Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cells ; 31(2): 327-37, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23132593

RESUMO

Hematopoietic stem cells (HSCs) reside in proximity to bone marrow endothelial cells (BM ECs) and maintenance of the HSC pool is dependent upon EC-mediated c-kit signaling. Here, we used genetic models to determine whether radioprotection of BM ECs could facilitate hematopoietic regeneration following radiation-induced myelosuppression. We developed mice bearing deletion of the proapoptotic proteins, BAK and BAX, in Tie2(+) ECs and HSCs (Tie2Bak/Bax(Fl/-) mice) and compared their hematopoietic recovery following total body irradiation (TBI) with mice which retained Bax in Tie2(+) cells. Mice bearing deletion of Bak and Bax in Tie2(+) cells demonstrated protection of BM HSCs, preserved BM vasculature, and 100% survival following lethal dose TBI. In contrast, mice that retained Bax expression in Tie2(+) cells demonstrated depletion of BM HSCs, disrupted BM vasculature, and 10% survival post-TBI. In a complementary study, VEcadherinBak/Bax(Fl/-) mice, which lack Bak and Bax in VEcadherin(+) ECs, also demonstrated increased recovery of BM stem/progenitor cells following TBI compared to mice which retained Bax in VEcadherin(+) ECs. Importantly, chimeric mice that lacked Bak and Bax in HSCs but retained Bak and Bax in BM ECs displayed significantly decreased HSC content and survival following TBI compared to mice lacking Bak and Bax in both HSCs and BM ECs. These data suggest that the hematopoietic response to ionizing radiation is dependent upon HSC-autonomous responses but is regulated by BM EC-mediated mechanisms. Therefore, BM ECs may be therapeutically targeted as a means to augment hematopoietic reconstitution following myelosuppression.


Assuntos
Células da Medula Óssea/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Lesões Experimentais por Radiação/metabolismo , Receptores Proteína Tirosina Quinases/genética , Irradiação Corporal Total , Animais , Células da Medula Óssea/patologia , Células da Medula Óssea/efeitos da radiação , Caderinas/genética , Caderinas/metabolismo , Células Endoteliais/patologia , Células Endoteliais/efeitos da radiação , Expressão Gênica/efeitos da radiação , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/efeitos da radiação , Camundongos , Camundongos Transgênicos , Lesões Experimentais por Radiação/genética , Lesões Experimentais por Radiação/mortalidade , Lesões Experimentais por Radiação/patologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptor TIE-2 , Regeneração/efeitos da radiação , Transdução de Sinais/efeitos da radiação , Análise de Sobrevida , Proteína Killer-Antagonista Homóloga a bcl-2/deficiência , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína X Associada a bcl-2/deficiência , Proteína X Associada a bcl-2/genética
2.
Sci Signal ; 5(234): ra52, 2012 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-22827996

RESUMO

Radiation therapy, which is used for the treatment of some cancers, can cause delayed heart damage. In the heart, p53 influences myocardial injury that occurs after multiple types of stress. Here, we demonstrated that p53 functioned in endothelial cells to protect mice from myocardial injury after whole-heart irradiation. Mice with an endothelial cell-specific deletion of p53 succumbed to heart failure after whole-heart irradiation as a result of myocardial necrosis, systolic dysfunction, and cardiac hypertrophy. Moreover, the onset of cardiac dysfunction was preceded by alterations in myocardial vascular permeability and density, which resulted in cardiac ischemia and myocardial hypoxia. Mechanistic studies with primary cardiac endothelial cells irradiated in vitro indicated that p53 signaling caused mitotic arrest and protected cardiac endothelial cells from cell death resulting from abnormal mitosis or mitotic catastrophe. Furthermore, mice lacking the cyclin-dependent kinase inhibitor p21, which is a transcriptional target of p53, were also sensitized to myocardial injury after whole-heart irradiation. Together, our results demonstrate that the p53-p21 axis functions to prevent radiation-induced myocardial injury in mice.


Assuntos
Cardiomegalia/patologia , Células Endoteliais/metabolismo , Miocárdio/patologia , Lesões Experimentais por Radiação/prevenção & controle , Radioterapia/efeitos adversos , Sístole/efeitos da radiação , Proteína Supressora de Tumor p53/metabolismo , Quinases Ativadas por p21/metabolismo , Análise de Variância , Animais , Permeabilidade Capilar/genética , Cardiomegalia/etiologia , Fluoroscopia , Deleção de Genes , Integrases , Camundongos , Camundongos Transgênicos , Necrose , Receptor TIE-2/genética , Proteína Supressora de Tumor p53/deficiência , Quinases Ativadas por p21/deficiência
3.
Radiat Res ; 177(3): 307-10, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22165824

RESUMO

Exposure of the gastrointestinal (GI) tract to high doses of radiation can lead to lethality from the GI syndrome. Although the molecular mechanism regulating the GI syndrome remains to be fully defined, we have recently demonstrated that p53 within the GI epithelial cells controls the radiation-induced GI syndrome. Mice lacking p53 in the GI epithelium were sensitized to the GI syndrome, while transgenic mice with one additional copy of p53 called "Super p53" mice were protected from the GI syndrome. Here, we crossed Super p53 mice to p21⁻/⁻ mice that lack the cyclin-dependent kinase inhibitor p21. Super p53; p21⁻/⁻ mice were sensitized to the GI syndrome compared to Super p53 mice that retain one p21 allele. In addition, mice lacking p21 were not protected from the GI syndrome with one extra copy of p53. These results suggest that p21 protects Super p53 mice from the GI syndrome.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Gastroenteropatias/genética , Gastroenteropatias/metabolismo , Lesões por Radiação/genética , Lesões por Radiação/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Inibidor de Quinase Dependente de Ciclina p21/deficiência , Dosagem de Genes , Camundongos , Camundongos Endogâmicos C57BL
4.
Science ; 327(5965): 593-6, 2010 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-20019247

RESUMO

Acute exposure to ionizing radiation can cause lethal damage to the gastrointestinal (GI) tract, a condition called the GI syndrome. Whether the target cells affected by radiation to cause the GI syndrome are derived from the epithelium or endothelium and whether the target cells die by apoptosis or other mechanisms are controversial issues. Studying mouse models, we found that selective deletion of the proapoptotic genes Bak1 and Bax from the GI epithelium or from endothelial cells did not protect mice from developing the GI syndrome after sub-total-body gamma irradiation. In contrast, selective deletion of p53 from the GI epithelium, but not from endothelial cells, sensitized irradiated mice to the GI syndrome. Transgenic mice overexpressing p53 in all tissues were protected from the GI syndrome after irradiation. These results suggest that the GI syndrome is caused by the death of GI epithelial cells and that these epithelial cells die by a mechanism that is regulated by p53 but independent of apoptosis.


Assuntos
Apoptose , Raios gama/efeitos adversos , Enteropatias/fisiopatologia , Mucosa Intestinal/efeitos da radiação , Intestino Delgado/efeitos da radiação , Lesões por Radiação/fisiopatologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Morte Celular , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Células Epiteliais/efeitos da radiação , Deleção de Genes , Genes p53 , Enteropatias/etiologia , Enteropatias/patologia , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiopatologia , Intestino Delgado/patologia , Intestino Delgado/fisiopatologia , Mesoderma/citologia , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Doses de Radiação , Lesões por Radiação/etiologia , Lesões por Radiação/patologia , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
5.
Circ Heart Fail ; 2(4): 351-60, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19808359

RESUMO

BACKGROUND: The direct consequences of a persistently increased myocardial expression of the unique matrix metalloproteinase (MMP) membrane type-1 (MT1-MMP) on myocardial remodeling remained unexplored. METHODS AND RESULTS: Cardiac-restricted MT1-MMPexp was constructed in mice using the full-length human MT1-MMP gene ligated to the myosin heavy chain promoter, which yielded approximately a 200% increase in MT1-MMP when compared with age/strain-matched wild-type (WT) mice. Left ventricular (LV) function and geometry was assessed by echocardiography in 3-month ("young") WT (n=32) and MT1-MMPexp (n=20) mice and compared with 14-month ("middle-aged") WT (n=58) and MT1-MMPexp (n=35) mice. LV end-diastolic volume was similar between the WT and MT1-MMPexp young groups, as was LV ejection fraction. In the middle-aged WT mice, LV end-diastolic volume and ejection fraction was similar to young WT mice. However, in the MT1-MMPexp middle-aged mice, LV end-diastolic volume was approximately 43% higher and LV ejection fraction 40% lower (both P<0.05). Moreover, in the middle-aged MT1-MMPexp mice, myocardial fibrillar collagen increased by nearly 2-fold and was associated with approximately 3-fold increase in the processing of the profibrotic molecule, latency-associated transforming growth factor binding protein. In a second study, 14-day survival after myocardial infarction was significantly lower in middle-aged MT1-MMPexp mice. CONCLUSIONS: Persistently increased myocardial MT1-MMP expression, in and of itself, caused LV remodeling, myocardial fibrosis, dysfunction, and reduced survival after myocardial injury. These findings suggest that MT1-MMP plays a mechanistic role in adverse remodeling within the myocardium.


Assuntos
Envelhecimento/genética , Metaloproteinase 14 da Matriz/biossíntese , Miocárdio/metabolismo , Remodelação Ventricular/genética , Animais , Expressão Gênica , Humanos , Camundongos
6.
Am J Physiol Heart Circ Physiol ; 297(5): H1744-51, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19734364

RESUMO

Cardiac pathology, such as myocardial infarction (MI), activates intracellular proteases that often trigger programmed cell death and contribute to maladaptive changes in myocardial structure and function. To test whether inhibition of calpain, a Ca(2+)-dependent cysteine protease, would prevent these changes, we used a mouse MI model. Calpeptin, an aldehydic inhibitor of calpain, was intravenously administered at 0.5 mg/kg body wt before MI induction and then at the same dose subcutaneously once per day. Both calpeptin-treated (n = 6) and untreated (n = 6) MI mice were used to study changes in myocardial structure and function after 4 days of MI, where end-diastolic volume (EDV) and left ventricular ejection fraction (EF) were measured by echocardiography. Calpain activation and programmed cell death were measured by immunohistochemistry, Western blotting, and TdT-mediated dUTP nick-end labeling (TUNEL). In MI mice, calpeptin treatment resulted in a significant improvement in EF [EF decreased from 67 + or - 2% pre-MI to 30 + or - 4% with MI only vs. 41 + or - 2% with MI + calpeptin] and attenuated the increase in EDV [EDV increased from 42 + or - 2 microl pre-MI to 73 + or - 4 microl with MI only vs. 55 + or - 4 microl with MI + calpeptin]. Furthermore, calpeptin treatment resulted in marked reduction in calpain- and caspase-3-associated changes and TUNEL staining. These studies indicate that calpain contributes to MI-induced alterations in myocardial structure and function and that it could be a potential therapeutic target in treating MI patients.


Assuntos
Calpaína/antagonistas & inibidores , Cardiotônicos/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/enzimologia , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Calpaína/metabolismo , Cardiotônicos/administração & dosagem , Caspase 3/metabolismo , Inibidores de Cisteína Proteinase/administração & dosagem , Dipeptídeos/administração & dosagem , Modelos Animais de Doenças , Ativação Enzimática , Injeções Intravenosas , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Volume Sistólico/efeitos dos fármacos , Fatores de Tempo , Ultrassonografia , Remodelação Ventricular/efeitos dos fármacos
7.
Ann Thorac Surg ; 86(4): 1268-76, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18805174

