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.
Toxicol Pathol ; 46(8): 991-998, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30392455

RESUMO

The ReninAAV db/db uNx model of diabetic kidney disease (DKD) exhibits hallmarks of advanced human disease, including progressive elevations in albuminuria and serum creatinine, loss of glomerular filtration rate, and pathological changes. Microarray analysis of renal transcriptome changes were more similar to human DKD when compared to db/db eNOS-/- model. The model responds to treatment with arterial pressure lowering (lisinopril) or glycemic control (rosiglitazone) at early stages of disease. We hypothesized the ReninAAV db/db uNx model with advanced disease would have residual disease after treatment with lisinopril, rosiglitazone, or combination of both. To test this, ReninAAV db/db uNx mice with advanced disease were treated with lisinopril, rosiglitazone, or combination of both for 10 weeks. All treatment groups showed significant lowering of urinary albumin to creatinine ratio compared to baseline; however, only combination group exhibited lowering of serum creatinine. Treatment improved renal pathological scores compared to baseline values with residual disease evident in all treatment groups when compared to db/m controls. Gene expression analysis by TaqMan supported pathological changes with increased fibrotic and inflammatory markers. The results further validate this model of DKD in which residual disease is present when treated with agents to lower arterial pressure and glycemic control.


Assuntos
Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Modelos Animais de Doenças , Animais , Humanos , Camundongos , Camundongos Endogâmicos , Transcriptoma
2.
Toxicol Pathol ; 46(8): 970-975, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30213245

RESUMO

Diabetes mellitus (types 1 and 2) is the leading cause of glomerular disease and end-stage renal disease in most developed countries, with estimates that one-third of people living with diabetes will develop diabetic kidney disease (DKD). The current standard of care medications slow but do not arrest progression of kidney disease, and therefore, therapy for DKD is a highly unmet medical need for patients. To discover and test novel and durable new therapies, it is necessary to develop animal models of human DKD, which authentically recapitulate the human disease state and provide translatable efficacy to human patients. Here, we review selected mouse models of human DKD, which demonstrate many of the features of type 2 human DKD.


Assuntos
Nefropatias Diabéticas , Modelos Animais de Doenças , Animais , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 2/complicações , Humanos , Camundongos
4.
Mol Metab ; 8: 1-12, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29289646

RESUMO

OBJECTIVES: The autonomic nervous system is critically involved in mediating the control by leptin of many physiological processes. Here, we examined the role of the leptin receptor (LepR) in proopiomelanocortin (POMC) and agouti-related peptide (AgRP) neurons in mediating the effects of leptin on regional sympathetic and parasympathetic nerve activity. METHODS: We analyzed how deletion of the LepR in POMC neurons (POMCCre/LepRfl/fl mice) or AgRP neurons (AgRPCre/LepRfl/fl mice) affects the ability of leptin to increase sympathetic and parasympathetic nerve activity. We also studied mice lacking the catalytic p110α or p110ß subunits of phosphatidylinositol-3 kinase (PI3K) in POMC neurons. RESULTS: Leptin-evoked increase in sympathetic nerve activity subserving thermogenic brown adipose tissue was partially blunted in mice lacking the LepR in either POMC or AgRP neurons. On the other hand, loss of the LepR in AgRP, but not POMC, neurons interfered with leptin-induced sympathetic nerve activation to the inguinal fat depot. The increase in hepatic sympathetic traffic induced by leptin was also reduced in mice lacking the LepR in AgRP, but not POMC, neurons whereas LepR deletion in either AgRP or POMC neurons attenuated the hepatic parasympathetic nerve activation evoked by leptin. Interestingly, the renal, lumbar and splanchnic sympathetic nerve activation caused by leptin were significantly blunted in POMCCre/LepRfl/fl mice, but not in AgRPCre/LepRfl/fl mice. However, loss of the LepR in POMC or AgRP neurons did not interfere with the ability of leptin to increase sympathetic traffic to the adrenal gland. Furthermore, ablation of the p110α, but not the p110ß, isoform of PI3K from POMC neurons eliminated the leptin-elicited renal sympathetic nerve activation. Finally, we show trans-synaptic retrograde tracing of both POMC and AgRP neurons from the kidneys. CONCLUSIONS: POMC and AgRP neurons are differentially involved in mediating the effects of leptin on autonomic nerve activity subserving various tissues and organs.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Sistema Nervoso Autônomo/fisiologia , Leptina/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Tecido Adiposo Marrom/metabolismo , Proteína Relacionada com Agouti/genética , Animais , Sistema Nervoso Autônomo/citologia , Sistema Nervoso Autônomo/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Pró-Opiomelanocortina/genética , Receptores para Leptina/genética , Receptores para Leptina/metabolismo
5.
J Am Soc Nephrol ; 29(2): 477-491, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29061652

RESUMO

Progress in research and developing therapeutics to prevent diabetic kidney disease (DKD) is limited by a lack of animal models exhibiting progressive kidney disease. Chronic hypertension, a driving factor of disease progression in human patients, is lacking in most available models of diabetes. We hypothesized that superimposition of hypertension on diabetic mouse models would accelerate DKD. To test this possibility, we induced persistent hypertension in three mouse models of type 1 diabetes and two models of type 2 diabetes by adeno-associated virus delivery of renin (ReninAAV). Compared with LacZAAV-treated counterparts, ReninAAV-treated type 1 diabetic Akita/129 mice exhibited a substantial increase in albumin-to-creatinine ratio (ACR) and serum creatinine level and more severe renal lesions. In type 2 models of diabetes (C57BKLS db/db and BTBR ob/ob mice), compared with LacZAAV, ReninAAV induced significant elevations in ACR and increased the incidence and severity of histopathologic findings, with increased serum creatinine detected only in the ReninAAV-treated db/db mice. The uninephrectomized ReninAAV db/db model was the most progressive model examined and further characterized. In this model, separate treatment of hyperglycemia with rosiglitazone or hypertension with lisinopril partially reduced ACR, consistent with independent contributions of these disorders to renal disease. Microarray analysis and comparison with human DKD showed common pathways affected in human disease and this model. These results identify novel models of progressive DKD that provide researchers with a facile and reliable method to study disease pathogenesis and support the development of therapeutics.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/etiologia , Modelos Animais de Doenças , Hipertensão/complicações , Renina/genética , Animais , Anti-Hipertensivos/uso terapêutico , Creatinina/sangue , Dependovirus , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/patologia , Progressão da Doença , Feminino , Vetores Genéticos , Hipertensão/tratamento farmacológico , Hipertensão/genética , Hipoglicemiantes/uso terapêutico , Janus Quinases/metabolismo , Óperon Lac/genética , Lisinopril/uso terapêutico , Masculino , Camundongos , Nefrectomia , Óxido Nítrico Sintase Tipo III/genética , Rosiglitazona/uso terapêutico , Fatores de Transcrição STAT/metabolismo , Albumina Sérica/metabolismo , Índice de Gravidade de Doença , Transdução de Sinais
6.
Am J Physiol Renal Physiol ; 312(6): F951-F962, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28249836

RESUMO

Transforming growth factor-alpha (TGFA) has been shown to play a role in experimental chronic kidney disease associated with nephron reduction, while its role in diabetic kidney disease (DKD) is unknown. We show here that intrarenal TGFA mRNA expression, as well as urine and serum TGFA, are increased in human DKD. We used a TGFA neutralizing antibody to determine the role of TGFA in two models of renal disease, the remnant surgical reduction model and the uninephrectomized (uniNx) db/db DKD model. In addition, the contribution of TGFA to DKD progression was examined using an adeno-associated virus approach to increase circulating TGFA in experimental DKD. In vivo blockade of TGFA attenuated kidney disease progression in both nondiabetic 129S6 nephron reduction and Type 2 diabetic uniNx db/db models, whereas overexpression of TGFA in uniNx db/db model accelerated renal disease. Therapeutic activity of the TGFA antibody was enhanced with renin angiotensin system inhibition with further improvement in renal parameters. These findings suggest a pathologic contribution of TGFA in DKD and support the possibility that therapeutic administration of neutralizing antibodies could provide a novel treatment for the disease.


Assuntos
Nefropatias Diabéticas/metabolismo , Rim/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Idoso , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Pressão Sanguínea , Células Cultivadas , Dependovirus/genética , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Receptores ErbB/metabolismo , Matriz Extracelular/metabolismo , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Taxa de Filtração Glomerular , Humanos , Hipertensão/complicações , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Rim/cirurgia , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Pessoa de Meia-Idade , Nefrectomia , Fosforilação , Sistema Renina-Angiotensina , Transdução de Sinais , Fatores de Tempo , Fator de Crescimento Transformador alfa/antagonistas & inibidores , Fator de Crescimento Transformador alfa/deficiência , Fator de Crescimento Transformador alfa/genética
7.
Am J Physiol Regul Integr Comp Physiol ; 309(5): R467-74, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26108870

RESUMO

Mice provide a unique platform to dissect disease pathogenesis, with the availability of recombinant inbred strains and diverse genetically modified strains. Leveraging these reagents to elucidate the mechanisms of hypertensive tissue injury has been hindered by difficulty establishing persistent hypertension in these inbred lines. ANG II infusion provides relatively short-term activation of the renin-angiotensinogen system (RAS) with concomitant elevated arterial pressure. Longer-duration studies using renin transgenic mice are powerful models of chronic hypertension, yet are limited by the genetic background on which the transgene exists and the exposure throughout development. The present studies characterized hypertension produced by transduction with a renin-coding adeno-associated virus (ReninAAV). ReninAAV mice experienced elevated circulating renin with concurrent elevations in arterial pressure. Following a single injection of ReninAAV, arterial pressure increased on average +56 mmHg, an increase that persisted for at least 12 wk in three distinct and widely used strains of adult mice: 129/S6, C56BL/6, and DBA/2J. This was accomplished without surgical implantation of pumps or complex breeding and backcrossing. In addition, ReninAAV mice developed pathophysiological changes associated with chronic hypertension, including increased heart weight and albuminuria. Thus ReninAAV provides a unique tool to study the onset of and effects of persistent hypertension in diverse murine models. This model should facilitate our understanding of the pathogenesis of hypertensive injury.


Assuntos
Pressão Arterial , Dependovirus/metabolismo , Vetores Genéticos , Hipertensão/metabolismo , Sistema Renina-Angiotensina , Renina/biossíntese , Transdução Genética , Albuminúria/genética , Albuminúria/metabolismo , Animais , Pressão Arterial/genética , Cardiomegalia/genética , Cardiomegalia/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Progressão da Doença , Predisposição Genética para Doença , Hipertensão/genética , Hipertensão/fisiopatologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Mutação , Fenótipo , Renina/genética , Sistema Renina-Angiotensina/genética , Fatores de Tempo
8.
J Pharmacol Exp Ther ; 349(2): 330-43, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24518034

RESUMO

At least seven distinct epidermal growth factor (EGF) ligands bind to and activate the EGF receptor (EGFR). This activation plays an important role in the embryo and in the maintenance of adult tissues. Importantly, pharmacologic EGFR inhibition also plays a critical role in the pathophysiology of diverse disease states, especially cancer. The roles of specific EGFR ligands are poorly defined in these disease states. Accumulating evidence suggests a role for transforming growth factor α (TGFα) in skin, lung, and kidney disease. To explore the role of Tgfa, we generated a monoclonal antibody (mAb41) that binds to and neutralizes human Tgfa with high affinity (KD = 36.5 pM). The antibody also binds human epiregulin (Ereg) (KD = 346.6 pM) and inhibits ligand induced myofibroblast cell proliferation (IC50 values of 0.52 and 1.12 nM for human Tgfa and Ereg, respectively). In vivo, a single administration of the antibody to pregnant mice (30 mg/kg s.c. at day 14 after plug) or weekly administration to neonate mice (20 mg/kg s.c. for 4 weeks) phenocopy Tgfa knockout mice with curly whiskers, stunted growth, and expansion of the hypertrophic zone of growth plate cartilage. Humanization of this monoclonal antibody to a human IgG4 antibody (LY3016859) enables clinical development. Importantly, administration of the humanized antibody to cynomolgus monkeys is absent of the skin toxicity observed with current EGFR inhibitors used clinically and no other pathologies were noted, indicating that neutralization of Tgfa could provide a relatively safe profile as it advances in clinical development.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Neutralizantes/metabolismo , Anticorpos Neutralizantes/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Epirregulina , Humanos , Imunoglobulina G/imunologia , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Miofibroblastos/citologia , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Ligação Proteica , Fator de Crescimento Transformador alfa/genética
9.
Mol Metab ; 2(2): 69-73, 2013 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-24199153

RESUMO

The adipocyte-derived hormone, leptin, is a master regulator of energy homeostasis. Leptin action in the central nervous system also contributes to arterial pressure regulation through its capacity to increase renal sympathetic outflow. The accumulating evidence pointing to a key role for leptin in the adverse sympathetic and cardiovascular consequences of excessive adiposity highlight the importance of understanding the mechanisms underlying the sympathetic and cardiovascular effects of leptin. The ability of the leptin receptor to stimulate various intracellular pathways allows leptin to regulate physiological processes in a specific manner. In this review, we examine the role of the PI3K pathway emanating from the leptin receptor in mediating the sympathetic and arterial pressure effects of leptin. We also discuss the relevance of PI3K signaling for obesity-induced hypertension through its role in mediating selective leptin resistance.

10.
Cell Metab ; 17(4): 599-606, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23541372

RESUMO

The fundamental importance of the hypothalamus in the regulation of autonomic and cardiovascular functions is well established. However, the molecular processes involved are not well understood. Here, we show that the mammalian (or mechanistic) target of rapamycin (mTOR) signaling in the hypothalamus is tied to the activity of the sympathetic nervous system and to cardiovascular function. Modulation of mTOR complex 1 (mTORC1) signaling caused dramatic changes in sympathetic traffic, blood flow, and arterial pressure. Our data also demonstrate the importance of hypothalamic mTORC1 signaling in transducing the sympathetic and cardiovascular actions of leptin. Moreover, we show that the PI3K pathway links the leptin receptor to mTORC1 signaling and that changes in its activity impact sympathetic traffic and arterial pressure. These findings establish mTORC1 activity in the hypothalamus as a key determinant of sympathetic and cardiovascular regulation and suggest that dysregulated hypothalamic mTORC1 activity may influence the development of cardiovascular diseases.


Assuntos
Hipotálamo/metabolismo , Leptina/metabolismo , Proteínas/metabolismo , Animais , Pressão Arterial/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Leucina/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas/antagonistas & inibidores , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores para Leptina/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo , Serina-Treonina Quinases TOR
11.
Clin Auton Res ; 23(1): 1-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22714900

RESUMO

Leptin is an adipocyte-derived hormone that relays a satiety signal to the brain. The effect of leptin on the sympathetic nervous system is an important aspect in the regulation of energy homeostasis as well as several other physiological functions. The arcuate nucleus of the hypothalamus is considered a major site for the regulation of physiological processes by leptin. However, there is growing recognition that other hypothalamic and extra-hypothalamic brain nuclei are important for leptin regulation of physiological processes including sympathetic nerve traffic. The current review discusses the various hypothalamic and extra-hypothalamic nuclei that have been implicated in leptin-induced increase in regional sympathetic nerve activity. The continuous rise in the prevalence of obesity underscores the importance of understanding the underlying neural mechanisms regulating sympathetic traffic to different tissues to design effective strategies to reverse obesity and associated diseases.


Assuntos
Encéfalo/fisiologia , Metabolismo Energético/fisiologia , Leptina/fisiologia , Sistema Nervoso Simpático/fisiologia , Animais , Humanos
12.
Circ Res ; 108(7): 808-12, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21311043

RESUMO

RATIONALE: The hypothalamic arcuate nucleus (ARC) is considered a major site for leptin signaling that regulates several physiological processes. OBJECTIVE: To test the hypothesis that leptin receptor in the ARC is required to mediate leptin-induced sympathetic activation. METHODS AND RESULTS: First, we used the ROSA Cre-reporter mice to establish the feasibility of driving Cre expression in the ARC in a controlled manner with bilateral microinjection of adenovirus-expressing Cre-recombinase (Ad-Cre). Ad-Cre microinjection into the ARC of ObR(flox/flox) mice robustly reduced ObR expression and leptin-induced Stat3 activation in the ARC but not in the adjacent nuclei, confirming the efficacy and selectivity of the ARC deletion of ObR. Critically, deletion of ObR in the ARC attenuated brown adipose tissue and renal sympathetic nerve responses to leptin. We also examined whether ObR in the ARC is required for the preserved leptin-induced increase in renal sympathetic activity in dietary obesity. We found that deletion of ARC ObR abrogated leptin-induced increases in renal sympathetic discharge and resolved arterial pressure elevation in diet-induced obese ObR(flox/flox) mice. CONCLUSIONS: These data demonstrate a critical role for ObR in the ARC in mediating the sympathetic nerve responses to leptin and in the adverse sympathoexcitatory effects of leptin in obesity.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Deleção de Genes , Leptina/farmacologia , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Proteínas de Fluorescência Verde , Homozigoto , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/fisiopatologia , Sistema Nervoso Simpático/fisiopatologia
13.
Hypertension ; 57(3): 627-32, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21263121

RESUMO

Leptin acts in the brain to regulate food intake and energy expenditure. Leptin also increases renal sympathetic nerve activity and arterial pressure. The divergent signaling capacities of the leptin receptor (ObRb) mediate the stimulation of various intracellular pathways that are important for leptin control of physiological processes. We evaluated the cardiovascular and sympathetic consequences of disrupting the signal emanating from tyrosine985 of ObRb. For this, we used Lepr(L985) (l/l) mice, which carry a loss of function mutation replacing tyrosine985 of ObRb with leucine. Body weight of l/l mice was not significantly different from wild-type controls. In contrast, radiotelemetry measurements revealed that the l/l mice had higher arterial pressure and heart rate as compared with controls. Ganglionic blockade caused a greater arterial pressure fall in the l/l mice relative to controls. In addition, leptin treatment induced a larger increase in arterial pressure and heart rate in the l/l versus wild-type mice. Finally, we compared the response of renal and brown adipose tissue sympathetic nerve activity to intracerebroventricular injection of leptin (2 µg) between l/l and control mice. Leptin-induced increase in renal sympathetic nerve activity was greater in l/l mice relative to controls. In contrast, the brown adipose tissue sympathetic nerve activity response to leptin was attenuated in the l/l mice relative to controls. These data indicate that selective loss of leptin receptor signaling emanating from tyrosine985 enhances the cardiovascular and renal sympathetic effects of leptin. These findings provide important insight into the molecular mechanisms underlying leptin's effects on the sympathetic cardiovascular function and arterial pressure.


Assuntos
Pressão Sanguínea/genética , Frequência Cardíaca/genética , Mutação , Receptores para Leptina/genética , Sistema Nervoso Simpático/fisiologia , Tecido Adiposo Marrom/inervação , Tecido Adiposo Marrom/metabolismo , Substituição de Aminoácidos , Análise de Variância , Animais , Pressão Sanguínea/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Rim/inervação , Rim/metabolismo , Leptina/metabolismo , Leptina/farmacologia , Camundongos , Camundongos Transgênicos , Receptores para Leptina/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos
14.
Anat Rec (Hoboken) ; 291(12): 1574-86, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18951515

RESUMO

The cardiac-specific -497 bp promoter of rat cardiac troponin T (cTnT) contains two similar modules, D and F, each of which possesses TCTG(G/C) direct repeats and A/T-rich sites. To identify cis-elements critical for cardiac specificity, a -249 bp promoter containing only module F and its site-directed mutations were used to generate transgenic mice. Transgene expression of the -249 bp promoter remained cardiac-specific, despite low and nonuniform expression. The nonuniform expression pattern of the transgene coincided with differential expression of HMGB1, which appeared to be the predominant form of HMGB family proteins in the heart. The HMGB1 binds to the A/T-rich/MEF2-like sites of the cTnT promoter, as determined by chromatin immunoprecipitation assays. Mice carrying the -249 bp promoter with point mutations disrupting the direct repeats expressed transgene at lower levels in the heart and ectopically in the brain. Ectopic expression of transgene was also observed in developing limbs and head. These results suggest an important role for the direct repeat in determining the cardiac specificity. Furthermore, mice carrying a mutant promoter simultaneously disrupting the direct repeats and overlapping GATA site failed to express the transgene in any tissues tested. Therefore, the direct repeat and overlapping GATA site are critical for the expression level and cardiac specificity. The F module controls one level of cardiac specificity. For a uniform and high level of cardiac-specific expression, the upstream element (-497 to -250 bp) is further required, possibly through the D enhancer module and the combination of Nkx2.5 and GATA sites.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Coração/embriologia , Miocárdio/metabolismo , Regiões Promotoras Genéticas/genética , Sequências Repetitivas de Ácido Nucleico/genética , Troponina T/genética , Animais , Sequência de Bases/genética , Padronização Corporal/genética , Sinalização do Cálcio/genética , Diferenciação Celular/genética , Elementos Facilitadores Genéticos/genética , Fatores de Transcrição GATA/genética , Camundongos , Camundongos Transgênicos , Contração Muscular/genética , Mutagênese Sítio-Dirigida , Miocárdio/ultraestrutura , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Mutação Puntual/genética , Elementos Reguladores de Transcrição/genética , Transcrição Gênica/genética , Transgenes/genética , Troponina T/biossíntese
15.
J Biol Chem ; 282(49): 36024-36, 2007 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-17925400

RESUMO

Targeted deletion of mXinalpha results in cardiac hypertrophy and cardiomyopathy with conduction defects (Gustafson-Wagner, E., Sinn, H. W., Chen, Y.-L., Wang, D.-Z., Reiter, R. S., Lin, J. L.-C., Yang, B., Williamson, R. A., Chen, J. N., Lin, C.-I., and Lin, J. J.-C. (2007) Am. J. Physiol. 293, H2680-H2692). To understand the underlying mechanisms leading to such cardiac defects, the functional domains of mXinalpha and its interacting proteins were investigated. Interaction studies using co-immunoprecipitation, pull-down, and yeast two-hybrid assays revealed that mXinalpha directly interacts with beta-catenin. The beta-catenin-binding site on mXinalpha was mapped to amino acids 535-636, which overlaps with the known actin-binding domains composed of the Xin repeats. The overlapping nature of these domains provides insight into the molecular mechanism for mXinalpha localization and function. Purified recombinant glutathione S-transferase- or His-tagged mXinalpha proteins are capable of binding and bundling actin filaments, as determined by co-sedimentation and electron microscopic studies. The binding to actin was saturated at an approximate stoichiometry of nine actin monomers to one mXinalpha. A stronger interaction was observed between mXinalpha C-terminal deletion and actin as compared with the interaction between full-length mXinalpha and actin. Furthermore, force expression of green fluorescent protein fused to an mXinalpha C-terminal deletion in cultured cells showed greater stress fiber localization compared with force-expressed GFP-mXinalpha. These results suggest a model whereby the C terminus of mXinalpha may prevent the full-length molecule from binding to actin, until the beta-catenin-binding domain is occupied by beta-catenin. The binding of mXinalpha to beta-catenin at the adherens junction would then facilitate actin binding. In support of this model, we found that the actin binding and bundling activity of mXinalpha was enhanced in the presence of beta-catenin.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Junções Aderentes/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , beta Catenina/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/ultraestrutura , Actinas/genética , Junções Aderentes/genética , Junções Aderentes/patologia , Sequência de Aminoácidos/genética , Animais , Células CHO , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Cricetinae , Cricetulus , Proteínas de Ligação a DNA/genética , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/patologia , Hipertrofia/genética , Hipertrofia/metabolismo , Hipertrofia/patologia , Camundongos , Modelos Biológicos , Proteínas Nucleares/genética , Mapeamento de Peptídeos , Ligação Proteica/genética , Estrutura Terciária de Proteína , Coelhos , Deleção de Sequência , Técnicas do Sistema de Duplo-Híbrido , beta Catenina/genética
16.
Methods Mol Biol ; 366: 183-201, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17568125

RESUMO

To understand how cardiac gene expression is regulated, the identification and characterization of cis-regulatory elements and their trans-acting factors by gel mobility shift assay (GMSA) or gel retardation assay are essential and common steps. In addition to providing a general protocol for GMSA, this chapter describes some applications of this assay to characterize cardiac-specific and ubiquitous trans-acting factors bound to regulatory elements [novel TCTG(G/C) direct repeat and A/T-rich region] of the rat cardiac troponin T promoter. In GMSA, the specificity of the binding of trans-acting factor to labeled DNA probe should be verified by the addition of unlabeled probe in the reaction mixture. The migratory property of DNA-protein complexes formed by protein extracts prepared from different tissues can be compared to determine the tissue specificity of trans-acting factors. GMSA, coupled with specific antibody to trans-acting factor (antibody supershift assay), is used to identify proteins present in the DNA-protein complex. The gel-shift competition assay with an unlabeled probe containing a slightly different sequence is a powerful technique used to assess the sequence specificity and relative binding affinity of a DNA-protein interaction. GMSA with SDS-PAGE fractionated proteins allows for the determination of the apparent molecular mass of bound trans-acting factor.


Assuntos
Ensaio de Desvio de Mobilidade Eletroforética/métodos , Miocárdio/metabolismo , Regiões Promotoras Genéticas , Elementos Reguladores de Transcrição/genética , Fatores de Transcrição/genética , Animais , Sequência de Bases , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Mucosa Gástrica/metabolismo , Fígado/metabolismo , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Ligação Proteica , Ratos , Homologia de Sequência do Ácido Nucleico , Troponina T/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...