Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Transgenic Res ; 13(6): 531-40, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15672834

RESUMO

Previous studies have shown that depletion of cardiac actin by targeted disruption is associated with increased expression of alternative actins in the mouse heart. Here we have studied the effects of transgenic overexpression of cardiac actin using the alpha-myosin heavy chain promoter. Lines carrying 7 or 8 copies of the transgene showed a 2-fold increase in cardiac actin mRNA and also displayed decreased expression of skeletal and vascular actin in their hearts. In contrast, a line with more than 250 copies of the transgene did not show a similar decrease in the expression of skeletal and vascular actin despite a 3-fold increase in cardiac actin mRNA. While the low copy number transgenic mice displayed hearts that were similar to non-transgenic controls, the high copy number transgenic line showed larger hearts with distinct atrial enlargement and cardiomyocyte hypertrophy. Further, while the low copy number transgenic mouse hearts were mildly hypocontractile when compared with non-transgenic mouse hearts, the high copy number transgenic mouse hearts were significantly so. We conclude that in the presence of a small number of copies of the cardiac actin transgene, homeostatic mechanisms involved in maintaining actin levels are active and negatively regulate skeletal and vascular actin levels in the heart in response to increased expression of cardiac actin. However, these putative mechanisms are either inoperative in the high copy number transgenic line or are countered by the enhanced expression of skeletal and vascular actin during cardiomyocyte hypertrophy.


Assuntos
Actinas/metabolismo , Cardiomegalia/etiologia , Regulação da Expressão Gênica , Miocárdio/metabolismo , Actinas/genética , Actinas/ultraestrutura , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Dosagem de Genes , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Contração Miocárdica/genética , Miocárdio/patologia , Miocárdio/ultraestrutura , Cadeias Pesadas de Miosina/genética , Tamanho do Órgão , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo
2.
Proc Natl Acad Sci U S A ; 98(24): 14114-9, 2001 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-11707570

RESUMO

Although aquaporin 5 (AQP5) is the major water channel expressed in alveolar type I cells in the lung, its actual role in the lung is a matter of considerable speculation. By using immunohistochemical staining, we show that AQP5 expression in mouse lung is not restricted to type I cells, but is also detected in alveolar type II cells, and in tracheal and bronchial epithelium. Aqp5 knockout (Aqp5(-/-)) mice were used to analyze AQP5 function in pulmonary physiology. Compared with Aqp5(+/+) mice, Aqp5(-/-) mice show a significantly increased concentration-dependent bronchoconstriction to intravenously administered Ach, as shown by an increase in total lung resistance and a decrease in dynamic lung compliance (P < 0.05). Likewise, Penh, a measure of bronchoconstriction, was significantly enhanced in Aqp5(-/-) mice challenged with aerosolized methacholine (P < 0.05). The hyperreactivity to bronchoconstriction observed in the Aqp5(-/-) mice was not due to differences in tracheal smooth muscle contractility in isolated preparations or to altered levels of surfactant protein B. These data suggest a novel pathway by which AQP5 influences bronchoconstriction. This observation is of special interest because studies to identify genetic loci involved in airway hyperresponsiveness associated with asthma bracket genetic intervals on human chromosome 12q and mouse chromosome 15, which contain the Aqp5 gene.


Assuntos
Acetilcolina/farmacologia , Aquaporinas/fisiologia , Broncoconstritores/farmacologia , Pulmão/efeitos dos fármacos , Proteínas de Membrana , Animais , Aquaporina 5 , Aquaporinas/biossíntese , Aquaporinas/genética , Broncoconstrição , Broncodilatadores/farmacologia , Feminino , Contração Isométrica , Isoproterenol/farmacologia , Pulmão/metabolismo , Pulmão/patologia , Pulmão/fisiologia , Masculino , Camundongos , Camundongos Knockout , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiologia , Tamanho do Órgão , Proteolipídeos/metabolismo , Troca Gasosa Pulmonar , Surfactantes Pulmonares/metabolismo , Traqueia/efeitos dos fármacos , Traqueia/fisiologia , Equilíbrio Hidroeletrolítico
3.
Am J Physiol Renal Physiol ; 281(4): F718-27, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11553519

RESUMO

To study the role of Na+/H+ exchanger isoform 2 (NHE2) and isoform 3 (NHE3) in sodium-fluid volume homeostasis and renal Na+ conservation, mice with Nhe2 (Nhe2-/-) and/or Nhe3 (Nhe3-/-) null mutations were fed a Na+-restricted diet, and urinary Na+ excretion, blood pressure, systemic acid-base and electrolyte status, and renal function were analyzed. Na+ -restricted Nhe2-/- mice, on either a wild-type or Nhe3 heterozygous mutant (Nhe3+/-) background, did not exhibit excess urinary Na+ excretion. After 15 days of Na+ restriction, blood pressure, fractional excretion of Na+, and the glomerular filtration rate (GFR) of Nhe2-/-Nhe3+/- mice were similar to those of Nhe2+/+ and Nhe3+/- mice, and no metabolic disturbances were observed. Nhe3-/- mice maintained on a Na+-restricted diet for 3 days exhibited hyperkalemia, urinary salt wasting, acidosis, sharply reduced blood pressure and GFR, and evidence of hypovolemic shock. These results negate the hypothesis that NHE2 plays an important renal function in sodium-fluid volume homeostasis; however, they demonstrate that NHE3 is critical for systemic electrolyte, acid-base, and fluid volume homeostasis during dietary Na+ restriction and that its absence leads to renal salt wasting.


Assuntos
Rim/fisiologia , Sódio na Dieta/farmacocinética , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Equilíbrio Hidroeletrolítico/fisiologia , Equilíbrio Ácido-Base/fisiologia , Animais , Pressão Sanguínea , Dieta Hipossódica , Ingestão de Líquidos/fisiologia , Fezes/química , Taxa de Filtração Glomerular/fisiologia , Camundongos , Camundongos Mutantes , Potássio/análise , Potássio/urina , Sódio na Dieta/análise , Sódio na Dieta/urina , Trocador 3 de Sódio-Hidrogênio , Urina
4.
Circulation ; 103(19): 2402-7, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11352891

RESUMO

BACKGROUND: The consequence of upregulation of desmin in the heart is unknown. Mutations in desmin have been linked to desmin-related myopathy (DRM), which is characterized by abnormal intrasarcoplasmic accumulation of desmin, but direct causative evidence that a desmin mutation leads to aberrant intrasarcoplasmic desmin accumulation, aggregation, and cardiomyopathy is lacking. METHODS AND RESULTS: Multiple transgenic mouse lines that expressed either murine wild-type desmin or a 7-amino acid deletion (R173 through E179) desmin (D7-des) mutation linked to DRM were made. The distribution of desmin protein was unchanged, and no overt phenotype was detected in the wild-type desmin transgenic mice. In contrast, the D7-des mouse heart showed aberrant intrasarcoplasmic and electron-dense granular filamentous aggregates that were desmin-positive and characteristic of human DRM. The desmin filament network was significantly disrupted, and myofibril alignment was visibly compromised. Although systolic function at the whole-organ level was substantially conserved in the young adult animals, the ability of the heart to respond to beta-agonist stimulation, as measured in the intact animal, was significantly blunted. CONCLUSIONS: Upregulation of desmin protein at moderate levels is not detrimental. However, the D7-des mutation is dominant negative, and expression of the mutant protein leads to the appearance of aggregates that are characteristic of and diagnostic for human desmin-related cardiomyopathy.


Assuntos
Cardiomiopatias/genética , Desmina/genética , Modelos Animais de Doenças , Sequência de Aminoácidos , Animais , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Desmina/metabolismo , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Ventrículos do Coração/ultraestrutura , Hipertrofia/genética , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica , Dados de Sequência Molecular , Mutação , Contração Miocárdica/genética
5.
Circulation ; 103(6): 889-96, 2001 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-11171800

RESUMO

BACKGROUND: Relieving the inhibition of sarcoplasmic reticular function by phospholamban is a major target of beta-adrenergic stimulation. Chronic beta-adrenergic receptor activity has been suggested to be detrimental, on the basis of transgenic overexpression of the receptor or its signaling effectors. However, it is not known whether physiological levels of sympathetic tone, in the absence of preexisting heart failure, are similarly detrimental. METHODS AND RESULTS: Transgenic mice overexpressing phospholamban at 4-fold normal levels were generated, and at 3 months, they exhibited mildly depressed ventricular contractility without heart failure. As expected, transgenic cardiomyocyte mechanics and calcium kinetics were depressed, but isoproterenol reversed the inhibitory effects of phospholamban on these parameters. In vivo cardiac function was substantially depressed by propranolol administration, suggesting enhanced sympathetic tone. Indeed, plasma norepinephrine levels and the phosphorylation status of phospholamban were elevated, reflecting increased adrenergic drive in transgenic hearts. On aging, the chronic enhancement of adrenergic tone was associated with a desensitization of adenylyl cyclase (which intensified the inhibitory effects of phospholamban), the development of overt heart failure, and a premature mortality. CONCLUSIONS: The unique interaction between phospholamban and increased adrenergic drive, elucidated herein, provides the first evidence that compensatory increases in catecholamine stimulation can, even in the absence of preexisting heart failure, be a primary causative factor in the development of cardiomyopathy and early mortality.


Assuntos
Envelhecimento , Proteínas de Ligação ao Cálcio/metabolismo , Cardiomiopatias/etiologia , Receptores Adrenérgicos beta/metabolismo , Adenilil Ciclases/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Animais , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Cardiomiopatias/sangue , Cardiomiopatias/mortalidade , Ecocardiografia , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/mortalidade , Isoproterenol/farmacologia , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Miocárdio/patologia , Norepinefrina/sangue , Fosforilação , Propranolol/farmacologia , Função Ventricular Esquerda
6.
Curr Opin Nephrol Hypertens ; 10(1): 65-9, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11195054

RESUMO

Transgenic and gene-targeting techniques have opened a new era of physiologic investigation: the field of functional genomics. The nearly exclusive use of the mouse in this discipline has necessitated the development and adaptation of sophisticated techniques for evaluating murine physiology at the cellular, tissue, organ and whole animal levels. Although many of the methodologies for exploring cardiorenal function have been successfully adapted from their use in the rat, there are important limitations and considerations that must be recognized when applying them in the mouse. Investigators have been successful in measuring a wide variety of functional variables at the whole kidney and even single nephron levels. Reviewed here are recent advances in the measurement of blood pressure, renal blood flow, whole kidney electrolyte excretion and clearance rates, single-nephron glomerular filtration rate and transport, and tubuloglomerular feedback.


Assuntos
Engenharia Genética , Rim/fisiologia , Camundongos/fisiologia , Animais , Pressão Sanguínea/fisiologia , Camundongos/genética , Perfusão , Punções
7.
Radiology ; 218(2): 471-6, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11161164

RESUMO

PURPOSE: To evaluate interventional radiologic placement of tunneled hemodialysis catheters in small thyrocervical collateral veins or in occluded veins in the neck or chest in patients with limited venous access. MATERIALS AND METHODS: A femoral venous approach was used to recanalize occluded veins or catheterize small collateral veins in 24 patients in whom all major central veins were occluded. A loop snare or catheter was used as a target for antegrade puncture. Metallic stents were deployed if necessary. Once antegrade access was secured, catheters were placed in a conventional fashion. RESULTS: Technical success was achieved in 22 (88%) of 25 procedures (one patient underwent two procedures). All catheters functioned immediately after placement. There were two procedural complications: a vasovagal episode requiring intravenously administered atropine sulfate and an episode of respiratory distress requiring intubation. There were no instances of pneumothorax, nerve injury, or bleeding complications. Catheter malfunction requiring exchange occurred at a rate of 0.67 per 100 catheter days. Infection requiring catheter removal occurred at a rate of 0.06 per 100 catheter days. Primary patency was 90% at 1 month, 71% at 6 months, and 25% at 12 months. Secondary patency was 100% at 6 months and 70% at 12 months. CONCLUSION: In patients undergoing hemodialysis in whom conventional venous access sites have been exhausted, interventional radiologic venous recanalization for the placement of permanent catheters is safe and effective. Catheters placed in recanalized veins or small collateral veins have shorter primary patency rates compared with those of conventionally placed catheters, but the former can be maintained for relatively long periods.


Assuntos
Cateterismo Periférico/métodos , Cateteres de Demora , Radiografia Intervencionista , Diálise Renal/instrumentação , Circulação Colateral , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Tempo , Grau de Desobstrução Vascular , Veias
8.
J Physiol ; 530(Pt 3): 359-66, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11158268

RESUMO

The Na+-H+ exchanger NHE3 and the thiazide-sensitive Na+-Cl- cotransporter NCC are the major apical sodium transporters in the proximal convoluted tubule and the distal convoluted tubule of the kidney, respectively. We investigated the mechanism of compensation that allows maintenance of sodium balance in NHE3 knockout mice and in NCC knockout mice. We used a so-called 'targeted proteomics' approach, which profiles the entire renal tubule with regard to changes in Na+ transporter and aquaporin abundance in response to the gene deletions. Specific antibodies to the Na+ transporters and aquaporins expressed along the nephron were utilized to determine the relative abundance of each transporter. Semiquantitative immunoblotting was used which gives an estimate of the percentage change in abundance of each transporter in knockout compared with wild-type mice. In NHE3 knockout mice three changes were identified which could compensate for the loss of NHE3-mediated sodium absorption. (a) The proximal sodium-phosphate cotransporter NaPi-2 was markedly upregulated. (b) In the collecting duct, the 70 kDa form of the y-subunit of the epithelial sodium channel, ENaC, exhibited an increase in abundance. This is thought to be an aldosterone-stimulated form of y-ENaC. (c) Glomerular filtration was significantly reduced. In the NCC knockout mice, amongst all the sodium transporters expressed along the renal tubule, only the 70 kDa form of the y-subunit of the epithelial sodium channel, ENaC, exhibited an increase in abundance. In conclusion, both mouse knockout models demonstrated successful compensation for loss of the deleted transporter. More extensive adaptation occurred in the case of the NHE3 knockout, presumably because NHE3 is responsible for much more sodium absorption in normal mice than in NCC knockout mice.


Assuntos
Proteínas de Transporte/fisiologia , Túbulos Renais/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Simportadores , Animais , Bicarbonatos/sangue , Pressão Sanguínea , Proteínas de Transporte/genética , Cloretos/sangue , Canais Epiteliais de Sódio , Taxa de Filtração Glomerular , Frequência Cardíaca , Homozigoto , Camundongos , Camundongos Knockout , Potássio/sangue , Proteoma , Sódio/sangue , Canais de Sódio/fisiologia , Simportadores de Cloreto de Sódio , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética
9.
Am J Physiol Lung Cell Mol Physiol ; 280(2): L334-41, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11159013

RESUMO

We previously reported that substance P (SP) and ATP evoke transient, epithelium-dependent relaxation of mouse tracheal smooth muscle. Since both SP and ATP are known to evoke transepithelial Cl- secretion across epithelial monolayers, we tested the hypothesis that epithelium-dependent relaxation of mouse trachea depends on Cl- channel function. In perfused mouse tracheas, the responses to SP and ATP were both inhibited by the Cl- channel inhibitors diphenylamine-2-carboxylate and 5-nitro-2-(3-phenylpropylamino)benzoate. Relaxation to ATP or SP was unaffected by 4,4'-dinitrostilbene-2,2'-disulfonic acid (DNDS), and relaxation to SP was unaffected by either DIDS or DNDS. Replacing Cl- in the buffer solutions with the impermeable anion gluconate on both sides of the trachea inhibited relaxation to SP or ATP. In contrast, increasing the gradient for Cl- secretion using Cl- free medium only in the tracheal lumen enhanced the relaxation to SP or ATP. We conclude that Cl- channel function is linked to receptor-mediated, epithelium-dependent relaxation. The finding that relaxation to SP was not blocked by DIDS suggested the involvement of a DIDS-insensitive Cl- channel, potentially the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel. To test this hypothesis, we evaluated tracheas from CFTR-deficient mice and found that the peak relaxation to SP or ATP was not significantly different from those responses in wild-type littermates. This suggests that a DIDS-insensitive Cl- channel other than CFTR is active in the SP response. This work introduces a possible role for Cl- pathways in the modulation of airway smooth muscle function and may have implications for fundamental studies of airway function as well as therapeutic approaches to pulmonary disease.


Assuntos
Broncoconstrição/fisiologia , Canais de Cloreto/metabolismo , Relaxamento Muscular/fisiologia , Mucosa Respiratória/metabolismo , Traqueia/metabolismo , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Broncoconstrição/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Gluconatos/farmacologia , Técnicas In Vitro , Contração Isométrica/efeitos dos fármacos , Contração Isométrica/fisiologia , Camundongos , Camundongos Endogâmicos CFTR , Relaxamento Muscular/efeitos dos fármacos , Nitrobenzoatos/farmacologia , Perfusão , Reprodutibilidade dos Testes , Mucosa Respiratória/efeitos dos fármacos , Estilbenos/farmacologia , Substância P/farmacologia , Traqueia/efeitos dos fármacos , ortoaminobenzoatos/farmacologia
10.
Cardiovasc Toxicol ; 1(4): 285-98, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12213967

RESUMO

Epidemiologic studies have linked dioxin exposure to increased mortality caused by ischemic heart disease. To test the hypothesis that dioxin exposure may constitute an environmental risk factor for atherosclerosis, we exposed C57BL/6J mice to 5 microg/kg of dioxin daily for 3 d, and measured various molecular and physiological markers of heart disease. Dioxin treatment led to an increase in the urinary excretion of vasoactive eicosanoids and an elevation in the mean tail-cuff blood pressure. In addition, dioxin exposure led to an increase in triglycerides, but not in high-density lipoproteins, in both Apoe(+/+) mice and in hyperlipidemic Apoe(-/- mice. Dioxin exposure also led to an increase in low-density lipoproteins in Apoe(-/-) mice. After treatment, dioxin was associated with low-density lipoprotein particles, which might serve as a vehicle to deliver the compound to atherosclerotic plaques. Dioxin treatment of vascular smooth-muscle cells taken from C57Bl/6J mice resulted in the deregulation of several genes involved in cell proliferation and apoptosis. Subchronic treatment of Apoe(-/-) mice with dioxin (150 ng/kg, three times weekly) for 7 or 26 wk caused a trend toward earlier onset and greater severity of atherosclerotic lesions compared to those of vehicle treated mice. These results suggest that dioxin may increase the incidence of ischemic heart disease by exacerbating its severity.


Assuntos
Poluentes Ambientais/toxicidade , Isquemia Miocárdica/induzido quimicamente , Dibenzodioxinas Policloradas/toxicidade , Animais , Aorta Torácica/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Arteriosclerose/induzido quimicamente , Arteriosclerose/patologia , Arteriosclerose/fisiopatologia , Pressão Sanguínea/efeitos dos fármacos , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiologia , Células Cultivadas , Colesterol/sangue , Eicosanoides/metabolismo , Lipoproteínas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/patologia , RNA/genética , RNA/isolamento & purificação , Fatores de Risco
11.
Circ Res ; 86(11): 1173-9, 2000 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-10850970

RESUMO

Protein kinase C (PKC) is a key mediator of many diverse physiological and pathological responses. Although little is known about the specific in vivo roles of the various cardiac PKC isozymes, activation-induced translocation of PKC is believed to be the primary determinant of isozyme-specific functions. Recently, we have identified a catalytically inactive peptide translocation inhibitor (epsilonV1) and translocation activator (psiepsilonRACK [receptors for activated C kinase]) specifically targeting PKCepsilon. Using cardiomyocyte-specific transgenic expression of these peptides, we combined loss- and gain-of-function approaches to elucidate the in vivo consequences of myocardial PKCepsilon signaling. As expected for a PKCepsilon RACK binding peptide, confocal microscopy showed that epsilonV1 decorated cross-striated elements and intercalated disks of cardiac myocytes. Inhibition of cardiomyocyte PKCepsilon by epsilonV1 at lower expression levels upregulated alpha-skeletal actin gene expression, increased cardiomyocyte cell size, and modestly impaired left ventricular fractional shortening. At high expression levels, epsilonV1 caused a lethal dilated cardiomyopathy. In contrast, enhancement of PKCepsilon translocation with psiepsilonRACK resulted in selectively increased beta myosin heavy chain gene expression and normally functioning concentric ventricular remodeling with decreased cardiomyocyte size. These results identify for the first time a role for PKCepsilon signaling in normal postnatal maturational myocardial development and suggest the potential for PKCepsilon activators to stimulate "physiological" cardiomyocyte growth.


Assuntos
Coração/fisiologia , Isoenzimas/fisiologia , Proteína Quinase C/fisiologia , Actinas/genética , Animais , Transporte Biológico/fisiologia , Cardiomegalia/etiologia , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Cardiomiopatia Dilatada/etiologia , Expressão Gênica/fisiologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos/genética , Contração Miocárdica/fisiologia , Miocárdio/patologia , Cadeias Pesadas de Miosina/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Quinase C-épsilon , Receptores de Quinase C Ativada , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Remodelação Ventricular/fisiologia
12.
Circulation ; 101(14): 1707-14, 2000 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-10758054

RESUMO

BACKGROUND: Transgenic cardiac beta(2)-adrenergic receptor (AR) overexpression has resulted in enhanced signaling and cardiac function in mice, whereas relatively low levels of transgenically expressed G(alphas) or beta(1)AR have resulted in phenotypes of ventricular failure. Potential relationships between the levels of betaAR overexpression and biochemical, molecular, and physiological consequences have not been reported. METHODS AND RESULTS: We generated transgenic mice expressing beta(2)AR at 3690, 7120, 9670, and 23 300 fmol/mg in the heart, representing 60, 100, 150, and 350 times background betaAR expression. All lines showed enhanced basal adenylyl cyclase activation but a decrease in forskolin- and NaF-stimulated adenylyl cyclase activities. Mice of the highest-expressing line developed a rapidly progressive fibrotic dilated cardiomyopathy and died of heart failure at 25+/-1 weeks of age. The 60-fold line exhibited enhanced basal cardiac function without increased mortality when followed for 1 year, whereas 100-fold overexpressors developed a fibrotic cardiomyopathy and heart failure, with death occurring at 41+/-1 weeks of age. Adenylyl cyclase activation did not correlate with early or delayed decompensation. Propranolol administration reduced baseline +dP/dt(max) to nontransgenic levels in all beta(2)AR transgenics except the 350-fold overexpressors, indicating that spontaneous activation of beta(2)AR was present at this level of expression. CONCLUSIONS: These data demonstrate that the heart tolerates enhanced contractile function via 60-fold beta(2)AR overexpression without detriment for a period of >/=1 year and that higher levels of expression result in either aggressive or delayed cardiomyopathy. The consequences for enhanced betaAR function in the heart appear to be highly dependent on which signaling elements are increased and to what extent.


Assuntos
Miocárdio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Animais , Canais de Cálcio/metabolismo , Canais de Cálcio/fisiologia , Baixo Débito Cardíaco/etiologia , Baixo Débito Cardíaco/mortalidade , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Cardiomiopatia Dilatada/diagnóstico por imagem , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Ecocardiografia , Condutividade Elétrica , Fibrose , Hemodinâmica , Humanos , Camundongos , Camundongos Transgênicos/genética , Contração Miocárdica/fisiologia , Miocárdio/patologia , Concentração Osmolar , Estudos Prospectivos , Fatores de Tempo
13.
Nat Genet ; 24(2): 171-4, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10655064

RESUMO

Smad proteins are intracellular mediators of signalling initiated by Tgf-betasuperfamily ligands (Tgf-betas, activins and bone morphogenetic proteins (Bmps)). Smads 1, 2, 3, 5 and 8 are activated upon phosphorylation by specific type I receptors, and associate with the common partner Smad4 to trigger transcriptional responses. The inhibitory Smads (6 and 7) are transcriptionally induced in cultured cells treated with Tgf-beta superfamily ligands, and downregulate signalling in in vitro assays. Gene disruption in mice has begun to reveal specific developmental and physiological functions of the signal-transducing Smads. Here we explore the role of an inhibitory Smad in vivo by targeted mutation of Madh6 (which encodes the Smad6 protein). Targeted insertion of a LacZ reporter demonstrated that Smad6 expression is largely restricted to the heart and blood vessels, and that Madh6 mutants have multiple cardiovascular abnormalities. Hyperplasia of the cardiac valves and outflow tract septation defects indicate a function for Smad6 in the regulation of endocardial cushion transformation. The role of Smad6 in the homeostasis of the adult cardiovascular system is indicated by the development of aortic ossification and elevated blood pressure in viable mutants. These defects highlight the importance of Smad6 in the tissue-specific modulation of Tgf-beta superfamily signalling pathways in vivo.


Assuntos
Anormalidades Cardiovasculares/genética , Sistema Cardiovascular/embriologia , Sistema Cardiovascular/crescimento & desenvolvimento , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Músculo Liso Vascular/fisiologia , Transdução de Sinais/fisiologia , Transativadores/genética , Transativadores/metabolismo , Animais , Proteínas de Ligação a DNA/deficiência , Feminino , Biblioteca Genômica , Homeostase , Homozigoto , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Músculo Liso Vascular/patologia , Mutagênese Insercional , Proteínas Recombinantes de Fusão/metabolismo , Mapeamento por Restrição , Proteína Smad6 , Transativadores/deficiência
14.
Biochemistry ; 38(50): 16706-13, 1999 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-10600134

RESUMO

The limiting element in beta-adrenergic receptor (betaAR)-G(s)-adenylyl cyclase (AC) signal transduction in the cardiomyocyte is not known, but it has been proposed that the level of adenylyl cyclase expression constrains betaAR signaling. To alter the above equilibrium, type V AC was overexpressed in a myocyte-specific manner in the hearts of transgenic mice using the alpha-myosin heavy chain promoter. Expression of type V AC was approximately 75% over endogenous levels as quantitated by [(3)H]forskolin binding. Functional activity of the transgene product was evident in cardiac membrane AC studies, where basal (45 +/- 11 vs 19 +/- 5 pmol min(-)(1) mg(-)(1)) and forskolin+Mn(2+) (695 +/- 104 vs 386 +/- 34 pmol min(-)(1) mg(-)(1)) stimulated activities were increased compared to activities in nontransgenic (NTG) littermates. However, while isoproterenol stimulated activities were higher (74 +/- 12 vs 46 +/- 9.8 pmol min(-)(1) mg(-)(1)), the fold stimulation over basal was not increased in ACV overexpressors compared to NTG (line 14.3 = 2.29 +/- 0.44-fold, line 15.1 = 1.70 +/- 0.1-fold, NTG = 2.62 +/- 0.18-fold). Similarly, in whole cell patch-clamp studies, betaAR-mediated opening of L-type Ca(2+) channels was not found to be enhanced in transgenic ACV myocytes (225 +/- 15 vs 216 +/- 10% of basal currents). Basal and isoproterenol stimulated PKA activities were elevated in the ACV mice compared to NTG, but again the extent of stimulation over basal was not enhanced. Phosphorylated phospholamban was approximately 2-fold greater in myocytes from ACV hearts compared to NTG, indicating that distal elements of the contractile cascade are activated by AC overexpression. ACV mice displayed increased heart rates and fractional shortening as assessed by echocardiography. However, in vivo hemodynamic studies revealed that heart rate and contractility responses to agonist infusion were not enhanced in ACV mice compared to NTG. We conclude that at native stoichiometries, the levels of adenylyl cyclase influence basal activities and cardiac function, but do not constrain betaAR signaling in the cardiomyocyte.


Assuntos
Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Miocárdio/enzimologia , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Adenilil Ciclases/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Western Blotting , Canais de Cálcio Tipo L/metabolismo , Catálise , Eletrocardiografia , Testes de Função Cardíaca , Frequência Cardíaca/genética , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Receptores Adrenérgicos beta/fisiologia , Transdução de Sinais/genética
15.
Proc Natl Acad Sci U S A ; 96(22): 12798-803, 1999 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-10536002

RESUMO

Brief periods of cardiac ischemia trigger protection from subsequent prolonged ischemia (preconditioning). epsilon Protein kinase C (epsilonPKC) has been suggested to mediate preconditioning. Here, we describe an epsilonPKC-selective agonist octapeptide, psiepsilon receptor for activated C-kinase (psiepsilonRACK), derived from an epsilonPKC sequence homologous to its anchoring protein, epsilonRACK. Introduction of psiepsilonRACK into isolated cardiomyocytes, or its postnatal expression as a transgene in mouse hearts, increased epsilonPKC translocation and caused cardio-protection from ischemia without any deleterious effects. Our data demonstrate that epsilonPKC activation is required for protection from ischemic insult and suggest that small molecules that mimic this epsilonPKC agonist octapeptide provide a powerful therapeutic approach to protect hearts at risk for ischemia.


Assuntos
Cardiotônicos/uso terapêutico , Isoenzimas/metabolismo , Isquemia Miocárdica/prevenção & controle , Oligopeptídeos/uso terapêutico , Proteína Quinase C/metabolismo , Receptores de Superfície Celular/uso terapêutico , Sequência de Aminoácidos , Animais , Transporte Biológico , Cardiotônicos/síntese química , Cardiotônicos/farmacologia , Morte Celular/efeitos dos fármacos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Oligopeptídeos/síntese química , Oligopeptídeos/farmacologia , Proteína Quinase C-épsilon , Ratos , Ratos Wistar , Receptores de Quinase C Ativada
16.
J Biol Chem ; 274(38): 26946-55, 1999 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-10480906

RESUMO

In chloride-secretory epithelia, the basolateral Na-K-2Cl cotransporter (NKCC1) is thought to play a major role in transepithelial Cl(-) and fluid transport. Similarly, in marginal cells of the inner ear, NKCC1 has been proposed as a component of the entry pathway for K(+) that is secreted into the endolymph, thus playing a critical role in hearing. To test these hypotheses, we generated and analyzed an NKCC1-deficient mouse. Homozygous mutant (Nkcc1(-/-)) mice exhibited growth retardation, a 28% incidence of death around the time of weaning, and mild difficulties in maintaining their balance. Mean arterial blood pressure was significantly reduced in both heterozygous and homozygous mutants, indicating an important function for NKCC1 in the maintenance of blood pressure. cAMP-induced short circuit currents, which are dependent on the CFTR Cl(-) channel, were reduced in jejunum, cecum, and trachea of Nkcc1(-/-) mice, indicating that NKCC1 contributes to cAMP-induced Cl(-) secretion. In contrast, secretion of gastric acid in adult Nkcc1(-/-) stomachs and enterotoxin-stimulated fluid secretion in the intestine of suckling Nkcc1(-/-) mice were normal. Finally, homozygous mutants were deaf, and histological analysis of the inner ear revealed a collapse of the membranous labyrinth, consistent with a critical role for NKCC1 in transepithelial K(+) movements involved in generation of the K(+)-rich endolymph and the endocochlear potential.


Assuntos
Proteínas de Transporte/fisiologia , Cloretos/metabolismo , Surdez/etiologia , Proteínas de Membrana/metabolismo , Potássio/metabolismo , Sódio/metabolismo , Animais , Animais Lactentes , Pressão Sanguínea , Proteínas de Transporte/genética , Surdez/patologia , Sistema Digestório/patologia , Células Epiteliais/metabolismo , Genótipo , Camundongos , Camundongos Mutantes , Simportadores de Cloreto de Sódio-Potássio , Taxa de Sobrevida
17.
Am J Physiol ; 277(3): F447-53, 1999 09.
Artigo em Inglês | MEDLINE | ID: mdl-10484528

RESUMO

The Na/H exchanger isoform 3 (NHE3) is expressed in the proximal tubule and thick ascending limb and contributes to the reabsorption of fluid and electrolytes in these segments. The contribution of NHE3 to fluid reabsorption was assessed by micropuncture in homozygous (Nhe3-/-) and heterozygous (Nhe3+/-) knockout mice, and in their wild-type (WT, Nhe3+/+) littermates. Arterial pressure was lower in the Nhe3-/- mice (89 +/- 6 mmHg) compared with Nhe3+/+ (118 +/- 4) and Nhe3+/- (108 +/- 5). Collections from proximal and distal tubules demonstrated that proximal fluid reabsorption was blunted in both Nhe3+/- and Nhe3-/- mice (WT, 4. 2 +/- 0.3; Nhe3+/-, 3.4 +/- 0.2; and Nhe3-/-, 2.6 +/- 0.3 nl/min; P < 0.05). However, distal delivery of fluid was not different among the three groups of mice (WT, 3.3 +/- 0.4 nl/min; Nhe3+/-, 3.3 +/- 0.2 nl/min; and Nhe3-/-, 3.0 +/- 0.4 nl/min; P < 0.05). In Nhe3-/- mice, this compensation was largely attributable to decreased single-nephron glomerular filtration rate (SNGFR): 10.7 +/- 0.9 nl/min in the Nhe3+/+ vs. 6.6 +/- 0.8 nl/min in the Nhe3-/-, measured distally. Proximal-distal SNGFR differences in Nhe3-/- mice indicated that much of the decrease in SNGFR was due to activation of tubuloglomerular feedback (TGF), and measurements of stop-flow pressure confirmed that TGF is intact in Nhe3-/- animals. In contrast to Nhe3-/- mice, normalization of early distal flow rate in Nhe3+/- mice was not related to decreased SNGFR (9.9 +/- 0.7 nl/min), but rather, to increased fluid reabsorption in the loop segment (Nhe3+/+, 2.6 +/- 0.2; Nhe3+/-, 3.6 +/- 0.5 nl/min). We conclude that NHE3 is a major Na/H exchanger isoform mediating Na+ and fluid reabsorption in the proximal tubule. In animals with NHE3 deficiency, normalization of fluid delivery to the distal tubule is achieved through alterations in filtration rate and/or downstream transport processes.


Assuntos
Néfrons/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Animais , Pressão Sanguínea , Taxa de Filtração Glomerular , Transporte de Íons , Camundongos , Camundongos Knockout , Isoformas de Proteínas/fisiologia
18.
J Clin Invest ; 104(6): 709-19, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10491406

RESUMO

In vitro, fibroblast growth factor-2 (FGF2) has been implicated in cardiomyocyte growth and reexpression of fetal contractile genes, both markers of hypertrophy. However, its in vivo role in cardiac hypertrophy during pressure overload is not well characterized. Mice with or without FGF2 (Fgf2(+/+) and Fgf2(-/-), respectively) were subjected to transverse aortic coarctation (AC). Left ventricular (LV) mass and wall thickness were assessed by echocardiography preoperatively and once a week postoperatively for 10 weeks. In vivo LV function during dobutamine stimulation, cardiomyocyte cross-sectional area, and recapitulation of fetal cardiac genes were also measured. AC Fgf2(-/-) mice develop significantly less hypertrophy (4-24% increase) compared with AC Fgf2(+/+) mice (41-52% increase). Cardiomyocyte cross-sectional area is significantly reduced in AC Fgf2(-/-) mice. Noncoarcted (NC) and AC Fgf2(-/-) mice have similar beta-adrenergic responses, but those of AC Fgf2(+/+) mice are blunted. A lack of mitotic growth in both AC Fgf2(+/+) and Fgf2(-/-) hearts indicates a hypertrophic response of cardiomyocytes. Consequently, FGF2 plays a major role in cardiac hypertrophy. Comparison of alpha- and beta-cardiac myosin heavy chain mRNA and protein levels in NC and AC Fgf2(+/+) and Fgf2(-/-) mice indicates that myosin heavy chain composition depends on hemodynamic stress rather than on FGF2 or hypertrophy, and that isoform switching is transcriptionally, not posttranscriptionally, regulated.


Assuntos
Cardiomegalia/etiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Animais , Dobutamina/farmacologia , Ecocardiografia , Feminino , Hemodinâmica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Cadeias Pesadas de Miosina/biossíntese , Cadeias Pesadas de Miosina/genética , Pressão
19.
Proc Natl Acad Sci U S A ; 96(11): 6400-5, 1999 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-10339599

RESUMO

Transgenic overexpression of Galphaq in the heart triggers events leading to a phenotype of eccentric hypertrophy, depressed ventricular function, marked expression of hypertrophy-associated genes, and depressed beta-adrenergic receptor (betaAR) function. The role of betaAR dysfunction in the development of this failure phenotype was delineated by transgenic coexpression of the carboxyl terminus of the betaAR kinase (betaARK), which acts to inhibit the kinase, or concomitant overexpression of the beta2AR at low (approximately 30-fold, Galphaq/beta2ARL), moderate (approximately 140-fold, Galphaq/beta2ARM), and high (approximately 1,000-fold, Galphaq/beta2ARH) levels above background betaAR density. Expression of the betaARK inhibitor had no effect on the phenotype, consistent with the lack of increased betaARK levels in Galphaq mice. In marked contrast, Galphaq/beta2ARL mice displayed rescue of hypertrophy and resting ventricular function and decreased cardiac expression of atrial natriuretic factor and alpha-skeletal actin mRNA. These effects occurred in the absence of any improvement in basal or agonist-stimulated adenylyl cyclase (AC) activities in crude cardiac membranes, although restoration of a compartmentalized beta2AR/AC signal cannot be excluded. Higher expression of receptors in Galphaq/beta2ARM mice resulted in salvage of AC activity, but hypertrophy, ventricular function, and expression of fetal genes were unaffected or worsened. With approximately 1,000-fold overexpression, the majority of Galphaq/beta2ARH mice died with cardiomegaly at 5 weeks. Thus, although it appears that excessive, uncontrolled, or generalized augmentation of betaAR signaling is deleterious in heart failure, selective enhancement by overexpressing the beta2AR subtype to limited levels restores not only ventricular function but also reverses cardiac hypertrophy.


Assuntos
Cardiomegalia/fisiopatologia , Proteínas de Ligação ao GTP/genética , Coração/fisiopatologia , Receptores Adrenérgicos beta 2/genética , Animais , Fator Natriurético Atrial/fisiologia , Cardiomegalia/genética , Cardiomegalia/patologia , Cruzamentos Genéticos , Ecocardiografia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Proteínas de Ligação ao GTP/fisiologia , Regulação da Expressão Gênica , Coração/fisiologia , Heterozigoto , Humanos , Camundongos , Camundongos Transgênicos , Miocárdio/patologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Adrenérgicos beta 2/fisiologia , Transdução de Sinais , Função Ventricular Esquerda
20.
Endocrinology ; 140(4): 1815-25, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10098520

RESUMO

PTH-related protein (PTHrP) and its receptor are expressed in vascular smooth muscle cells and are believed to participate in the local regulation of vascular tone. To explore the function of locally produced PTHrP in vascular smooth muscle in vivo, we developed transgenic mice that overexpress PTHrP in smooth muscle using a smooth muscle alpha-actin promoter to direct expression of the transgene. In the PTHrP-overexpressing mice, messenger RNA expression was mainly restricted to smooth muscle-containing tissues. Several founders also expressed the transgene in bone and heart and exhibited striking abnormalities in the development of these tissues. In PTHrP-overexpressing mice, blood pressure was significantly lower than that in wild-type controls (121 +/- 3 vs. 135 +/- 2 mm Hg; P < 0.01). Moreover, the magnitude of the vasorelaxant response to iv infusions of PTHrP-(1-34)NH2 was significantly attenuated in the transgenic animals. A similar desensitization to PTHrP was observed in aortic ring and portal vein preparations. Surprisingly, PTHrP-overexpressing mice were also significantly less responsive to the hypotensive action of infused acetylcholine in vivo and to the relaxant actions of acetylcholine on aortic vessel preparations in vitro. In summary, we have successfully targeted overexpression of PTHrP to the smooth muscle of transgenic mice. When expressed in its normal autocrine/paracrine setting, PTHrP lowers systemic blood pressure and decreases vascular responsiveness to further relaxation by PTHrP and other endothelium-dependent vasorelaxants such as acetylcholine. We postulate that the heterologous desensitization to acetylcholine-induced relaxation in PTHrP-overexpressing blood vessels involves desensitization of second messenger/effector signaling pathways common to PTHrP and acetylcholine.


Assuntos
Pressão Sanguínea/fisiologia , Expressão Gênica , Marcação de Genes , Contração Muscular/fisiologia , Músculo Liso Vascular/fisiologia , Proteínas/genética , Actinas/genética , Animais , Aorta/efeitos dos fármacos , Aorta/fisiologia , Feminino , Hemodinâmica , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/química , Proteína Relacionada ao Hormônio Paratireóideo , Fragmentos de Peptídeos/farmacologia , Veia Porta/efeitos dos fármacos , Veia Porta/fisiologia , Regiões Promotoras Genéticas , Proteínas/farmacologia , Proteínas/fisiologia , RNA Mensageiro/análise , Vasodilatação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...