RESUMO

BACKGROUND: Left ventricular (LV) remodeling after myocardial infarction (MI) commonly causes infarct expansion (IE). This study sought to interrupt IE through microinjections of a biocompatible composite material into the post-MI myocardium. METHODS: MI was created in 21 pigs (coronary ligation). Radiopaque markers (2-mm diameter) were placed for IE (fluoroscopy). Pigs were randomized for microinjections (25 injections; 2- x 2-cm array; 200 microL/injection) at 7 days post-MI of a fibrin-alginate composite (Fib-Alg; fibrinogen, fibronectin, factor XIII, gelatin-grafted alginate, thrombin; n = 11) or saline (n = 10). RESULTS: At 7 days after injection (14 days post-MI), LV posterior wall thickness was higher in the Fib-Alg group than in the saline group (1.07 +/- 0.11 vs 0.69 +/- 0.07 cm, respectively, p = 0.002). At 28 days post-MI, the area within the markers (IE) increased from baseline (1 cm2) in the saline (1.71 +/- 0.13 cm2, p = 0.010) and Fib-Alg groups (1.44 +/- 0.23 cm2, p < 0.001). However, the change in IE at 21 and 28 days post-MI was reduced in the Fib-Alg group (p=0.043 and p=0.019). Total collagen content within the MI region was similar in the saline and Fib-Alg groups (12.8 +/- 1.7 and 11.6 +/- 1.5 microg/mg, respectively, p = NS). However, extractable collagen, indicative of solubility, was lower in the Fib-Alg group than the saline group (59.1 +/- 3.5 vs 71.0 +/- 6.1 microg/mL, p = 0.020). CONCLUSIONS: Targeted myocardial microinjection of the biocomposite attenuated the post-MI decrease in LV wall thickness and infarct expansion. Thus, intraoperative microinjections of biocompatible material may provide a novel approach for interrupting post-MI LV remodeling.


Assuntos
Materiais Biocompatíveis/administração & dosagem , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Remodelação Ventricular , Animais , Modelos Animais de Doenças , Feminino , Injeções Intralesionais , Masculino , Microinjeções , Infarto do Miocárdio/patologia , Probabilidade , Distribuição Aleatória , Valores de Referência , Sensibilidade e Especificidade , Suínos , Remodelação Ventricular/fisiologia
8.
Ann Thorac Surg ; 86(2): 568-75, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18640335

RESUMO

BACKGROUND: Cardiac surgery can result in left ventricular ischemia and reperfusion (I/R), the release of cytokines such as tumor necrosis factor, and oxidative stress with release of myeloperoxidase. Although aprotinin has been used in cardiac surgery, the likely multiple effects of this serine protease inhibitor limit clinical utility. This study tested the hypothesis that different aprotinin doses cause divergent effects on left ventricular contractility, cytokine release, and oxidative stress in the context of I/R. METHODS: Left ventricular I/R (30 minutes I, 60 minutes R) was induced in mice, and left ventricular contractility (maximal end-systolic elastance) determined. Mice were randomly allocated to 2 x 10(4) kallikrein inhibitory units (KIU)/kg aprotinin (n = 11), 4 x 10(4) KIU/kg aprotinin (n = 10), and vehicle (saline, n = 10). Based upon a fluorogenic assay, aprotinin doses of 2 and 4 x 10(4) KIU/kg resulted in plasma concentrations similar to those of the half and full Hammersmith doses, respectively. RESULTS: After I/R, maximal end-systolic elastance fell by more than 40% from baseline (p < 0.05), and this effect was attenuated by 2 x 10(4) KIU/kg but not 4 x 10(4) KIU/kg aprotinin. Tumor necrosis factor increased by more than 60% from control (p < 0.05) with I/R, but was reduced with 4 x 10(4) KIU/kg aprotinin. Myeloperoxidase increased with I/R, and was reduced to the greatest degree by 2 x 10(4) KIU/kg aprotinin. CONCLUSIONS: Aprotinin influences left ventricular contractility, cytokine release, and oxidative stress, which are dose dependent. These results provide mechanistic evidence that multiple pathways are differentially affected by aprotinin in a context relevant to cardiac surgery.


Assuntos
Aprotinina/administração & dosagem , Contração Miocárdica/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Serina Proteinase/administração & dosagem , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Procedimentos Cirúrgicos Cardíacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Elasticidade , Processamento de Imagem Assistida por Computador , Interleucina-6/análise , Camundongos , Camundongos Endogâmicos , Miocárdio/enzimologia , Peroxidase/análise , Fator de Necrose Tumoral alfa/sangue , Função Ventricular Esquerda/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...