KAP3B expression was confirmed by RT-PCR analysis. Thus, cell surface-binding to extracellular interstitial collagen may trigger intracellular signaling and alteration in gene expression, and HSCs not only produce various ECM components but also change their morphology and gene expression in response to ECM components adhering to the cells.

19.
Lab Invest ; 82(9): 1209-17, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12218082

RESUMO

Recent studies suggested that the vascular endothelial cells function as a resident antigen-presenting cell (APC) in certain situations such as organ transplantation, and the ischemia/reperfusion injury, an inevitable event in organ transplantation, leads to an enhanced biosynthesis of cell adhesion molecules. We have demonstrated that the hepatic sinusoidal endothelial cells have potential ability as APCs by expressing the costimulatory adhesion molecule proteins, CD80 (B7-1) and CD86 (B7-2), of which expression was enhanced by warm ischemia/reperfusion of the rat liver. In this study, we assessed the localization of CD80, CD86, and intercellular adhesion molecule 1 in the rat kidneys and the influence of warm ischemia/reperfusion with or without a hypercreatinemic condition on the expression of these adhesion molecules in the renal tissues. Wistar male rats weighing 150 to 230 g were divided into group A, receiving a sham-operation (control), group B, receiving 1-hour clamping of the left renal pedicle (temporary ischemia), and group C, receiving right nephrectomy and 1-hour clamping of the left renal pedicle (temporary ischemia with hypercreatinemia). The left kidneys were submitted to immunohistochemical and molecular analyses sequentially for the period of 14 days. We found that CD80, CD86, and intercellular adhesion molecule 1 proteins localized on the glomerular and peritubular endothelium and were up-regulated after ischemia/reperfusion. The up-regulation of these three proteins was enhanced by the hypercreatinemic condition. The relative mRNA levels analyzed by real-time reverse transcription polymerase chain reaction showed that CD80 and CD86 expressions were constitutively observed and significantly increased for 14 days after the warm ischemia reperfusion with a peak level at Day 3 (6.7- and 20.8-fold increase for CD80 and CD86, respectively). Our results suggested that the glomerular endothelial cells will play a pivotal role as a APC by expressing CD80 and CD86 in the induction of renal tissue injury associated with the ischemia/reperfusion at renal transplantation surgery, as well as the peritubular endothelium.


Assuntos
Antígenos CD/análise , Antígeno B7-1/análise , Endotélio Vascular/química , Isquemia/metabolismo , Glomérulos Renais/química , Glicoproteínas de Membrana/análise , Traumatismo por Reperfusão/metabolismo , Animais , Células Apresentadoras de Antígenos/fisiologia , Antígenos CD/genética , Antígeno B7-1/genética , Antígeno B7-2 , Creatinina/sangue , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Molécula 1 de Adesão Intercelular/análise , Molécula 1 de Adesão Intercelular/genética , Glomérulos Renais/metabolismo , Masculino , Glicoproteínas de Membrana/genética , RNA Mensageiro/análise , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA