Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
J Leukoc Biol ; 88(4): 779-89, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20631258

RESUMO

Infectious pathogens produce compounds called Toll ligands that activate TLRs on lymphocytes. Acute activation triggered by certain TLRs appears to "jump start" the innate immune response, characterized by the release of inflammatory cytokines and cellular expansion. In some individuals, there is a failure to control acute inflammation, resulting in postinfectious, chronic inflammation. Susceptibility to chronic inflammation is strongly associated with an individual's MHC genes. Recent clinical trials for several autoimmune diseases characterized by chronic inflammation suggest that B lymphocyte depletion therapies dampen chronic immune activation. However, currently, there is no known mechanism that accounts for the correlation among TLR activation, MHC genetics, and a pathological role for B-lymphocytes. Our hypothesis is that TLR-activated B cells (B cells that have been polyclonally activated in the absence of antigen-specific signals) are not controlled properly by T cell-dependent B cell death, thereby causing B cell-dependent chronic inflammation. Here, we show that treatment with Toll ligands results in polyclonal B cell activation accompanied by ectopic expression of CLIP. Furthermore, by adoptively transferring purified CLIP+ B cells in syngeneic animals, we find that CLIP+ B cells induce production of TNF-α by host T cells. Finally, we demonstrate that CLIP-targeted peptide competition results in the death of polyclonally activated CLIP+ B cells.


Assuntos
Antígenos de Diferenciação de Linfócitos B/imunologia , Linfócitos B/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Inflamação/imunologia , Ativação Linfocitária/imunologia , Receptores Toll-Like/imunologia , Transferência Adotiva , Animais , Antígenos de Diferenciação de Linfócitos B/biossíntese , Linfócitos B/metabolismo , Separação Celular , Células Cultivadas , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL/metabolismo
2.
Neuromuscul Disord ; 15(8): 541-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16014330

RESUMO

The primary receptor for Adenovirus and Coxsackie virus (CAR) serves as main port of entry of the adenovirus vector mediating gene transfer into skeletal muscle. Information about CAR expression in normal and diseased human skeletal muscle is lacking. C'- or N'-terminally directed polyclonal antibodies against CAR were generated and immunohistochemical analysis of CAR on morphologically normal and regenerating human skeletal muscle of children and adults was performed. In morphologically normal human muscle fibers, CAR immunoreactivity was limited to the neuromuscular junction. In regenerating muscle fibers, CAR was abundantly co-expressed with markers of regeneration. The function of CAR at the neuromuscular junction is currently unknown. Co-expression of CAR with markers of regeneration suggests that CAR is developmentally regulated, and may serve as a marker of skeletal muscle fiber regeneration.


Assuntos
Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo , Receptores Virais/metabolismo , Regeneração/fisiologia , Adolescente , Adulto , Fatores Etários , Idoso , Criança , Pré-Escolar , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Desmina/metabolismo , Humanos , Imunoglobulinas/imunologia , Imuno-Histoquímica/métodos , Lactente , Pessoa de Meia-Idade , Distrofia Muscular de Duchenne/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Junção Neuromuscular/metabolismo , Junção Neuromuscular/fisiologia , Polimiosite/metabolismo , Receptores Virais/química , Coloração e Rotulagem/métodos , Utrofina/metabolismo
3.
Mol Ther ; 4(5): 499-507, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11708887

RESUMO

Duchenne muscular dystrophy (DMD) is caused by defects in the dystrophin gene. In young dystrophic mdx mice, immature regenerating myofibers represent the principal substrate for adenovirus vector (AdV)-mediated dystrophin gene transfer. However, in DMD patients immature regenerating myofibers are generally sparse. Such a situation also exists in old mdx mice, which may represent a more realistic model. Therefore, here we have used old mdx mice (of 14- to 17 months of age) to test the hypothesis that one-time administration of a myonecrotic agent can transiently re-establish a population of immature myofibers susceptible to AdV-mediated dystrophin gene transfer. This strategy led to upregulation of the coxsackie/adenovirus attachment receptor by means of induction of regenerating myofibers, significantly augmented AdV-mediated dystrophin gene expression, and enhanced force-generating capacity. In addition, it led to an increased resistance to contraction-induced injury compared with untreated controls. The latter protective effect was positively correlated with the number of dystrophin-expressing myofibers (r=0.83, P<0.05). Accordingly, the risk:benefit ratio associated with the sequential use of forced myofiber regeneration and AdV-mediated dystrophin gene transfer was favorable in old mdx mice despite advanced disease. These findings have implications for the potential applicability of AdV-mediated gene therapy to DMD and other muscle diseases in which immature regenerating myofibers are lacking.


Assuntos
Envelhecimento/fisiologia , Distrofina/genética , Distrofina/uso terapêutico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Miofibrilas/fisiologia , Regeneração , Adenoviridae/genética , Animais , Western Blotting , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Distrofina/administração & dosagem , Venenos Elapídicos/farmacologia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Imuno-Histoquímica , Contração Isométrica , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Miofibrilas/efeitos dos fármacos , Miofibrilas/genética , Miofibrilas/patologia , Receptores Virais/biossíntese , Receptores Virais/genética , Regeneração/efeitos dos fármacos , Estresse Mecânico , Transgenes/genética , Regulação para Cima/efeitos dos fármacos
4.
Hum Gene Ther ; 12(14): 1741-55, 2001 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-11560768

RESUMO

Helper-dependent adenoviruses (HDAd) are Ad vectors lacking all or most viral genes. They hold great promise for gene therapy of diseases such as Duchenne muscular dystrophy (DMD), because they are less immunogenic than E1/E3-deleted Ad (first-generation Ad or FGAd) and can carry the full-length (Fl) dystrophin (dys) cDNA (12 kb). We have compared the transgene expression of a HDAd (HDAdCMVDysFl) and a FGAd (FGAdCMV-dys) in cell culture (HeLa, C2C12 myotubes) and in the muscle of mdx mice (the mouse model for DMD). Both vectors encoded dystrophin regulated by the same cytomegalovirus (CMV) promoter. We demonstrate that the amount of dystrophin expressed was significantly higher after gene transfer with FGAdCMV-dys compared to HDAdCMVDysFl both in vitro and in vivo. However, gene transfer with HDAdCMVDysFl in the presence of a FGAd resulted in a significant increase of dystrophin expression indicating that gene products synthesized by the FGAd increase, in trans, the amount of dystrophin produced. This enhancement occurred in cell culture and after gene transfer in the muscle of mdx mice and dystrophic golden retriever (GRMD) dogs, another animal model for DMD. The E4 region of Ad is required for the enhancement, because no increase of dystrophin expression from HDAdCMVDysFl was observed in the presence of an E1/E4-deleted Ad in vitro and in vivo. The characterization of these enhancing gene products followed by their inclusion into an HDAd may be required to produce sufficient dystrophin to mitigate the pathology of DMD by HDAd-mediated gene transfer.


Assuntos
Adenoviridae/genética , Distrofina/biossíntese , Técnicas de Transferência de Genes , Músculos/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Linhagem Celular , Células Cultivadas , Citomegalovirus/genética , DNA Complementar/metabolismo , Cães , Vetores Genéticos , Células HeLa , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos mdx , Fenótipo , Regiões Promotoras Genéticas , Mapeamento por Restrição , Ativação Transcricional , Transgenes
5.
J Virol ; 75(9): 4276-82, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11287577

RESUMO

Significant levels of adenovirus (Ad)-mediated gene transfer occur only in immature muscle or in regenerating muscle, indicating that a developmentally regulated event plays a major role in limiting transgene expression in mature skeletal muscle. We have previously shown that in developing mouse muscle, expression of the primary Ad receptor CAR is severely downregulated during muscle maturation. To evaluate how global expression of CAR throughout muscle affects Ad vector (AdV)-mediated gene transfer into mature skeletal muscle, we produced transgenic mice that express the CAR cDNA under the control of the muscle-specific creatine kinase promoter. Five-month-old transgenic mice were compared to their nontransgenic littermates for their susceptibility to AdV transduction. In CAR transgenics that had been injected in the tibialis anterior muscle with AdVCMVlacZ, increased gene transfer was demonstrated by the increase in the number of transduced muscle fibers (433 +/- 121 in transgenic mice versus 8 +/- 4 in nontransgenic littermates) as well as the 25-fold increase in overall beta-galactosidase activity. Even when the reporter gene was driven by a more efficient promoter (the cytomegalovirus enhancer-chicken beta-actin gene promoter), differential transducibility was still evident (893 +/- 149 versus 153 +/- 30 fibers; P < 0.001). Furthermore, a fivefold decrease in the titer of injected AdV still resulted in significant transduction of muscle (253 +/- 130 versus 14 +/- 4 fibers). The dramatic enhancement in AdV-mediated gene transfer to mature skeletal muscle that is observed in the CAR transgenics indicates that prior modulation of the level of CAR expression can overcome the poor AdV transducibility of mature skeletal muscle and significant transduction can be obtained at low titers of AdV.


Assuntos
Adenovírus Humanos/fisiologia , Técnicas de Transferência de Genes , Vetores Genéticos/fisiologia , Músculo Esquelético/metabolismo , Receptores Virais/fisiologia , Adenovírus Humanos/genética , Animais , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Creatina Quinase/genética , Creatina Quinase Forma MM , Expressão Gênica , Genes Reporter , Vetores Genéticos/genética , Humanos , Isoenzimas/genética , Óperon Lac , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Receptores Virais/biossíntese , Receptores Virais/genética
6.
Glia ; 33(3): 217-24, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11241739

RESUMO

Fas (APO-1/CD95) is a cell surface receptor initially identified in lymphoid cells, but more recently detected in the central nervous system under pathological, usually inflammatory, conditions. In most Fas expressing cells, triggering of Fas by its ligand or by antagonistic antibodies leads to apoptosis. Human fetal astrocytes (HFA) constitutively express Fas yet are resistant to cell death following Fas ligation. In the current study, using dissociated cultures of human fetal central nervous system-derived cells, we attempted to identify a basis for HFA resistance to Fas-mediated injury. We compared the components of the Fas signaling pathway of HFA to those of two human cell lines susceptible to Fas-mediated injury, U251 glioma and Jurkat T-cells. We found that HFA did not express caspase 8 (FLICE), the caspase primarily activated on Fas signaling. Although we could induce caspase 8 in HFA with the inflammatory cytokines IFNgamma and TNFalpha, HFA remained resistant to Fas-mediated injury. Addition of inflammatory cytokines to the extracellular milieu also increased FLIP mRNA (FLICE inhibitory protein). Furthermore, upon triggering of cytokine-treated cells with FasL, we observed upregulation of the cleavage product of FLIP (p43-FLIP) previously shown to associate with the DISC and to block caspase 8 recruitment, thereby inhibiting Fas-mediated death. Our findings indicate that caspase 8 and its regulators play a central role in determining the response to Fas ligation of HFA and support a role for Fas signaling in the developing central nervous system other than related to cytotoxicity.


Assuntos
Astrócitos/enzimologia , Caspases/genética , Caspases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Transdução de Sinais/fisiologia , Receptor fas/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Astrócitos/citologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 8 , Caspase 9 , Córtex Cerebral/citologia , Citotoxinas/metabolismo , Ativação Enzimática/fisiologia , Feto/citologia , Regulação Enzimológica da Expressão Gênica , Humanos , Interferon gama/farmacologia , Células Jurkat , RNA Mensageiro/análise , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
7.
J Neural Transm Suppl ; (58): 193-203, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11128609

RESUMO

Development of neuroprotective therapies for multiple sclerosis is dependent on defining the precise mechanisms whereby immune effector cells and molecules are able to induce relatively selective injury of oligodendrocytes (OLs) and their myelin membranes. The selectivity of this injury could be conferred either by the properties of the effectors or the targets. The former would involve antigen specific recognition by either antibody or T cell receptor of the adaptive immune system. OLs are also susceptible to non antigen restricted injury mediated by components of the innate immune system including macrophages/microglia and NK cells. Target related selectivity could reflect the expression of death inducing surface receptors (such as Fas or TNFR-1) required for interaction with effector mediators and subsequent intracellular signaling pathways, including the caspase cascade. Development of therapeutic delivery systems, which would reach the site of disease activity within the CNS, will permit the administration of inhibitors either of the cell death pathway or of effector target interaction and opens new avenues to neuroprotection approach.


Assuntos
Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/fisiopatologia , Fármacos Neuroprotetores/uso terapêutico , Doenças Autoimunes/patologia , Doenças do Sistema Nervoso Central/patologia , Humanos , Esclerose Múltipla/etiologia , Esclerose Múltipla/imunologia , Oligodendroglia/patologia , Oligodendroglia/fisiologia , Linfócitos T Reguladores/fisiologia
8.
J Neuropathol Exp Neurol ; 59(10): 896-906, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11079780

RESUMO

The kinin B1 receptor is an inducible receptor expressed in response to inflammatory mediators. We sought to determine whether kinin B1 receptor can be expressed on human brain endothelial cells (HBECs) in vitro and whether signaling via this receptor can regulate permeability and chemokine production properties of these cells. Multiplex RT-PCR amplification and western blot techniques were used to evaluate B1 receptor expression by HBECs. Although B1 receptor mRNA and protein could not be detected on resting HBECs, interferon-gamma induced a dose- and time-dependent up-regulation of B1 receptor mRNA and protein on HBECs. Stimulation of interferon-gamma-treated HBECs with the selective B1 agonist R-838 (Sar [D-Phe8] des Arg9-BK) induced a dose- and time-dependent increase in the production of inositol 3,4,5 tri-phosphate and nitric oxide. Permeability of the HBECs monolayer, as measured by BSA diffusion, was significantly increased by application of the B1 agonist. This biological effect of R-838 could be prevented by R-715, a B1 receptor antagonist and by L-NAME, a nitric oxide synthase blocker. R-838 also inhibited interleukin-8 release from HBECs. We demonstrate that B1 receptors can be up regulated on the surface of HBECs by molecules released during inflammatory response and that signaling via this receptor can regulate BBB permeability and chemokine production in vitro.


Assuntos
Barreira Hematoencefálica/fisiologia , Bradicinina/análogos & derivados , Química Encefálica/fisiologia , Receptores da Bradicinina/genética , Receptores da Bradicinina/metabolismo , Soroalbumina Bovina/farmacocinética , Adulto , Barreira Hematoencefálica/efeitos dos fármacos , Bradicinina/farmacologia , Células Cultivadas , Endotélio Vascular/química , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Fluoresceína-5-Isotiocianato/farmacocinética , Corantes Fluorescentes/farmacocinética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Humanos , Fosfatos de Inositol/biossíntese , Interferon gama/farmacologia , Interleucina-8/biossíntese , Interleucina-8/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/biossíntese , RNA Mensageiro/metabolismo , Receptor B1 da Bradicinina , Receptores da Bradicinina/análise , Lobo Temporal/química , Lobo Temporal/metabolismo
9.
Physiol Genomics ; 3(3): 133-44, 2000 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-11015608

RESUMO

Duchenne muscular dystrophy (DMD) is a fatal disease caused by defects in the gene encoding dystrophin. Dystrophin is a cytoskeletal protein, which together with its associated protein complex, helps to protect the sarcolemma from mechanical stresses associated with muscle contraction. Gene therapy efforts aimed at supplying a normal dystrophin gene to DMD muscles could be hampered by host immune system recognition of dystrophin as a "foreign" protein. In contrast, a closely related protein called utrophin is not foreign to DMD patients and is able to compensate for dystrophin deficiency when overexpressed throughout development in transgenic mice. However, the issue of which of the two candidate molecules is superior for DMD therapy has remained an open question. In this study, dystrophin and utrophin gene transfer effects on dystrophic muscle function were directly compared in the murine (mdx) model of DMD using E1/E3-deleted adenovirus vectors containing either a dystrophin (AdV-Dys) or a utrophin (AdV-Utr) transgene. In immunologically immature neonatal animals, AdV-Dys and AdV-Utr improved tibialis anterior muscle histopathology, force-generating capacity, and the ability to resist injury caused by high-stress contractions to an equivalent degree. By contrast, only AdV-Utr was able to achieve significant improvement in force generation and the ability to resist stress-induced injury in the soleus muscle of immunocompetent mature mdx animals. In addition, in mature mdx mice, there was significantly greater transgene persistence and reduced inflammation with utrophin compared to dystrophin gene transfer. We conclude that dystrophin and utrophin are largely equivalent in their intrinsic abilities to prevent the development of muscle necrosis and weakness when expressed in neonatal mdx animals with an immature immune system. However, because immunity against dystrophin places an important limitation on the efficacy of dystrophin gene replacement in an immunocompetent mature host, the use of utrophin as an alternative to dystrophin gene transfer in this setting appears to offer a significant therapeutic advantage.


Assuntos
Proteínas do Citoesqueleto/genética , Distrofina/genética , Proteínas de Membrana/genética , Distrofia Muscular de Duchenne/terapia , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/fisiologia , DNA Recombinante/genética , Distrofina/análise , Distrofina/fisiologia , Técnicas de Transferência de Genes , Terapia Genética , Membro Posterior , Imunocompetência , Imuno-Histoquímica , Inflamação/genética , Inflamação/patologia , Proteínas de Membrana/análise , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos mdx , Contração Muscular , Músculos/química , Músculos/metabolismo , Músculos/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Estresse Mecânico , Utrofina
10.
Pathol Res Pract ; 196(4): 243-50, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10782468

RESUMO

p27kip1 and p21cip1 are cyclin-dependent kinase (cdk) inhibitors which along with p53 play critical roles in the control of cell cycle progression. Accumulation of p27kip1 in post-mitotic neurons is a major event of neurogenesis. We hypothesized that a dysregulation of the expression of p53 and these cdk inhibitors underlies cellular proliferation in medulloblastomas, and tested this hypothesis by investigating p27kip1, p21cip1, Bcl2 and p53 immunoreactivity in 14 medulloblastoma tumors. We noted an inverse relationship between p27kip1 expression and cellular proliferation (MIB1). Focal islands of neuroblastic or glial differentiation expressed high levels of p27kip1, while the undifferentiated, highly-proliferative population of tumor cells showed no detectable p27kip1 expression, thus suggesting a role for p27kip1 in cell cycle control in medulloblastoma. In addition, there was no detectable p21cip1 expression in any of the medulloblastomas studied. The low level of apoptosis displayed by these tumors was not associated with the expression of Bcl-2. A significant relationship was found between detection of p53 protein and poor survival. Since, p21cip1 and p27kip1 are often co-expressed with other INK4 family of cdk inhibitors during the induction of cellular differentiation and are synergistic in their effect, a deregulation of their coordinate expression may underlie the lack of complete differentiation in medulloblastoma.


Assuntos
Proteínas de Ciclo Celular , Neoplasias Cerebelares/metabolismo , Meduloblastoma/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Proteína Supressora de Tumor p53/biossíntese , Proteínas Supressoras de Tumor , Adolescente , Apoptose , Divisão Celular , Neoplasias Cerebelares/patologia , Criança , Pré-Escolar , Inibidor de Quinase Dependente de Ciclina p27 , Humanos , Imuno-Histoquímica , Lactente , Meduloblastoma/patologia , Proteínas Associadas aos Microtúbulos/análise , Análise de Sobrevida , Proteína Supressora de Tumor p53/análise
11.
Hum Gene Ther ; 11(5): 701-14, 2000 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-10757350

RESUMO

Duchenne muscular dystrophy (DMD) and other inherited myopathies lead to progressive destruction of most skeletal muscles in the body, including those responsible for maintaining respiration. DMD is a fatal disorder caused by defects in the dystrophin gene. Recombinant adenovirus vectors (AdV) are considered a promising means for therapeutic delivery of a functional dystrophin gene to DMD muscles. If AdV-mediated dystrophin gene replacement in DMD is to be successful, development of a systemic delivery method for targeting the large number of diseased muscles will be required. In this study we investigated two major factors preventing efficient AdV-mediated gene transfer to skeletal muscles of adult animals after intravascular AdV administration: (1) an inability of AdV particles to breach the endothelial barrier and enter into contact with myofibers, and (2) a relatively nonpermissive myofiber population for AdV infection due at least in part to insufficient levels of the coxsackie/adenovirus attachment receptor (CAR). On the basis of established principles governing the transendothelial flux of macromolecules, we further hypothesized that an alteration in Starling forces (increased hydrostatic and decreased osmotic pressures) within the intravascular compartment would facilitate AdV transendothelial flux via convective transport. In addition, experimental muscle regeneration was employed to increase the prevalence of immature myofibers in which CAR expression is upregulated. Here we report that by employing the above-described strategy, high-level heterologous reporter gene expression was achievable in hindlimb muscles of normal rats as well as dystrophic (mdx) mice (genetic homolog of DMD) after a single intraarterial injection of AdV. Microsphere studies confirmed enhanced transport into muscle of fluorescent tracer particles in the size range of AdV, and there was a high concordance between CAR upregulation and myofiber transduction after intraarterial AdV delivery. Furthermore, in mdx mice examined 10 days after intraarterial AdV delivery, the aforementioned procedures had no adverse effects on the force-generating capacity of targeted muscles. These findings have implications for eventual AdV-mediated gene therapy of generalized skeletal muscle diseases such as DMD using a systemic intraarterial delivery approach.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/patologia , Animais , Fenômenos Biomecânicos , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Distrofina/genética , Regulação da Expressão Gênica , Injeções Intra-Arteriais , Camundongos , Camundongos Mutantes , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Distrofias Musculares/terapia , Ratos , Ratos Sprague-Dawley , Receptores Virais/genética , Receptores Virais/metabolismo
13.
Neuroreport ; 11(1): 9-14, 2000 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-10683821

RESUMO

The neuronal apoptosis inhibitory protein (NAIP) is known to have anti-apoptotic functions, and its gene is often mutated in severe cases of spinal muscular atrophy (SMA), a disease characterized by motor neuron degeneration. In this study, we examined the distribution of the endogenous NAIP protein in normal human spinal cord and brain tissue by using a polyclonal antibody against NAIP. Immunohistochemical staining demonstrated that NAIP is strongly expressed in anterior horn and motor cortex neurons of normal brains, and it is not altered in the remaining motor neurons of patients with amyotrophic lateral sclerosis (ALS). NAIP is also located in human fetal neurons and in adult choroid plexus cells. These results suggest that the anti apoptotic molecule NAIP may be important in motor neurons, but it specifically does not appear to be altered in ALS.


Assuntos
Apoptose/fisiologia , Química Encefálica/fisiologia , Encéfalo/anatomia & histologia , Proteínas do Tecido Nervoso/metabolismo , Medula Espinal/anatomia & histologia , Medula Espinal/metabolismo , Adenoviridae/genética , Adulto , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Células do Corno Anterior/metabolismo , Células do Corno Anterior/patologia , Encéfalo/patologia , Química Encefálica/genética , Células Cultivadas , Células HeLa , Humanos , Imuno-Histoquímica , Microscopia Confocal , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Proteínas do Tecido Nervoso/genética , Proteína Inibidora de Apoptose Neuronal , Neurônios/metabolismo , Inclusão em Parafina , Medula Espinal/patologia , Transdução Genética/genética
14.
Neurology ; 53(6): 1352-4, 1999 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-10522901

RESUMO

McArdle's disease is due to a genetic deficiency of glycogen phosphorylase and results in a lack of glucose mobilization from glycogen during anaerobic exercise. A genetic defect in Merino sheep produces a similar picture. We constructed a first-generation adenoviral recombinant containing the full-length human phosphorylase cDNA under the control of the Rous sarcoma virus promoter. Primary myoblast cultures from phosphorylase-deficient human and sheep muscle were efficiently transduced with this vector, resulting in restoration of the phosphorylase activity. A similar correction of the genetic defect in muscles of McArdle's patients in vivo appears feasible, preferably with the use of an adeno-associated viral vector.


Assuntos
Técnicas de Transferência de Genes , Doença de Depósito de Glicogênio Tipo V/genética , Fosforilases/genética , Animais , Humanos , Immunoblotting , Ovinos
15.
J Neurochem ; 73(2): 605-11, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10428056

RESUMO

Oligodendrocytes (OLs) and their myelin membranes are the primary targets in the autoimmune disease multiple sclerosis (MS). The inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) has been implicated as a mediator of OL cell injury. TNF-alpha is detectable within MS lesions and induces apoptosis of mature human OLs in vitro. One possible mechanism by which TNF-alpha mediates cell death is through the activation of c-jun N-terminal kinase (JNK). We have previously shown that treatment of human OLs with TNF-alpha leads to activation of JNK. Here we provide evidence that p53, a regulator of the cell cycle and apoptosis, is a mediator of TNF-alpha-induced apoptosis of OLs. Although p53 was undetectable by western blot analysis in adult human OLs, its levels increased within 24 h after TNF-alpha treatment (100 ng/ml). The induced p53 was immunolocalized to the nucleus prior to the appearance of significant numbers of apoptotic cells. Overexpression of p53 by adenovirus-mediated gene transfer into human OLs in vitro resulted in marked apoptosis as revealed by in situ cleavage of DNA (TUNEL positive), decreased mitochondrial function, and release of lactate dehydrogenase into the culture medium. These in vitro studies demonstrate that increased p53 levels are associated with apoptosis of human OLs. The findings further implicate p53 as a target for the JNK pathway activated during TNF-alpha-mediated cell death of human adult OLs.


Assuntos
Apoptose/fisiologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Proteína Supressora de Tumor p53/genética , Adenoviridae , Infecções por Adenoviridae , Adulto , Apoptose/efeitos dos fármacos , Encéfalo/citologia , Células Cultivadas , Feto/citologia , Regulação Viral da Expressão Gênica , Técnicas de Transferência de Genes , Humanos , Marcação In Situ das Extremidades Cortadas , Óperon Lac , Oligodendroglia/virologia , Proteínas Recombinantes/genética
16.
Hippocampus ; 9(3): 201-5, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10401636

RESUMO

To investigate the possible involvement of beta-amyloid (A beta) in disrupting neuronal function during ischemia, we examined whether overexpression of C-terminal fragments of beta-amyloid precursor protein (beta-APP) in transgenic (Tg) mice is capable of altering the capacity of hippocampus slices to recover synaptic transmission after transient hypoxic episodes. Recovery of synaptic transmission was monitored in area CA1 of perfused hippocampal slices prepared from both control and Tg mice. The results obtained indicate that hippocampal slices prepared from Tg mice exhibited a much lower level of recovery in synaptic transmission following reoxygenation. This reduction in the capacity of Tg slices to recover from hypoxia-induced impairment of synaptic transmission in the hippocampus does not appear to be related to pre-existing alterations in either functional or biochemical properties of glutamate receptors in Tg mice. The present results provide the first experimental evidence that overexpression of the C-terminal fragment of APP exacerbates functional damage of hippocampal neurons after hypoxic episodes.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Hipocampo/fisiopatologia , Hipóxia Encefálica/fisiopatologia , Fragmentos de Peptídeos/biossíntese , Transmissão Sináptica/fisiologia , Precursor de Proteína beta-Amiloide/química , Animais , Potenciais Pós-Sinápticos Excitadores , Hipocampo/patologia , Hipóxia Encefálica/patologia , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Neurônios/patologia , Neurônios/fisiologia , Receptores de Glutamato/metabolismo
17.
Hum Gene Ther ; 10(8): 1299-310, 1999 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-10365661

RESUMO

Utrophin is a close homolog of dystrophin, the protein whose mutations cause Duchenne muscular dystrophy (DMD). Utrophin is present at low levels in normal and dystrophic muscle, whereas dystrophin is largely absent in DMD. In such cases, the replacement of dystrophin using a utrophin gene transfer strategy could be more advantageous because utrophin would not be a neoantigen. To establish if adenovirus (AV)-mediated utrophin gene transfer is a possible option for the treatment of DMD, an AV vector expressing a shortened version of utrophin (AdCMV-Utr) was constructed. The effect of utrophin overexpression was investigated following intramuscular injection of this AV into mdx mice, the mouse model of DMD. When the tibialis anterior (TA) muscles of 3- to 5-day-old animals were injected with 5 microl of AdCMV-Utr (7.0 x 10(11) virus/ml), an average of 32% of fibers were transduced and the transduction level remained stable for at least 60 days. The presence of utrophin restored the normal histochemical pattern of the dystrophin-associated protein complex at the cell surface and resulted in a reduction in the number of centrally nucleated fibers. The transduced fibers were largely impermeable to the tracer dye Evans blue, suggesting that utrophin protects the surface membrane from breakage. In vitro measurements of the force decline in response to high-stress eccentric contractions demonstrated that the muscles overexpressing utrophin were more resistant to mechanical stress-induced injury. Taken together, these data indicate that AV-mediated utrophin gene transfer can correct various aspects of the dystrophic phenotype. However, a progressive reduction in the number of transduced fibers was observed when the TA muscles of 30- to 45-day-old mice were injected with 25 microl of AdCMV-Utr. This reduction coincides with a humoral response to the AV and transgene, which consists of a hybrid mouse-human cDNA.


Assuntos
Adenoviridae , Proteínas do Citoesqueleto/uso terapêutico , Técnicas de Transferência de Genes , Vetores Genéticos , Proteínas de Membrana/uso terapêutico , Distrofias Musculares/terapia , Animais , Animais Recém-Nascidos , Proteínas do Citoesqueleto/genética , Expressão Gênica , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos mdx , Músculos/patologia , Distrofias Musculares/patologia , Fenótipo , Utrofina
18.
Hum Gene Ther ; 10(6): 1009-19, 1999 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-10223734

RESUMO

Skeletal muscle fibers are infected efficiently by adenoviral vectors only in neonatal animals. This lack of tropism for mature skeletal muscle may be partly due to inefficient binding of adenoviral particles to the cell surface. We evaluated in developing mouse muscle the expression levels of two high-affinity receptors for adenovirus, MHC class I and the coxsackie and adenovirus receptor (CAR). The moderate levels of MHC class I transcripts that were detected in quadriceps, gastrocnemius, and heart muscle did not vary between postnatal day 3 and day 60 adult tissue. A low level of CAR expression was detected on postnatal day 3 in quadriceps and gastrocnemius muscles, but CAR expression was barely detectable in adult skeletal muscle even by reverse transcriptase-polymerase chain reaction. In contrast, CAR transcripts were moderately abundant at all stages of heart muscle development. Ectopic expression of CAR in C2C12 mouse myoblast cells increased their transducibility by adenovirus at all multiplicities of infection (MOIs) tested as measured by lacZ reporter gene activity following AVCMVlacZ infection, with an 80-fold difference between CAR-expressing cells and control C2C12 cells at an MOI of 50. Primary myoblasts ectopically expressing CAR were injected into muscles of syngeneic hosts; following incorporation of the exogenous myoblasts into host myofibers, an increased transducibility of adult muscle fibers by AVCMVlacZ was observed in the host. Expression of the lacZ reporter gene in host myofibers coincided with CAR immunoreactivity. Furthermore, sarcolemmal CAR expression was markedly increased in regenerating muscle fibers of the dystrophic mdx mouse, fibers that are susceptible to adenovirus transduction. These analyses show that CAR expression by skeletal muscle correlates with its susceptibility to adenovirus transduction, and that forced CAR expression in mature myofibers dramatically increases their susceptibility to adenovirus transduction.


Assuntos
Adenoviridae/metabolismo , Regulação para Baixo/genética , Enterovirus/metabolismo , Técnicas de Transferência de Genes , Músculo Esquelético/citologia , Receptores Virais/metabolismo , Adenoviridae/genética , Animais , Northern Blotting , Divisão Celular , Linhagem Celular , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Imuno-Histoquímica , Camundongos , Músculo Esquelético/metabolismo , Receptores Virais/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Cancer Res ; 59(10): 2384-94, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10344748

RESUMO

Direct in vivo tumor-targeting with "suicide" viral vectors is limited by either inefficient gene transfer (i.e., retroviral vectors) or indiscriminate transfer of a conditionally toxic gene to surrounding nonmalignant tissue (i.e., adenoviral vectors). Retrovectors pseudotyped with the vesicular stomatitis virus G protein (VSVG) may serve as a remedy to this conundrum. These retroviral particles differ from standard murine retroviruses by their very broad tropism and the capacity to be concentrated by ultracentrifugation without loss of activity. We propose that a VSVG-typed retrovector can be used for efficient and tumor-specific herpes simplex virus thymidine kinase (TK) gene delivery in vivo. To test this hypothesis, we developed a bicistronic retroviral vector that expresses TK and green fluorescence protein (pTKiGFP). The 293GPG packaging cell line was used to generate vTKiGFP retroparticles. In cytotoxicity assays, vTKiGFP-transduced human glioma cell lines were sensitized to the cytotoxic effects of gangciclovir (GCV) 10,000-fold. Subsequently, virus was concentrated by ultracentrifugation to a titer of 2.3 x 10(10) cfu/ml. We tested the antitumor activity of vTKiGFP retroparticles in a rat C6 glioma model of brain cancer. Concentrated retrovector stock (9 microl volume) was injected stereotactically in preestablished intracerebral tumor. Subsequently, rats were treated with GCV for 10 days. Control rats (no GCV) had a mean survival of 38 days (range, 20-52 days). Sections performed on postmortem brain tissue revealed large tumors with evidence of high efficiency retrovector transfer and expression (as assessed by GFP fluorescence). Fluorescence was restricted to malignant tissue. In the experimental group (GCV treated), 8 of 12 remain alive and well >120 days after glioma implantation. In conclusion, vTKiGFP is very efficient at transducing human glioma cell lines in vitro and leads to significant GCV sensitization. Recombinant retroviral particles can be concentrated to titers that allow in vivo intratumoral delivery of large viral doses. The therapeutic efficiency of this reagent has been demonstrated in a preclinical model of brain cancer.


Assuntos
Antivirais/uso terapêutico , Neoplasias Encefálicas/terapia , Gammaretrovirus/genética , Ganciclovir/uso terapêutico , Terapia Genética , Vetores Genéticos/genética , Glioma/terapia , Timidina Quinase/genética , Vírus da Estomatite Vesicular Indiana/fisiologia , Proteínas não Estruturais Virais/genética , Animais , Antivirais/farmacocinética , Biotransformação , Neoplasias Encefálicas/patologia , Capsídeo , Ganciclovir/farmacocinética , Genes Reporter , Vetores Genéticos/administração & dosagem , Glioma/patologia , Proteínas de Fluorescência Verde , Humanos , Injeções Intralesionais , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Transplante de Neoplasias , Ratos , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/metabolismo , Proteínas não Estruturais Virais/metabolismo
20.
Clin Cancer Res ; 5(3): 637-42, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10100717

RESUMO

Malignant gliomas of astrocytic origin are good candidates for gene therapy because they have proven incurable with conventional treatments. Although mutation or inactivation of the p53 tumor suppressor gene occurs at early stages in gliomas and is associated with tumor progression, many tumors including high-grade glioblastoma multiforme carry a functionally intact p53 gene. To evaluate the effectiveness of p53-based therapy in glioma cells that contain endogenous wild-type p53, a clinically relevant model of malignant human glioma was established in athymic nu/nu mice. Intracerebral, rapidly growing tumors were produced by stereotactic injection of the human U87 MG glioma cell line that had been genetically modified for tracking purposes to express the Escherichia coli lacZ gene encoding beta-galactosidase. Overexpression of the p53 gene by adenovirus-mediated delivery into the tumor mass resulted in rapid cell death with the eradication of beta-galactosidase-expressing glioma cells through apoptosis. In long-term experiments, the survival of mice treated with the p53 adenoviral recombinant was significantly longer than that of mice that had received control adenoviral recombinant. During the observation period of 1 year, a complete cure was achieved in 27% of animals after a single injection of p53 adenoviral recombinant, and 38% of the animals were tumor free in the group receiving multiple injections of p53 adenoviral recombinant into a larger tumor mass. These experiments demonstrate that overexpression of p53 in gliomas, even in the presence of endogenous functional wildtype p53, leads to efficient elimination of tumor cells. These results point to the potential therapeutic usefulness of this approach for all astrocytic brain tumors.


Assuntos
Terapia Genética , Glioma/terapia , Proteína Supressora de Tumor p53/genética , Adenoviridae/genética , Animais , Progressão da Doença , Genes Supressores de Tumor , Glioma/genética , Glioma/mortalidade , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Taxa de Sobrevida , Transplante Heterólogo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA