Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Pain ; 6: 65, 2010 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-20923570

RESUMO

BACKGROUND: A chronic compressed dorsal root ganglion (CCD) in rat produces pain behavior and an enhanced excitability of neurons within the compressed ganglion. Kir2.1 is an inwardly rectifying potassium channel that acts to stabilize the resting potential of certain cell types. We hypothesized that an inducible expression of Kir2.1 channels in CCD neurons might suppress neuronal excitability in the dorsal root ganglion (DRG) and reduce the associated pain behavior. RESULTS: We delivered, by microinjection into the fourth lumbar (L4) DRG, an adenoviral vector containing a reporter gene encoding the enhanced green fluorescent protein (GFP) and a Kir2.1 channel (AdKir). At the same time the ganglion was compressed by implantation of a rod through the intervertebral foramen (CCD). The in vivo expression of the transferred gene was controlled by an ecdysone analog via an ecdysone-inducible promoter in the viral vector. In comparison with the effects of vehicle or a control vector containing only the GFP gene, AdKir significantly reduced the neuronal hyperexcitability after CCD. Electrophysiological recordings, in vivo, from nociceptive and non-nociceptive DRG neurons expressing the virally produced Kir2.1 channels revealed a hyperpolarized resting membrane potential, an increased rheobase, and lack of spontaneous activity. Inducing the Kir2.1 gene at the beginning of CCD surgery partially prevented the development of mechanical hyperalgesia. However, a delayed induction of the Kir2.1 gene (3 days after CCD surgery) produced no significant effect on the pain behavior. CONCLUSIONS: We found that an inducible expression of Kir2.1 channels in chronically compressed DRG neurons can effectively suppress the neuronal excitability and, if induced at the beginning of CCD injury, prevent the development of hyperalgesia. We hypothesize that a higher level of neuronal hyperexcitability in the DRG is required to initiate than to maintain the hyperalgesia and that the hyperexcitability contributing to neuropathic pain is best inhibited as soon as possible after injury.


Assuntos
Potenciais de Ação/fisiologia , Adenoviridae/genética , Gânglios Espinais/fisiopatologia , Vetores Genéticos/genética , Hiperalgesia/etiologia , Síndromes de Compressão Nervosa/fisiopatologia , Neurônios/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Comportamento Animal , Doença Crônica , Feminino , Regulação da Expressão Gênica , Terapia Genética , Hiperalgesia/fisiopatologia , Síndromes de Compressão Nervosa/complicações , Canais de Potássio Corretores do Fluxo de Internalização/genética , Ratos , Ratos Sprague-Dawley
2.
Cell Commun Adhes ; 15(3): 289-303, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18923946

RESUMO

Little is known about connexin expression and function in murine cardiac fibroblasts. The authors isolated native ventricular fibroblasts from adult mice and determined that although they expressed both connexin43 (Cx43) and connexin45 (Cx45), the relative abundance of Cx45 was greater than that of Cx43 in fibroblasts compared to myocytes, and the electrophoretic mobility of both Cx43 and Cx45 differed in fibroblasts and in myocytes. Increasing Cx43 expression by adenoviral infection increased intercellular coupling, whereas decreasing Cx43 expression by genetic ablation decreased coupling. Interestingly, increasing Cx43 expression reduced fibroblast proliferation, whereas decreasing Cx43 expression increased proliferation. These data demonstrate that native fibroblasts isolated from the mouse heart exhibit intercellular coupling via gap junctions containing both Cx43 and Cx45. Fibroblast proliferation is inversely related to the expression level of Cx43. Thus, connexin expression and remodeling is likely to alter fibroblast function, maintenance of the extracellular matrix, and ventricular remodeling in both normal and diseased hearts.


Assuntos
Comunicação Celular/fisiologia , Conexina 43/metabolismo , Fibroblastos/metabolismo , Miócitos Cardíacos/metabolismo , Adenoviridae/genética , Animais , Antígenos de Diferenciação/biossíntese , Comunicação Celular/genética , Proliferação de Células , Separação Celular , Células Cultivadas , Conexina 43/química , Conexina 43/genética , Conexinas/química , Conexinas/genética , Conexinas/metabolismo , Fibroblastos/citologia , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Ventrículos do Coração/citologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Fosforilação
3.
PLoS One ; 3(1): e1511, 2008 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-18231597

RESUMO

Hyperpolarization-activated, cyclic nucleotide sensitive (HCN) channels underlie the pacemaker current I(f), which plays an essential role in spontaneous cardiac activity. HCN channel subunits (HCN1-4) are believed to be modulated by additional regulatory proteins, which still have to be identified. Using biochemistry, molecularbiology and electrophysiology methods we demonstrate a protein-protein interaction between HCN2 and the K(+) channel regulator protein 1, named KCR1. In coimmunoprecipitation experiments we show that KCR1 and HCN2 proteins are able to associate. Heterologously expressed HCN2 whole-cell current density was significantly decreased by KCR1. KCR1 profoundly suppressed I(HCN2) single-channel activity, indicating a functional interaction between KCR1 and the HCN2 channel subunit. Endogenous KCR1 expression could be detected in adult and neonatal rat ventriculocytes. Adenoviral-mediated overexpression of KCR1 in rat cardiomyocytes (i) reduced I(f) whole-cell currents, (ii) suppressed most single-channel gating parameters, (iii) altered the activation kinetics, (iv) suppressed spontaneous action potential activity, and (v) the beating rate. More importantly, siRNA-based knock-down of endogenous KCR1 increased the native I(f) current size and single-channel activity and accelerated spontaneous beating rate, supporting an inhibitory action of endogenous KCR1 on native I(f). Our observations demonstrate for the first time that KCR1 modulates I(HCN2)/I(f) channel gating and indicate that KCR1 serves as a regulator of cardiac automaticity.


Assuntos
Relógios Biológicos , Contração Miocárdica/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio/fisiologia , Adenoviridae/genética , Animais , Nó Atrioventricular/metabolismo , Células Cultivadas , Primers do DNA , Imunoprecipitação , Ativação do Canal Iônico , Proteínas do Tecido Nervoso/genética , Canais de Potássio/genética , RNA Interferente Pequeno , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
4.
Mol Ther ; 15(8): 1495-503, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17565351

RESUMO

Conditionally replication competent adenoviruses (CRAds) represent one of the most intensely studied gene therapy strategies for a variety of malignancies, including prostate cancer. These viruses can be generated by placing a tissue or cancer-specific promoter upstream of one or more of the viral genes required for replication (e.g., E1A, E1B). We report here that E1A inhibits androgen receptor (AR) target gene induction and, correspondingly, activated AR inhibits adenoviral replication. This mutual inhibition appears to be an indirect effect, possibly through competition for shared transcriptional co-activators. The net effect is that the oncolytic effect of prostate-specific CRAds is attenuated by these interactions. Fusion of the E1A to AR ameliorates this inhibition, while enhancing specificity. These findings have significant implications in the development of prostate-specific CRAd therapies.


Assuntos
Adenoviridae/fisiologia , Receptores Androgênicos/metabolismo , Replicação Viral , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Animais , Linhagem Celular Tumoral , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos Nus , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Próstata/metabolismo , Receptores Androgênicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Ann Neurol ; 60(1): 118-27, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16729292

RESUMO

OBJECTIVE: Potassium (K(+)) channels on immune cells have gained attention recently as promising targets of therapy for immune-mediated neurological diseases such as multiple sclerosis (MS). We examined K(+) channels on dendritic cells (DCs), which infiltrate the brain in MS and may impact disease course. METHODS: We identified K(+) channels on blood-derived DCs by whole-cell patch-clamp analysis, confirmed by immunofluorescent staining. We also stained K(+) channels in brain sections from MS patients and control subjects. To test functionality, we blocked K(v)1.3 and K(v)1.5 in stimulated DCs with pharmacological blockers or with an inducible dominant-negative K(v)1.x adenovirus construct and analyzed changes in costimulatory molecule upregulation. RESULTS: Electrophysiological analysis of DCs showed an inward-rectifying K(+) current early after stimulation, replaced by a mix of voltage-gated K(v)1.3- and K(v)1.5-like channels at later stages of maturation. K(v)1.3 and K(v)1.5 were also highly expressed on DCs infiltrating MS brain tissue. Of note, we found that CD83, CD80, CD86, CD40, and interleukin-12 upregulation were significantly impaired on K(v)1.3 and K(v)1.5 blockade. INTERPRETATION: These data support a functional role of K(v)1.5 and K(v)1.3 on activated human DCs and further define the mechanisms by which K(+) channel blockade may act to suppress immune-mediated neurological diseases.


Assuntos
Encéfalo/imunologia , Doenças do Sistema Nervoso Central/imunologia , Células Dendríticas/fisiologia , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/metabolismo , Adenoviridae/genética , Adulto , Idoso , Encéfalo/citologia , Doenças do Sistema Nervoso Central/patologia , Células Dendríticas/efeitos dos fármacos , Feminino , Imunofluorescência , Genes Dominantes , Humanos , Imuno-Histoquímica , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/genética , Lipopolissacarídeos/farmacologia , Masculino , Pessoa de Meia-Idade , Mutagênese , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Transfecção
6.
Circ Res ; 98(2): e11-8, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16397147

RESUMO

Beta-adrenoceptor stimulation robustly increases cardiac L-type Ca2+ current (ICaL); yet the molecular mechanism of this effect is still not well understood. Previous reports have shown in vitro phosphorylation of a consensus protein kinase A site at serine 1928 on the carboxyl terminus of the alpha1C subunit; however, the functional role of this site has not been investigated in cardiac myocytes. Here, we examine the effects of truncating the distal carboxyl terminus of the alpha1C subunit at amino acid residue 1905 or mutating the putative protein kinase A site at serine 1928 to alanine in adult guinea pig myocytes, using novel dihydropyridine-insensitive alpha1C adenoviruses, coexpressed with beta2 subunits. Expression of alpha1C truncated at 1905 dramatically attenuated the increase of peak ICaL induced by isoproterenol. However, the point mutation S1928A did not significantly attenuate the beta-adrenergic response. The findings indicate that the distal carboxyl-terminus of alpha1C plays an important role in beta-adrenergic upregulation of cardiac L-type Ca2+ channels, but that phosphorylation of serine 1928 is not required for this effect.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/química , Receptores Adrenérgicos beta/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Cobaias , Isoproterenol/farmacologia , Subunidades Proteicas , Serina
7.
Biochem Biophys Res Commun ; 329(2): 749-54, 2005 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-15737650

RESUMO

The alpha(1c) subunit of the cardiac L-type Ca(2+) channel, which contains the channel pore, voltage- and Ca(2+)-dependent gating structures, and drug binding sites, has been well studied in heterologous expression systems, but many aspects of L-type Ca(2+) channel behavior in intact cardiomyocytes remain poorly characterized. Here, we develop adenoviral constructs with E1, E3 and fiber gene deletions, to allow incorporation of full-length alpha(1c) gene cassettes into the adenovirus backbone. Wild-type (alpha(1c-wt)) and mutant (alpha(1c-D-)) Ca(2+) channel adenoviruses were constructed. The alpha(1c-D-) contained four point substitutions at amino acid residues known to be critical for dihydropyridine binding. Both alpha(1c-wt) and alpha(1c-D-) expressed robustly in A549 cells (peak L-type Ca(2+) current (I(CaL)) at 0 mV: alpha(1c-wt) -9.94+/-1.00pA/pF, n=9; alpha(1c-D-) -10.30pA/pF, n=12). I(CaL) carried by alpha(1c-D-) was markedly less sensitive to nitrendipine (IC(50) 17.1 microM) than alpha(1c-wt) (IC(50) 88 nM); a feature exploited to discriminate between engineered and native currents in transduced guinea-pig myocytes. 10 microM nitrendipine blocked only 51+/-5% (n=9) of I(CaL) in alpha(1c-D-)-expressing myocytes, in comparison to 86+/-8% (n=9) of I(CaL) in control myocytes. Moreover, in 20 microM nitrendipine, calcium transients could still be evoked in alpha(1c-D-)-transduced cells, but were largely blocked in control myocytes, indicating that the engineered channels were coupled to sarcoplasmic reticular Ca(2+) release. These alpha(1c) adenoviruses provide an unprecedented tool for structure-function studies of cardiac excitation-contraction coupling and L-type Ca(2+) channel regulation in the native myocyte background.


Assuntos
Adenoviridae/genética , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/fisiologia , Engenharia de Proteínas/métodos , Substituição de Aminoácidos , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Cobaias , Mutagênese Sítio-Dirigida , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Coelhos , Proteínas Recombinantes/metabolismo
8.
Blood ; 105(10): 3910-7, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15677570

RESUMO

Inflammation and thrombosis are increasingly recognized as interrelated biologic processes. Endothelial cell expression of thrombomodulin (TM), a key component of the anticoagulant protein C pathway, is potently inhibited by inflammatory cytokines. Because the mechanism underlying this effect is largely unknown, we investigated a potential role for the inflammatory transcription factor nuclear factor-kappa B (NF-kappaB). Blocking NF-kappaB activation effectively prevented cytokine-induced down-regulation of TM, both in vitro and in a mouse model of tumor necrosis factor-alpha (TNF-alpha)-mediated lung injury. Although the TM promoter lacks a classic NF-kappaB consensus site, it does contain tandem Ets transcription factor binding sites previously shown to be important for both constitutive TM gene expression and cytokine-induced repression. Using electrophoretic mobility shift assay and chromatin immunoprecipitation, we found that multiple Ets species bind to the TNF-alpha response element within the TM promoter. Although cytokine exposure did not alter Ets factor binding, it did reduce binding of p300, a coactivator required by Ets for full transcriptional activity. Overexpression of p300 also prevented TM repression by cytokines. We conclude that NF-kappaB is a critical mediator of TM repression by cytokines. Further evidence suggests a mechanism involving competition by NF-kappaB for limited pools of the transcriptional coactivator p300 necessary for TM gene expression.


Assuntos
Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Mediadores da Inflamação/farmacologia , Interleucina-1/farmacologia , NF-kappa B/metabolismo , Trombomodulina/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células Cultivadas , Proteína p300 Associada a E1A , Células Endoteliais/metabolismo , Humanos , Masculino , Camundongos , NF-kappa B/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ets , Trombomodulina/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
9.
Nucleic Acids Res ; 32(8): 2652-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15141036

RESUMO

The human adenovirus E4 ORF 6 34 kDa oncoprotein (E4 34k), in concert with the 55 kDa product of E1b, prevents concatenation of viral genomes in infected cells, inhibits the repair of double strand breaks (DSBs) in the viral genome, and inhibits V(D)J recombination in a plasmid transfection assay. These activities are consistent with a general inhibition by the E4 34k and E1b 55k proteins of DSB repair by non-homologous end joining (NHEJ) on extrachromosomal substrates. To determine whether inhibition of NHEJ extends to repair of DSBs in the cell chromosome, we have examined the effects of E4 34k on repair of chromosomal DSBs induced by ionizing radiation in a cell line in which E4 34k expression and biological activity is inducible and E1b 55k is produced constitutively. We demonstrate that in this cell line, induction of E4 34k inhibits chromosomal DSB repair. Recently, it has been shown that in infected cells, E4 34k and the adenovirus E1b 55k proteins cooperate to destabilize Mre11 and Rad50, components of mammalian NHEJ systems. Consistent with this, induction of expression of E4 34k in the inducible cell line also reduces the steady state level of Mre11 protein.


Assuntos
Proteínas E4 de Adenovirus/metabolismo , Quebra Cromossômica , Reparo do DNA , Proteínas E4 de Adenovirus/genética , Ciclo Celular , Divisão Celular , Linhagem Celular , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Genoma Humano , Humanos , Proteína Homóloga a MRE11
10.
J Pain ; 5(2): 104-10, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15042518

RESUMO

UNLABELLED: Bone is a common metastatic site for prostate and breast cancer, and bone cancer is usually associated with severe pain. Traditional treatments for cancer pain can sometimes be ineffective or associated with side effects. Thus an increasing number of patients seek alternative therapies. In this study we investigated the analgesic effects of a soy diet on 3 experimental models of bone cancer pain. Mice were fed a diet in which the protein source was either soy or casein. After 1 week on the diet, sarcoma cells (NCTC 2472) were injected into the medullary cavity of the humeri, femur, or calcaneus. Experimenters blinded to diet of the animal assessed the pain behavior in these animals, forelimb grip force in the humerus model and paw withdrawal frequency to mechanical stimuli in the calcaneus and femur models. The effect of morphine on cancer-induced pain behavior was investigated in calcaneus and femur models. In addition, in the femur model, the effects of soy on tumor size and bone destruction were studied. The soy diet reduced secondary mechanical hyperalgesia in the femur model but had no effect on primary mechanical hyperalgesia in the calcaneus model or on movement-related hyperalgesia in the humerus model. No dietary impact was discerned in measurements of tumor size, bone destruction, and body weight in the femur model, suggesting that the soy diet had no effect on cancer growth. Morphine dose-dependently reduced hyperalgesia with no diet-based difference. These results suggest that a soy diet might provide analgesia in certain forms of hyperalgesia associated with bone cancer. PERSPECTIVE: The study raises the possibility of dietary supplements influencing aspects of cancer pain. Further research will help determine if use of nutritional supplements, such as soy proteins, can reduce opioid analgesic use in chronic pain states and help minimize the side effects associated with long term use of opioids.


Assuntos
Analgésicos/farmacologia , Neoplasias Ósseas/complicações , Dor/dietoterapia , Sarcoma/complicações , Proteínas de Soja/farmacologia , Analgésicos Opioides/farmacologia , Ração Animal , Animais , Peso Corporal , Neoplasias Ósseas/patologia , Calcâneo/patologia , Caseínas/farmacologia , Doença Crônica , Modelos Animais de Doenças , Fêmur/patologia , Úmero/patologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Morfina/farmacologia , Atividade Motora , Dor/tratamento farmacológico , Dor/etiologia , Sarcoma/patologia
11.
Am J Physiol Heart Circ Physiol ; 286(6): H2342-51, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14962839

RESUMO

Suppression of electrical alternans may be antiarrhythmic. Our previous computer simulations have suggested that increasing the rapid component of the delayed rectifier K(+) current (I(Kr)) suppresses alternans. To test this hypothesis, I(Kr) in isolated canine ventricular myocytes was increased by infection with an adenovirus containing the gene for the pore-forming domain of I(Kr) [human ether-a-go-go gene (HERG)]. With the use of the perforated or whole cell patch-clamp technique, action potentials recorded at different pacing cycle lengths (CLs) were applied to the myocytes as the command waveforms. HERG infection markedly increased peak I(Kr) during the action potential (from 0.54 +/- 0.03 pA/pF in control to 3.60 +/- 0.81 pA/pF). Rate-dependent alterations of peak I(Kr) were similar for freshly isolated myocytes and HERG-infected myocytes. In both cell types, I(Kr) increased when CL decreased from 1,000 to 500 ms and then decreased progressively as CL decreased further. During alternans at CL = 170 ms, peak I(Kr) was larger for the short than for the long action potential for both groups, but the difference in peak I(Kr) was larger for HERG-infected myocytes. The voltage at which peak I(Kr) occurred was significantly less negative in HERG-infected myocytes, in association with shifts of the steady-state voltage-dependent activation and inactivation curves to less negative potentials. Pacing at short CL induced stable alternans in freshly isolated myocytes and in cultured myocytes without HERG infection, but not in HERG-infected myocytes. These data support the idea that increasing I(Kr) may be a viable approach to suppressing electrical alternans.


Assuntos
Potenciais de Ação/fisiologia , Proteínas de Transporte de Cátions/genética , Modelos Biológicos , Miócitos Cardíacos/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Taquicardia Ventricular/fisiopatologia , Adenoviridae/genética , Animais , Simulação por Computador , Cães , Canais de Potássio Éter-A-Go-Go , Feminino , Expressão Gênica , Terapia Genética/métodos , Humanos , Masculino , Taquicardia Ventricular/terapia
12.
Mol Cell Biol ; 23(17): 6159-73, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12917338

RESUMO

The cyclin D1 gene is overexpressed in human breast cancers and is required for oncogene-induced tumorigenesis. Peroxisome proliferator-activated receptor gamma (PPAR gamma) is a nuclear receptor selectively activated by ligands of the thiazolidinedione class. PPAR gamma induces hepatic steatosis, and liganded PPAR gamma promotes adipocyte differentiation. Herein, cyclin D1 inhibited ligand-induced PPAR gamma function, transactivation, expression, and promoter activity. PPAR gamma transactivation induced by the ligand BRL49653 was inhibited by cyclin D1 through a pRB- and cdk-independent mechanism, requiring a region predicted to form an helix-loop-helix (HLH) structure. The cyclin D1 HLH region was also required for repression of the PPAR gamma ligand-binding domain linked to a heterologous DNA binding domain. Adipocyte differentiation by PPAR gamma-specific ligands (BRL49653, troglitazone) was enhanced in cyclin D1(-/-) fibroblasts and reversed by retroviral expression of cyclin D1. Homozygous deletion of the cyclin D1 gene, enhanced expression by PPAR gamma ligands of PPAR gamma and PPAR gamma-responsive genes, and cyclin D1(-/-) mice exhibit hepatic steatosis. Finally, reduction of cyclin D1 abundance in vivo using ponasterone-inducible cyclin D1 antisense transgenic mice, increased expression of PPAR gamma in vivo. The inhibition of PPAR gamma function by cyclin D1 is a new mechanism of signal transduction cross talk between PPAR gamma ligands and mitogenic signals that induce cyclin D1.


Assuntos
Neoplasias da Mama/metabolismo , Ciclina D1/metabolismo , Regulação da Expressão Gênica , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Tiazolidinedionas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células 3T3 , Animais , Mama/citologia , Mama/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Ciclina D1/química , Ciclina D1/efeitos dos fármacos , Ciclina D1/genética , Ecdisterona/análogos & derivados , Ecdisterona/farmacologia , Células Epiteliais/metabolismo , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Feminino , Humanos , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Modelos Moleculares , Mutação , Conformação Proteica , Valores de Referência , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Rosiglitazona , Tiazóis/farmacologia , Ativação Transcricional
13.
J Physiol ; 541(Pt 2): 435-52, 2002 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12042350

RESUMO

Recombinant adenoviruses were used to overexpress green fluorescent protein (GFP)-fused auxiliary Ca(2+) channel beta subunits (beta(1)-beta(4)) in cultured adult rat heart cells, to explore new dimensions of beta subunit functions in vivo. Distinct beta-GFP subunits distributed differentially between the surface sarcolemma, transverse elements, and nucleus in single heart cells. All beta-GFP subunits increased the native cardiac whole-cell L-type Ca(2+) channel current density, but produced distinctive effects on channel inactivation kinetics. The degree of enhancement of whole-cell current density was non-uniform between beta subunits, with a rank order of potency beta(2a) approximately equal to beta(4) > beta(1b) > beta(3). For each beta subunit, the increase in L-type current density was accompanied by a correlative increase in the maximal gating charge (Q(max)) moved with depolarization. However, beta subunits produced characteristic effects on single L-type channel gating, resulting in divergent effects on channel open probability (P(o)). Quantitative analysis and modelling of single-channel data provided a kinetic signature for each channel type. Spurred on by ambiguities regarding the molecular identity of the actual endogenous cardiac L-type channel beta subunit, we cloned a new rat beta(2) splice variant, beta(2b), from heart using 5' rapid amplification of cDNA ends (RACE) PCR. By contrast with beta(2a), expression of beta(2b) in heart cells yielded channels with a microscopic gating signature virtually identical to that of native unmodified channels. Our results provide novel insights into beta subunit functions that are unattainable in traditional heterologous expression studies, and also provide new perspectives on the molecular identity of the beta subunit component of cardiac L-type Ca(2+) channels. Overall, the work establishes a powerful experimental paradigm to explore novel functions of ion channel subunits in their native environments.


Assuntos
Canais de Cálcio/metabolismo , Miocárdio/metabolismo , Adenoviridae/metabolismo , Animais , Western Blotting , Canais de Cálcio/biossíntese , Canais de Cálcio Tipo L/efeitos dos fármacos , Primers do DNA , Eletrofisiologia , Vetores Genéticos , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Modelos Neurológicos , Miocárdio/química , Técnicas de Patch-Clamp , Plasmídeos , Ratos , Proteínas Recombinantes/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcolema/efeitos dos fármacos , Sarcolema/metabolismo , Transfecção
14.
J Biol Chem ; 277(29): 26436-43, 2002 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-11994284

RESUMO

Voltage-gated K(+) channels are multimeric proteins, consisting of four pore-forming alpha-subunits alone or in association with accessory subunits. Recently, for example, it was shown that the accessory Kv channel interacting proteins form complexes with Kv4 alpha-subunits and modulate Kv4 channel activity. The experiments reported here demonstrate that the neuronal calcium sensor protein-1 (NCS-1), another member of the recoverin-neuronal calcium sensor superfamily, is expressed in adult mouse ventricles and that NCS-1 co-immunoprecipitates with Kv4.3 from (adult mouse) ventricular extracts. In addition, co-expression studies in HEK-293 cells reveal that NCS-1 increases membrane expression of Kv4 alpha-subunits and functional Kv4-encoded K(+) current densities. Co-expression of NCS-1 also decreases the rate of inactivation of Kv4 alpha-subunit-encoded K(+) currents. In contrast to the pronounced effects of Kv channel interacting proteins on Kv4 channel gating, however, NCS-1 co-expression does not measurably affect the voltage dependence of steady-state inactivation or the rate of recovery from inactivation of Kv4-encoded K(+) currents. Taken together, these results suggest that NCS-1 is an accessory subunit of Kv4-encoded I(to,f) channels that functions to regulate I(to,f) density in the mammalian myocardium.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Miocárdio/metabolismo , Neuropeptídeos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Adenoviridae , Animais , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Eletrofisiologia , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Sensoras de Cálcio Neuronal , Canais de Potássio Shal
15.
Stroke ; 33(4): 1101-6, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11935067

RESUMO

BACKGROUND AND PURPOSE: Poly(ADP-ribose) polymerase (PARP-1; Enzyme Commission 2.4.30) is a nuclear DNA repair enzyme that mediates early neuronal ischemic injury. Using novel 3-dimensional, fast spin-echo-based diffusion-weighted imaging, we compared acute (21 hours) and long-term (3 days) ischemic volume after middle cerebral artery (MCA) occlusion in PARP-1-null mutants (PARP-/-) versus genetically matched wild-type mice (WT mice). PARP-/- mice were also treated with viral transfection of wild-type PARP-1 to determine whether protection from MCA occlusion is lost with restoration of the gene product. METHODS: Halothane-anesthetized mice were treated with reversible MCA occlusion via intraluminal suture technique. Ischemic volumes were delineated by diffusion-weighted imaging with high spatial and temporal resolution during MCA occlusion and reperfusion. Recombinant Sindbis virus carrying beta-galactosidase (lacZ) or PARP-1 was injected into ipsilateral striatum, then animals underwent MCA occlusion 3 days later. Infarction volume was measured at 22 hours of reperfusion (2,3,5-triphenyltetrazolium chloride histology). RESULTS: Reduction in regional water apparent diffusion coefficient (ADC) during occlusion or secondary ADC decline during reperfusion was not different between groups. Ischemic volume was smaller early in occlusion in PARP-/- versus WT mice and remained less at 21 hours of reperfusion. Ischemic volume then increased from 1 to 2 days in all mice, then stabilized without further change. Ischemic damage was smaller in PARP-/- than in WT mice at 3 days. Transfection of PARP-1 into PARP-/- mice increased stroke damage relative to lacZ-injected PARP-/- and increased damage to that of the WT mice. Intraischemic laser-Doppler flowmetry and physiological variables were not different among groups. CONCLUSIONS: PARP-1 deficiency provides both early and prolonged protection from experimental focal stroke. The mechanism is not linked to preservation of ADC and mitigation of secondary energy depletion during early reperfusion.


Assuntos
Infarto Cerebral/fisiopatologia , Infarto da Artéria Cerebral Média/fisiopatologia , Poli(ADP-Ribose) Polimerases/farmacologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/fisiopatologia , Animais , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Infarto Cerebral/etiologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Difusão , Modelos Animais de Doenças , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Homozigoto , Infarto da Artéria Cerebral Média/complicações , Fluxometria por Laser-Doppler , Imageamento por Ressonância Magnética/métodos , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Poli(ADP-Ribose) Polimerases/deficiência , Poli(ADP-Ribose) Polimerases/genética , Recuperação de Função Fisiológica/genética , Reperfusão , Sindbis virus/genética , Acidente Vascular Cerebral/etiologia , Tempo , Transfecção , Água/metabolismo
16.
Circ Res ; 90(5): 586-93, 2002 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-11909823

RESUMO

Previous studies have demonstrated a role for Kv4 alpha subunits in the generation of the fast transient outward K+ current, I(to,f), in the mammalian myocardium. The experiments here were undertaken to explore the role of homomeric/heteromeric assembly of Kv4.2 and Kv4.3 and of the Kv channel accessory subunit, KChIP2, in the generation of mouse ventricular I(to,f). Western blots reveal that the expression of Kv4.2 parallels the regional heterogeneity in I(to,f) density, whereas Kv4.3 and KChIP2 are uniformly expressed in adult mouse ventricles. Antisense oligodeoxynucleotides (AsODNs) targeted against Kv4.2 or Kv4.3 selectively attenuate I(to,f) in mouse ventricular cells. Adenoviral-mediated coexpression of Kv4.2 and Kv4.3 in HEK-293 cells and in mouse ventricular myocytes produces transient outward K+ currents with properties distinct from those produced on expression of Kv4.2 or Kv4.3 alone, and the gating properties of the heteromeric Kv4.2/Kv4.3 channels in ventricular cells are more similar to native I(to,f) than are the homomeric Kv4.2 or Kv4.3 channels. Biochemical studies reveal that Kv4.2, Kv4.3, and KChIP2 coimmunoprecipitate from adult mouse ventricles. In addition, most of the Kv4.2 and KChIP2 are associated with Kv4.3 in situ. Taken together, these results demonstrate that functional mouse ventricular I(to,f) channels are heteromeric, comprising Kv4.2/Kv4.3 alpha subunits and KChIP2. The results here also suggest that Kv4.2 is the primary determinant of the regional heterogeneity in I(to,f) expression in adult mouse ventricle.


Assuntos
Ventrículos do Coração/metabolismo , Miocárdio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Potássio/metabolismo , Subunidades Proteicas , Animais , Western Blotting , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Técnicas de Cultura de Células , Ventrículos do Coração/citologia , Ativação do Canal Iônico/efeitos dos fármacos , Proteínas Interatuantes com Canais de Kv , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Miocárdio/citologia , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio , Canais de Potássio/genética , Testes de Precipitina , Estrutura Quaternária de Proteína/fisiologia , Canais de Potássio Shal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA