Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Leukoc Biol ; 115(3): 573-582, 2024 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-38038378

RESUMO

CD115, the receptor for colony stimulating factor 1, is essential for survival and differentiation of monocytes and macrophages and is therefore frequently used to define monocyte subsets and their progenitors in immunological assays. However, CD115 surface expression and detection by flow cytometry is greatly influenced by cell isolation and processing methods, organ source, and disease context. In a systematic analysis of murine monocytes, we define experimental conditions that preserve or limit CD115 surface expression and staining by flow cytometry. We also find that, independent of conditions, CD115 surface levels are consistently lower in Ly6Clo monocytes than in Ly6Chi monocytes, with the exception of Ly6Clo monocytes in the bone marrow. Furthermore, in contrast to IL-34, the presence of colony stimulating factor 1 impairs CD115 antibody staining in a dose-dependent manner, which, in a model of ischemic kidney injury with elevated levels of colony stimulating factor 1, influenced quantification of kidney monocytes. Thus, staining and experimental conditions affect quantitative and qualitative analysis of monocytes and may influence experimental conclusions.


Assuntos
Monócitos , Receptor de Fator Estimulador de Colônias de Macrófagos , Camundongos , Animais , Monócitos/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Citometria de Fluxo , Macrófagos/metabolismo , Diferenciação Celular
2.
Front Immunol ; 14: 1240327, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37691936

RESUMO

Ischemia causes an inflammatory response featuring monocyte-derived macrophages (MF) involved in angiogenesis and tissue repair. Angiogenesis and ischemic macrophage differentiation are regulated by Notch signaling via Notch ligand Delta-like 1 (Dll1). Colony stimulating factor 1 (CSF-1) is an essential MF lineage factor, but its role in ischemic macrophage development and the interaction with Notch signaling is so far unclear. Using a mouse model of hind limb ischemia with CSF-1 inhibitor studies and Dll1 heterozygous mice we show that CSF-1 is induced in the ischemic niche by a subpopulation of stromal cells expressing podoplanin, which was paralleled by the development of ischemic macrophages. Inhibition of CSF-1 signaling with small molecules or blocking antibodies impaired macrophage differentiation but prolonged the inflammatory response, resulting in impaired perfusion recovery and tissue regeneration. Yet, despite high levels of CSF-1, macrophage maturation and perfusion recovery were impaired in mice with Dll1 haploinsufficiency, while inflammation was exaggerated. In vitro, CSF-1 was not sufficient to induce full MF differentiation from donor monocytes in the absence of recombinant DLL1, while the presence of DLL1 in a dose-dependent manner stimulated MF differentiation in combination with CSF-1. Thus, CSF-1 is an ischemic niche factor that cooperates with Notch signaling in a non-redundant fashion to instruct macrophage cell fate and maturation, which is required for ischemic perfusion recovery and tissue repair.


Assuntos
Fator Estimulador de Colônias de Macrófagos , Doenças Vasculares Periféricas , Receptores Notch , Isquemia , Macrófagos , Monócitos , Animais , Camundongos
3.
Sci Rep ; 13(1): 12542, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37532879

RESUMO

Flap surgery is a common treatment for severe wounds and a major determinant of surgical outcome. Flap survival and healing depends on adaptation of the local flap vasculature. Using a novel and defined model of fasciocutaneous flap surgery, we demonstrate that the Notch ligand Delta-like 1 (Dll1), expressed in vascular endothelial cells, regulates flap arteriogenesis, inflammation and flap survival. Utilizing the stereotyped anatomy of dorsal skin arteries, ligation of the major vascular pedicle induced strong collateral vessel development by end-to-end anastomosis in wildtype mice, which supported flap perfusion recovery over time. In mice with heterozygous deletion of Dll1, collateral vessel formation was strongly impaired, resulting in aberrant vascularization and subsequent necrosis of the tissue. Furthermore, Dll1 deficient mice showed severe inflammation in the flap dominated by monocytes and macrophages. This process is controlled by endothelial Dll1 in vivo, since the results were recapitulated in mice with endothelial-specific deletion of Dll1. Thus, our model provides a platform to study vascular adaptation to flap surgery and molecular and cellular regulators influencing flap healing and survival.


Assuntos
Células Endoteliais , Neovascularização Fisiológica , Camundongos , Animais , Neovascularização Fisiológica/fisiologia , Proteínas de Ligação ao Cálcio/genética , Cicatrização , Inflamação
4.
FASEB J ; 36(7): e22407, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35713543

RESUMO

Dietary phosphate intake in the Western population greatly exceeds the recommended dietary allowance and is linked to enhanced cardiovascular and all-cause mortality. It is unclear whether a chronic high phosphate diet (HPD) causes kidney injury in healthy individuals. Here, we show that feeding a 2% HPD in C57BL/6N mice for one up to six months resulted in hyperphosphatemia, hyperphosphaturia, increased plasma levels of fibroblast growth factor (FGF) 23, and parathyroid hormone (PTH) compared to mice on a 0.8% phosphate diet. Kidney injury was already noted after two months of HPD characterized by loss of proximal tubular (PT) cell polarity, flattened epithelia, disruption of brush border membranes, vacuolization, increased PT cell proliferation, marked interstitial mononuclear infiltration, and progressive accumulation of collagen fibers. HPD increased Stat3 activation and Kim-1 expression in PT epithelial cells and enhanced renal synthesis of chemokines recruiting monocytes and macrophages as well as macrophage related factors. Enhanced recruitment of F4/80+ macrophages around injured PT lesions was timely associated with increased Kim-1 synthesis, tubular MCP-1 expression, and degree of PT injury score. Likewise, tubulointerstitial fibrosis was associated with activation of Stat3/Kim-1 signaling pathway. The stimulation of human proximal tubular cells with high phosphate activated Stat3 signaling and induced HAVCR1 and CCL2 expression. We conclude that high phosphate results in progressive proximal tubular injury, indicating that high dietary phosphate intake may affect kidney health and therefore represents an underestimated health problem for the general population.


Assuntos
Nefropatias , Túbulos Renais Proximais , Animais , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Nefropatias/patologia , Túbulos Renais Proximais/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfatos/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
5.
Nat Commun ; 8(1): 952, 2017 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-29038527

RESUMO

Ischemia causes an inflammatory response that is intended to restore perfusion and homeostasis yet often aggravates damage. Here we show, using conditional genetic deletion strategies together with adoptive cell transfer experiments in a mouse model of hind limb ischemia, that blood vessels control macrophage differentiation and maturation from recruited monocytes via Notch signaling, which in turn promotes arteriogenesis and tissue repair. Macrophage maturation is controlled by Notch ligand Dll1 expressed in vascular endothelial cells of arteries and requires macrophage canonical Notch signaling via Rbpj, which simultaneously suppresses an inflammatory macrophage fate. Conversely, conditional mutant mice lacking Dll1 or Rbpj show proliferation and transient accumulation of inflammatory macrophages, which antagonizes arteriogenesis and tissue repair. Furthermore, the effects of Notch are sufficient to generate mature macrophages from monocytes ex vivo that display a stable anti-inflammatory phenotype when challenged with pro-inflammatory stimuli. Thus, angiocrine Notch signaling fosters macrophage maturation during ischemia.Molecular mechanisms of macrophage-mediated regulation of artery growth in response to ischemia are poorly understood. Here the authors show that vascular endothelium controls macrophage maturation and differentiation via Notch signaling, which in turn promotes arteriogenesis and ischemic tissue recovery.


Assuntos
Vasos Sanguíneos/fisiologia , Diferenciação Celular/fisiologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Isquemia/genética , Macrófagos/metabolismo , Neovascularização Fisiológica/genética , Transferência Adotiva , Animais , Proteínas de Ligação ao Cálcio , Deleção de Genes , Membro Posterior/irrigação sanguínea , Camundongos , Mutação , Receptores Notch , Transdução de Sinais
6.
Nat Commun ; 7: 12597, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27576369

RESUMO

A population of monocytes, known as Ly6C(lo) monocytes, patrol blood vessels by crawling along the vascular endothelium. Here we show that endothelial cells control their origin through Notch signalling. Using combinations of conditional genetic deletion strategies and cell-fate tracking experiments we show that Notch2 regulates conversion of Ly6C(hi) monocytes into Ly6C(lo) monocytes in vivo and in vitro, thereby regulating monocyte cell fate under steady-state conditions. This process is controlled by Notch ligand delta-like 1 (Dll1) expressed by a population of endothelial cells that constitute distinct vascular niches in the bone marrow and spleen in vivo, while culture on recombinant DLL1 induces monocyte conversion in vitro. Thus, blood vessels regulate monocyte conversion, a form of committed myeloid cell fate regulation.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Monócitos/fisiologia , Receptor Notch2/metabolismo , Transdução de Sinais/fisiologia , Transferência Adotiva , Animais , Antígenos Ly/metabolismo , Células da Medula Óssea/metabolismo , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Células Cultivadas , Células Endoteliais/metabolismo , Proteínas Ligadas por GPI/metabolismo , Voluntários Saudáveis , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Camundongos , Camundongos Knockout , Receptores de IgG/metabolismo , Proteínas Recombinantes/metabolismo , Baço/citologia
7.
Sci Rep ; 6: 25009, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-27146149

RESUMO

Activation of ß-catenin-dependent canonical Wnt signaling in endothelial cells plays a key role in angiogenesis during development and ischemic diseases, however, other roles of Wnt/ß-catenin signaling in endothelial cells remain poorly understood. Here, we report that sustained activation of ß-catenin signaling in endothelial cells causes cardiac dysfunction through suppressing neuregulin-ErbB pathway in the heart. Conditional gain-of-function mutation of ß-catenin, which activates Wnt/ß-catenin signaling in Bmx-positive arterial endothelial cells (Bmx/CA mice) led to progressive cardiac dysfunction and 100% mortality at 40 weeks after tamoxifen treatment. Electron microscopic analysis revealed dilatation of T-tubules and degeneration of mitochondria in cardiomyocytes of Bmx/CA mice, which are similar to the changes observed in mice with decreased neuregulin-ErbB signaling. Endothelial expression of Nrg1 and cardiac ErbB signaling were suppressed in Bmx/CA mice. The cardiac dysfunction of Bmx/CA mice was ameliorated by administration of recombinant neuregulin protein. These results collectively suggest that sustained activation of Wnt/ß-catenin signaling in endothelial cells might be a cause of heart failure through suppressing neuregulin-ErbB signaling, and that the Wnt/ß-catenin/NRG axis in cardiac endothelial cells might become a therapeutic target for heart failure.


Assuntos
Células Endoteliais/fisiologia , Receptores ErbB/antagonistas & inibidores , Insuficiência Cardíaca/fisiopatologia , Neuregulina-1/antagonistas & inibidores , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Análise de Sobrevida
8.
PLoS One ; 10(10): e0138542, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26431421

RESUMO

Arteriogenesis, the growth of natural bypass arteries, is triggered by hemodynamic forces within vessels and requires a balanced inflammatory response, involving induction of the chemokine MCP-1 and recruitment of leukocytes. However, little is known how these processes are coordinated. The MAP-kinase-activated-proteinkinase-2 (MK2) is a critical regulator of inflammatory processes and might represent an important link between cytokine production and cell recruitment during postnatal arteriogenesis. Therefore, the present study investigated the functional role of MK2 during postnatal arteriogenesis. In a mouse model of hindlimb ischemia (HLI) MK2-deficiency (MK2KO) significantly impaired ischemic blood flow recovery and growth of collateral arteries as well as perivascular recruitment of mononuclear cells and macrophages. This was accompanied by induction of endothelial MCP-1 expression in wildtype (WT) but not in MK2KO collateral arteries. Following HLI, MK2 activation rapidly occured in the endothelium of growing WT arteries in vivo. In vitro, inflammatory cytokines and cyclic stretch activated MK2 in endothelial cells, which was required for stretch- and cytokine-induced release of MCP-1. In addition, a monocyte cell autonomous function of MK2 was uncovered potentially regulating MCP-1-dependent monocyte recruitment to vessels: MCP-1 stimulation of WT monocytes induced MK2 activation and monocyte migration in vitro. The latter was reduced in MK2KO monocytes, while in vivo MK2 was activated in monocytes recruited to collateral arteries. In conclusion, MK2 regulates postnatal arteriogenesis by controlling vascular recruitment of monocytes/macrophages in a dual manner: regulation of endothelial MCP-1 expression in response to hemodynamic and inflammatory forces as well as MCP-1 dependent monocyte migration.


Assuntos
Artérias/crescimento & desenvolvimento , Endotélio Vascular/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/citologia , Monócitos/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Artérias/citologia , Células Cultivadas , Quimiocina CCL2/fisiologia , Quimiotaxia/fisiologia , Endotélio Vascular/enzimologia , Camundongos
9.
Arterioscler Thromb Vasc Biol ; 30(7): 1398-406, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20431064

RESUMO

OBJECTIVE: To determine the intracellular mechanisms mediating the angiogenic effects of integrin alpha v beta 5 overexpression in circulating angiogenic cells (CACs). METHODS AND RESULTS: Integrin alpha v beta 5 is expressed on angiogenic endothelial cells, and integrin alpha v beta 5 activation was shown to improve the reparative functions of endothelial progenitors within the cardiovascular system. CACs were transiently transfected with the full-length cDNA of human integrin beta 5 (CAC-ITGB5) or control-vector (CAC-vector). Integrin beta 5 overexpression was confirmed using flow cytometry, Western blot, and PCR analysis; it enhanced the angiogenic capacities of CACs in vitro (spheroid and Matrigel angiogenesis assay) and stimulated new vessel formation in vivo (murine hind limb ischemia model). Overexpression of ITGB5 resulted in integrin alpha v beta 5 phosphorylation and activation of Src kinase and signal transducer and activator of transcription (STAT) 3. Furthermore, elevated mRNA and protein expression of the CXC chemokine CXCL8 and the CC chemokine CCL2 was detected in CAC-ITGB5, and conditioned medium from CAC-ITGB5 enhanced the sprouting of coincubated human endothelial cells in a STAT3-, CXCL8-, and CCL2-dependent manner. CONCLUSIONS: Src kinase-mediated activation of STAT3 and subsequent angiogenic gene expression mediate the effects of integrin alpha v beta 5 and may be exploited to enhance the paracrine activities of CACs.


Assuntos
Células Endoteliais/enzimologia , Cadeias beta de Integrinas/metabolismo , Neovascularização Fisiológica , Comunicação Parácrina , Receptores de Vitronectina/metabolismo , Fator de Transcrição STAT3/metabolismo , Quinases da Família src/metabolismo , Animais , Western Blotting , Adesão Celular , Movimento Celular , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Citometria de Fluxo , Membro Posterior , Humanos , Cadeias beta de Integrinas/genética , Interleucina-8/genética , Interleucina-8/metabolismo , Isquemia/enzimologia , Isquemia/genética , Isquemia/fisiopatologia , Camundongos , Camundongos Nus , Músculo Esquelético/irrigação sanguínea , Fosforilação , RNA Mensageiro/metabolismo , Receptores de Vitronectina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção , Regulação para Cima
11.
J Mol Cell Cardiol ; 45(3): 394-403, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18671980

RESUMO

Cell-based therapy after myocardial infarction (MI) is a promising therapeutic option but the relevant cell subsets and dosage requirements are poorly defined. We hypothesized that cell therapy for myocardial infarction is improved by ex vivo expansion and high-dose transplantation of defined hematopoietic progenitor cells (HPCs). Since beta-catenin promotes self-renewal of stem cells we evaluated the therapeutic efficacy of beta-catenin-mediated ex vivo expansion of mouse HPCs in a mouse model of myocardial ischemia/reperfusion followed by intraarterial cell delivery. The impact of cell dose was determined by comparing a low-dose (LD, 5 x 10(5) cells) vs. a high-dose (HD, 1 x 10(7) cells) cell transplantation regimen of beta-catenin-HPCs. The impact of beta-catenin modification of HPCs was determined by comparing control-transduced HPCs (GFP-HPCs) vs. transgenic beta-catenin-HPCs. HD beta-catenin-HPCs significantly improved LV function and end-systolic and end-diastolic dimensions as compared to saline and LD beta-catenin-HPCs. Furthermore, while treatment with HD GFP-HPC resulted in a modest cardiac improvement the application of beta-catenin-HPCs was superior, resulting in a significant improvement in EF, FS and LVESD over saline and control GFP-HPC treatment. Although myocardial engraftment of HPCs was only transient, as determined by cell quantification after dye labeling, beta-catenin-HPC treatment significantly decreased infarct size, reduced cardiomyocyte apoptosis and increased capillary angiogenesis in vitro and in vivo. Ex vivo expanded HPCs improve cardiac function and remodeling post MI in a cell number- and beta-catenin-dependent manner.


Assuntos
Coração/fisiopatologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Miocárdio/metabolismo , Transdução Genética , beta Catenina/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Testes de Função Cardíaca , Contagem de Leucócitos , Camundongos , Infarto do Miocárdio/patologia , Miocárdio/patologia , Remodelação Ventricular/fisiologia , beta Catenina/fisiologia
12.
Cardiovasc Res ; 78(2): 294-300, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18093990

RESUMO

AIMS: Neovascularization is an important element of long-term functional recovery during chronic ischaemia. We postulated that haeme oxygenase (HO) is required for progenitor cell recruitment, neovascularization, and blood flow recovery after critical hindlimb ischaemia (HLI). METHODS AND RESULTS: The femoral artery was ligated in FVB/N mice proximal to its superficial and deep branches. Blood flow in the ischaemic hindlimb was determined by laser Doppler perfusion imaging. Capillary density was measured by isolectin staining, and mobilization of Sca-1(+)/Kdr(+) progenitor cells by FACS analysis. Progenitor cell recruitment to the ischaemic hindlimb was assessed after Tie2-lacZ transgenic bone marrow transplantation. Blood flow recovery after femoral artery ligation was significantly blunted in mice treated with the HO inhibitor tin protoporphyrin-IX (25 mg/kg i.p., every other day). HO-inhibited mice developed more pronounced limb necrosis, associated with impaired hindlimb motor function. Capillary density in the ischaemic hindlimb and mobilization of Sca-1(+)/Kdr(+) progenitor cells were significantly reduced after HO inhibition. After transplantation of Tie2-lacZ transgenic bone marrow cells into lethally irradiated wild-type mice, fewer LacZ(+) cells were detected in the ischaemic hindlimb muscle of HO-inhibited mice. Mechanistically, HO inhibition prevented the establishment of a stromal cell-derived factor-1 gradient for progenitor cell mobilization between the ischaemic hindlimb and bone marrow. CONCLUSION: HOs are required for progenitor cell recruitment, neovascularization, and functional recovery after HLI.


Assuntos
Movimento Celular , Células Endoteliais/enzimologia , Heme Oxigenase-1/metabolismo , Isquemia/enzimologia , Proteínas de Membrana/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Células-Tronco/enzimologia , Inibidores da Angiogênese/farmacologia , Animais , Velocidade do Fluxo Sanguíneo , Transplante de Medula Óssea , Capilares/metabolismo , Capilares/fisiopatologia , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Inibidores Enzimáticos/farmacologia , Heme Oxigenase-1/antagonistas & inibidores , Membro Posterior , Isquemia/diagnóstico por imagem , Isquemia/fisiopatologia , Fluxometria por Laser-Doppler , Masculino , Proteínas de Membrana/antagonistas & inibidores , Metaloporfirinas/farmacologia , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica/efeitos dos fármacos , Protoporfirinas/farmacologia , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Fluxo Sanguíneo Regional , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Fatores de Tempo , Ultrassonografia
13.
Circ Res ; 100(3): 363-71, 2007 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-17234965

RESUMO

Growth of functional arteries is essential for the restoration of blood flow to ischemic organs. Notch signaling regulates arterial differentiation upstream of ephrin-B2 during embryonic development, but its role during postnatal arteriogenesis is unknown. Here, we identify the Notch ligand Delta-like 1 (Dll1) as an essential regulator of postnatal arteriogenesis. Dll1 expression was specifically detected in arterial endothelial cells, but not in venous endothelial cells or capillaries. During ischemia-induced arteriogenesis endothelial Dll1 expression was strongly induced, Notch signaling activated and ephrin-B2 upregulated, whereas perivascular cells expressed proangiogenic vascular endothelial growth factor, and the ephrin-B2 activator EphB4. In heterozygous Dll1 mutant mice endothelial Notch activation and ephrin-B2 induction after hindlimb ischemia were absent, arterial collateral growth was abrogated and recovery of blood flow was severely impaired, but perivascular vascular endothelial growth factor and EphB4 expression was unaltered. In vitro, angiogenic growth factors synergistically activated Notch signaling by induction of Dll1, which was necessary and sufficient to regulate ephrin-B2 expression and to induce ephrin-B2 and EphB4-dependent branching morphogenesis in human arterial EC. Thus, Dll1-mediated Notch activation regulates ephrin-B2 expression and postnatal arteriogenesis.


Assuntos
Artérias/citologia , Endotélio Vascular/citologia , Regulação da Expressão Gênica/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Isquemia/fisiopatologia , Proteínas de Membrana/fisiologia , Neovascularização Fisiológica/fisiologia , Receptores Notch/fisiologia , Animais , Aorta/citologia , Artérias/química , Artérias/crescimento & desenvolvimento , Proteínas de Ligação ao Cálcio , Capilares/química , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Circulação Colateral/fisiologia , Constrição , Meios de Cultura Livres de Soro , Células Endoteliais/metabolismo , Inativação Gênica , Membro Posterior/irrigação sanguínea , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Isquemia/etiologia , Isquemia/genética , Camundongos , Camundongos Transgênicos , Morfogênese/genética , Morfogênese/fisiologia , Neovascularização Fisiológica/genética , Especificidade de Órgãos , RNA Interferente Pequeno/farmacologia , Receptor EphB2/biossíntese , Receptor EphB2/genética , Receptor EphB2/fisiologia , Receptor EphB4/biossíntese , Receptor EphB4/genética , Receptor EphB4/fisiologia , Veias/química
14.
Circ Res ; 100(1): 70-8, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17158336

RESUMO

Notch receptors are important mediators of cell fate during embryogenesis, but their role in adult physiology, particularly in postnatal angiogenesis, remains unknown. Of the Notch receptors, only Notch1 and Notch4 are expressed in vascular endothelial cells. Here we show that blood flow recovery and postnatal neovascularization in response to hindlimb ischemia in haploinsufficient global or endothelial-specific Notch1(+/-) mice, but not Notch4(-/-) mice, were impaired compared with wild-type mice. The expression of vascular endothelial growth factor (VEGF) in response to ischemia was comparable between wild-type and Notch mutant mice, suggesting that Notch1 is downstream of VEGF signaling. Treatment of endothelial cells with VEGF increases presenilin proteolytic processing, gamma-secretase activity, Notch1 cleavage, and Hes-1 (hairy enhancer of split homolog-1) expression, all of which were blocked by treating endothelial cells with inhibitors of phosphatidylinositol 3-kinase/protein kinase Akt or infecting endothelial cells with a dominant-negative Akt mutant. Indeed, inhibition of gamma-secretase activity leads to decreased angiogenesis and inhibits VEGF-induced endothelial cell proliferation, migration, and survival. Overexpression of the active Notch1 intercellular domain rescued the inhibitory effects of gamma-secretase inhibitors on VEGF-induced angiogenesis. These findings indicate that the phosphatidylinositol 3-kinase/Akt pathway mediates gamma-secretase and Notch1 activation by VEGF and that Notch1 is critical for VEGF-induced postnatal angiogenesis. These results suggest that Notch1 may be a novel therapeutic target for improving angiogenic response and blood flow recovery in ischemic limbs.


Assuntos
Animais Recém-Nascidos , Endotélio Vascular/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia/fisiopatologia , Neovascularização Fisiológica , Receptor Notch1/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Bovinos , Movimento Celular , Proliferação de Células , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ativação Enzimática , Isquemia/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Notch1/química , Receptor Notch1/efeitos dos fármacos , Fluxo Sanguíneo Regional , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/farmacologia
15.
Basic Res Cardiol ; 101(4): 301-10, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16705471

RESUMO

Efficient strategies for labelling and delivery of bone marrow derived stem cells (BMCs) are required to elucidate the cellular kinetics and therapeutic effects after BMC transfer for myocardial infarction (MI). Lineage negative (lin-) BMCs, labelled ex vivo in a simple procedure with the cell tracker dye tetramethyl-rhodamine (TAMRA), were reliably detected by fluorescence microscopy with higher specificity than retroviral enhanced green fluorescence protein (EGFP) marking and detection. Only few cells entered the ischemic myocardium after intravenous (i.v.) application, but this number increased more than 18-fold after transcoronary delivery. Time course and kinetic analysis over 12 h revealed that myocardial colonization seems to be a biphasic process of first order decay with different elimination half-lives. Most cells are eliminated rapidly during the first 2 h (t1/2 40 min), but the remaining cells are retained significantly longer in the ischemic heart (t1/2 5.2 h). In contrast, BMC colonization of the spleen increased rather in a linear fashion. Although transcoronary BMC transfusion did not alter infarct size, it increased capillary density in the infarct border zone and improved LV function 4 weeks after MI. In conclusion, BMCs delivered by transcoronary injection increase capillary density and improve LV function after MI although homing to the ischemic heart is only transient.


Assuntos
Transplante de Medula Óssea/métodos , Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Animais , Células da Medula Óssea/fisiologia , Contagem de Células , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reperfusão Miocárdica , Rodaminas
16.
Eur J Heart Fail ; 7(5): 722-9, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16158493

RESUMO

BACKGROUND: Transplantation of bone marrow derived adult stem cells (BMC) improves cardiac function after acute myocardial infarction (MI). However, the cell population mediating myocardial recovery and the fate of the transplanted cells are still controversial. AIMS: We determined the effects of Sca-1+ c-kit+ lin- haematopoietic BMC on cardiac function after MI and the cell fate after transplantation. METHODS: Sca-1+ c-kit+ lin- BMC of male donor C57BL/6 mice were transplanted by intravenous injection into syngenic females after permanent MI. LV dimensions and function were determined by echocardiography and cardiac magnetic resonance imaging, transplanted BMC were identified by Y chromosome DNA in situ hybridization. RESULTS: BMC treatment completely prevented LV dilation (LV end-diastolic volume BMC 70 +/- 16 microl vs. control 122 +/- 41 microl; p < 0.05) and improved fractional shortening (BMC 22.9 +/- 8% vs. control 15.4 +/- 8.4%; p < 0.05) and ejection fraction BMC 68.2 +/- 6.6% vs. control 52 +/- 14.3%; p < 0.05) as early as 3 days after transplantation, but did not decrease infarct size (BMC 27 +/- 6% vs. control 28 +/- 7%, p = n.s.). After 4 weeks, only sporadic cells of male origin were identified in infarcted hearts (< 0.01% of periinfarct cells). CONCLUSION: Intravenous injection of Sca-1+ c-kit+ lin- in BMC after MI improves LV dimensions and function without evidence for long term engraftment.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Infarto do Miocárdio/cirurgia , Animais , Antígenos Ly , Dilatação Patológica , Feminino , Ventrículos do Coração/patologia , Células-Tronco Hematopoéticas , Hibridização In Situ , Imageamento por Ressonância Magnética , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Função Ventricular Esquerda
17.
Circulation ; 111(14): 1826-32, 2005 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-15809373

RESUMO

BACKGROUND: Notch signaling influences binary cell fate decisions in a variety of tissues. The Notch1 receptor is widely expressed during embryogenesis and is essential for embryonic development. Loss of global Notch1 function results in early embryonic lethality, but the cell type responsible for this defect is not known. Here, we identify the endothelium as the primary target tissue affected by Notch1 signaling. METHODS AND RESULTS: We generated an endothelium-specific deletion of Notch1 using Tie2Cre and conditional Notch1(flox/flox) mice. Mutant embryos lacking endothelial Notch1 died at approximately embryonic day 10.5 with profound vascular defects in placenta, yolk sac, and embryo proper, whereas heterozygous deletion had no effect. In yolk sacs of mutant embryos, endothelial cells formed a primary vascular plexus indicative of intact vasculogenesis but failed to induce the secondary vascular remodeling required to form a mature network of well-organized large and small blood vessels, which demonstrates a defect in angiogenesis. These vascular defects were also evident in the placenta, where blood vessels failed to invade the placental labyrinth, and in the embryo proper, where defective blood vessel maturation led to pericardial and intersomitic hemorrhage. Enhanced activation of caspase-3 was detected in endothelial and neural cells of mutant mice, which resulted in enhanced apoptotic degeneration of somites and the neural tube. CONCLUSIONS: These findings recapitulate the vascular phenotype of global Notch1-/- mutants and indicate an essential cell-autonomous role of Notch1 signaling in the endothelium during vascular development. These results may have important clinical implications with regard to Notch1 signaling in adult angiogenesis.


Assuntos
Endotélio Vascular/fisiologia , Neovascularização Fisiológica , Receptores de Superfície Celular/fisiologia , Fatores de Transcrição/fisiologia , Animais , Apoptose , Caspase 3 , Caspases/metabolismo , Perda do Embrião , Embrião de Mamíferos , Endotélio Vascular/química , Endotélio Vascular/embriologia , Genótipo , Hemorragia/etiologia , Camundongos , Camundongos Mutantes , Defeitos do Tubo Neural/etiologia , Defeitos do Tubo Neural/patologia , Placenta/irrigação sanguínea , Receptor Notch1 , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Transdução de Sinais , Somitos/patologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
19.
J Mol Med (Berl) ; 81(3): 168-74, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12682725

RESUMO

Many cellular responses to corticosteroids involve the transcriptional modulation of target genes by a prototypical nuclear receptor, the glucocorticoid receptor (GR). In the classic model of steroid hormone action GR acts as ligand-dependent transcription factor by either activating or repressing gene expression through direct interactions with DNA or other transcription factors. Recent evidence suggests an important role for nontranscriptional effects of GR in the vascular system. The nontranscriptional actions of GR involve the rapid activation of protein kinases, such as phosphatidylinositol-3 kinase and Akt, leading to the activation of endothelial nitric oxide synthase. This novel pathway of steroid hormone action protects against ischemic injury by augmenting blood flow and decreasing vascular inflammation.


Assuntos
Proteínas Serina-Treonina Quinases , Receptores de Glucocorticoides/metabolismo , Transcrição Gênica , Corticosteroides/metabolismo , Corticosteroides/uso terapêutico , DNA/metabolismo , Ativação Enzimática , Humanos , Isquemia/tratamento farmacológico , Isquemia/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptores de Glucocorticoides/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA