Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 40(4): e87-e104, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32078368

RESUMO

OBJECTIVE: Impaired ALK1 (activin receptor-like kinase-1)/Endoglin/BMP9 (bone morphogenetic protein 9) signaling predisposes to arteriovenous malformations (AVMs). Activation of SMAD1/5 signaling can be enhanced by shear stress. In the genetic disease hereditary hemorrhagic telangiectasia, which is characterized by arteriovenous malformations, the affected receptors are those involved in the activation of mechanosensitive SMAD1/5 signaling. To elucidate how genetic and mechanical signals interact in AVM development, we sought to identify targets differentially regulated by BMP9 and shear stress. Approach and Results: We identify Cx37 (Connexin37) as a differentially regulated target of ligand-induced and mechanotransduced SMAD1/5 signaling. We show that stimulation of endothelial cells with BMP9 upregulated Cx37, whereas shear stress inhibited this expression. This signaling was SMAD1/5-dependent, and in the absence of SMAD1/5, there was an inversion of the expression pattern. Ablated SMAD1/5 signaling alone caused AVM-like vascular malformations directly connecting the dorsal aorta to the inlet of the heart. In yolk sacs of mouse embryos with an endothelial-specific compound heterozygosity for SMAD1/5, addition of TNFα (tumor necrosis factor-α), which downregulates Cx37, induced development of these direct connections bypassing the yolk sac capillary bed. In wild-type embryos undergoing vascular remodeling, Cx37 was globally expressed by endothelial cells but was absent in regions of enlarging vessels. TNFα and endothelial-specific compound heterozygosity for SMAD1/5 caused ectopic regions lacking Cx37 expression, which correlated to areas of vascular malformations. Mechanistically, loss of Cx37 impairs correct directional migration under flow conditions. CONCLUSIONS: Our data demonstrate that Cx37 expression is differentially regulated by shear stress and SMAD1/5 signaling, and that reduced Cx37 expression is permissive for capillary enlargement into shunts.


Assuntos
Malformações Arteriovenosas/genética , Conexinas/genética , Regulação para Baixo , Mecanotransdução Celular , Proteína Smad1/genética , Proteína Smad5/genética , Regulação para Cima , Receptores de Activinas Tipo II/metabolismo , Animais , Malformações Arteriovenosas/metabolismo , Malformações Arteriovenosas/patologia , Capilares/patologia , Células Cultivadas , Conexinas/metabolismo , Embrião de Mamíferos , Endoglina/metabolismo , Células Endoteliais/metabolismo , Feminino , Fator 2 de Diferenciação de Crescimento/metabolismo , Humanos , Masculino , Camundongos Knockout , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Remodelação Vascular , Proteína alfa-4 de Junções Comunicantes
2.
Arterioscler Thromb Vasc Biol ; 38(9): 2174-2183, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29930007

RESUMO

Objective- Vascular fusion represents an important mechanism of vessel enlargement during development; however, its significance in postnatal vessel enlargement is still unknown. During fusion, 2 adjoining vessels merge to share 1 larger lumen. The aim of this research was to identify the molecular mechanism responsible for vascular fusion. Approach and Results- We previously showed that both low shear stress and DAPT ( N-[ N-(3,5-difluorophenacetyl)-L-alanyl]- S-phenylglycine t-butyl ester) treatment in the embryo result in a hyperfused vascular plexus and that increasing shear stress levels could prevent DAPT-induced fusion. We, therefore, investigated vascular endothelial-cadherin (VEC) phosphorylation because this is a common downstream target of low shear stress and DAPT treatment. VEC phosphorylation increases after DAPT treatment and decreased shear stress. The increased phosphorylation occurred independent of the cleavage of the Notch intracellular domain. Increasing shear stress rescues hyperfusion by DAPT treatment by causing the association of the phosphatase vascular endothelial-protein tyrosine phosphatase with VEC, counteracting VEC phosphorylation. Finally, Src (proto-oncogene tyrosine-protein kinase Src) inhibition prevents VEC phosphorylation in endothelial cells and can rescue hyperfusion induced by low shear stress and DAPT treatment. Moesin, a VEC target that was previously reported to mediate endothelial cell rearrangement during lumenization, relocalizes to cell membranes in vascular beds undergoing hyperfusion. Conclusions- This study provides the first evidence that VEC phosphorylation, induced by DAPT treatment and low shear stress, is involved in the process of fusion during vascular remodeling.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Estresse Mecânico , Remodelação Vascular , Animais , Membrana Celular/metabolismo , Células Cultivadas , Dipeptídeos/farmacologia , Embrião de Mamíferos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fosforilação , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
3.
Differentiation ; 84(1): 62-78, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22683047

RESUMO

Congenital cardiac abnormalities are, due to their relatively high frequency and severe impact on quality of life, an important focus in cardiovascular research. Recently, various human studies have revealed a high coincidence of VEGF and NOTCH polymorphisms with cardiovascular outflow tract anomalies, such as bicuspid aortic valves and Tetralogy of Fallot, next to predisposition for cardiovascular pathologies, including atherosclerosis and aortic valve calcification. This genetic association between VEGF/NOTCH mutations and congenital cardiovascular defects in humans has been supported by substantial proof from animal models, revealing interaction of both pathways in cellular processes that are crucial for cardiac development. This review focuses on the role of VEGF and NOTCH signaling and their interplay in cardiogenesis with special interest to coronary and outflow tract development. An overview of the association between congenital malformations and VEGF/NOTCH polymorphisms in humans will be discussed along with their potential mechanisms and processes as revealed by transgenic mouse models. The molecular and cellular interaction of VEGF and subsequent Notch-signaling in these processes will be highlighted.


Assuntos
Diferenciação Celular , Cardiopatias Congênitas/genética , Coração/embriologia , Miócitos Cardíacos/metabolismo , Receptor Notch1/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Aterosclerose/genética , Vasos Coronários/embriologia , Modelos Animais de Doenças , Predisposição Genética para Doença , Valvas Cardíacas/embriologia , Humanos , Camundongos , Mutação , Miócitos Cardíacos/citologia , Polimorfismo Genético , Receptor Notch1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 31(5): 1059-65, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21330605

RESUMO

OBJECTIVE: Notch has been implicated in neointima formation as reflected by increased Notch/Jagged expression on vascular injury and the promigratory effect of Notch signaling on smooth muscle cells. Soluble Jagged-1 (sJag1) has been shown to inhibit Notch signaling in vitro; however, its capacity to suppress neointima formation remains unknown. METHODS AND RESULTS: Balloon injury of rat carotid arteries induced Notch1, Notch3, and Jagged-1 expression at days 3 and 14 postinjury. Notch signaling was activated as shown by increased expression of the Notch target gene Herp2. Adenoviral sJag1 (Ad-sJag1) transfection reduced neointima formation in carotid artery and enhanced reendothelialization, whereas adenoviral full-length Jagged-1 (Ad-Fl-Jag1) or LacZ had no effect. Injury-induced Herp2 expression was absent in vessels treated with Ad-sJag1. Consistently, Herp2 expression was reduced in Ad-sJag1-infected or recombinant sJag1 -treated coronary artery smooth muscle cells (CASMCs). Ad-sJag1 had no effect on human umbilical endothelial cell behavior, but it significantly reduced proliferation and migration of CASMCs. Overexpression of Herp2 in sJag1-treated CASMCs rescued the migratory and proliferative capacity in vitro. CONCLUSIONS: Our results demonstrate that sJag1 can inhibit neointima formation after balloon injury by decreasing smooth muscle cell proliferation and migration through interference with Notch-Herp2 signaling.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/prevenção & controle , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Receptor Notch1/metabolismo , Receptores Notch/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Túnica Íntima/metabolismo , Análise de Variância , Animais , Proteínas de Ligação ao Cálcio/genética , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Hiperplasia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Proteínas de Membrana/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Ratos , Ratos Sprague-Dawley , Receptor Notch3 , Proteínas Serrate-Jagged , Fatores de Tempo , Transfecção , Túnica Íntima/patologia
5.
J Biol Chem ; 285(52): 40681-9, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20959466

RESUMO

The DELTA like-4 ligand (DLL4) belongs to the highly conserved NOTCH family and is specifically expressed in the endothelium. DLL4 regulates crucial processes in vascular growth, including endothelial cell (EC) sprouting and arterial specification. Its expression is increased by VEGF-A. In the present study, we show that VEGF-induced DLL4 expression depends on NOTCH activation. VEGF-induced DLL4 expression was prevented by the blockage of NOTCH signaling with γ-secretase or ADAM inhibitors in human cardiac microvascular ECs. Similar to VEGF-A, recombinant DLL4 itself stimulated NOTCH signaling and resulted in up-regulation of DLL4, suggesting a positive feed-forward mechanism. These effects were abrogated by NOTCH inhibitors but not by inhibition of VEGF signaling. NOTCH activation alone suffices to induce DLL4 expression as illustrated by the positive effect of NOTCH intracellular domain (NICD)-1 or -4 overexpression. To discriminate between NICD/RBP-Jκ and FOXC2-regulated DLL4 expression, DLL4 promoter activity was assessed in promoter deletion experiments. NICD induced promoter activity was dependent on RBP-Jκ site but independent of the FOXC2 binding site. Accordingly, constitutively active FOXC2 did not affect DLL4 expression. The notion that the positive feed-forward mechanism might propagate NOTCH activation to neighboring ECs was supported by our observation that DLL4-eGFP-transfected ECs induced DLL4 expression in nontransfected cells in their vicinity. In summary, our data provide evidence for a mechanism by which VEGF or ligand-induced NOTCH signaling up-regulates DLL4 through a positive feed-forward mechanism. By this mechanism, DLL4 could propagate its own expression and enable synchronization of NOTCH expression and signaling between ECs.


Assuntos
Comunicação Celular/fisiologia , Vasos Coronários/metabolismo , Células Endoteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Receptores Notch/metabolismo , Elementos de Resposta/fisiologia , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Proteínas de Ligação ao Cálcio , Células Cultivadas , Vasos Coronários/citologia , Células Endoteliais/citologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Estrutura Terciária de Proteína , Receptores Notch/genética , Fator A de Crescimento do Endotélio Vascular/genética
6.
Arterioscler Thromb Vasc Biol ; 30(11): 2188-95, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20814017

RESUMO

OBJECTIVE: To elucidate the downstream mechanisms of vascular endothelial growth factor receptor 2 (VEGFR2), a key receptor in angiogenesis, which has been associated with atherosclerotic plaque growth and instability. METHODS AND RESULTS: By using a yeast-2-hybrid assay, we identified A Disintegrin And Metalloprotease 10 (ADAM10) as a novel binding partner of VEGFR2. ADAM10 is a metalloprotease with sheddase activity involved in cell migration; however, its exact function in endothelial cells (ECs), angiogenesis, and atherosclerosis is largely unknown. For the first time to our knowledge, we show ADAM10 expression in human atherosclerotic lesions, associated with plaque progression and neovascularization. We demonstrate ADAM10 expression and activity in ECs to be induced by VEGF; also, ADAM10 mediates the ectodomain shedding of VEGFR2. Furthermore, VEGF induces ADAM10-mediated cleavage of vascular endothelium (VE)-cadherin, which could increase vascular permeability and facilitate EC migration. Indeed, VEGF increases vascular permeability in an ADAM10- and ADAM17-dependent way; inhibition of ADAM10 reduces EC migration and chemotaxis. CONCLUSIONS: These data provide the first evidence of ADAM10 expression in atherosclerosis and neovascularization. ADAM10 plays a functional role in VEGF-induced EC function. These data open perspectives for novel therapeutic interventions in vascular diseases.


Assuntos
Proteínas ADAM/fisiologia , Secretases da Proteína Precursora do Amiloide/fisiologia , Aterosclerose/fisiopatologia , Células Endoteliais/fisiologia , Proteínas de Membrana/fisiologia , Neovascularização Patológica/fisiopatologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Proteínas ADAM/biossíntese , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/biossíntese , Aterosclerose/metabolismo , Células Cultivadas , Progressão da Doença , Humanos , Proteínas de Membrana/biossíntese , Neovascularização Patológica/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/fisiopatologia
7.
Aging (Albany NY) ; 13(9): 12359-12377, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33952723

RESUMO

Age-related macular degeneration (AMD) is a worldwide leading cause of blindness affecting individuals over 50 years old. The most aggressive form, wet AMD, is characterized by choroidal neovascularization (CNV) and inflammation involving microglia recruitment. By using a laser-induced CNV mouse model, we provide evidence for a key role played by miR-142-3p during CNV formation. MiR-142-3p was overexpressed in murine CNV lesions and its pharmacological inhibition decreased vascular and microglia densities by 46% and 30%, respectively. Consistently, miR-142-3p overexpression with mimics resulted in an increase of 136% and 126% of blood vessels and microglia recruitment. Interestingly, miR-142-3p expression was linked to the activation state of mouse microglia cells as determined by morphological analysis (cell solidity) through a computational method. In vitro, miR-142-3p overexpression in human microglia cells (HMC3) modulated microglia activation, as shown by CD68 levels. Interestingly, miR142-3p modulation also regulated the production of VEGF-A, the main pro-angiogenic factor. Together, these data strongly support the unprecedented importance of miR-142-3p-dependent vascular-inflammation axis during CNV progression, through microglia activation.


Assuntos
Neovascularização de Coroide/tratamento farmacológico , Neovascularização de Coroide/genética , Lasers , MicroRNAs/farmacologia , Animais , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Injeções Intravítreas/métodos , Ativação de Macrófagos/genética , Degeneração Macular/tratamento farmacológico , Degeneração Macular/metabolismo , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo
8.
Elife ; 92020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32484440

RESUMO

Mechanical force is a determinant of Notch signalling but the mechanism of force detection and its coupling to Notch are unclear. We propose a role for Piezo1 channels, which are mechanically-activated non-selective cation channels. In cultured microvascular endothelial cells, Piezo1 channel activation by either shear stress or a chemical agonist Yoda1 activated a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10), a Ca2+-regulated transmembrane sheddase that mediates S2 Notch1 cleavage. Consistent with this observation, we found Piezo1-dependent increase in the abundance of Notch1 intracellular domain (NICD) that depended on ADAM10 and the downstream S3 cleavage enzyme, γ-secretase. Conditional endothelial-specific disruption of Piezo1 in adult mice suppressed the expression of multiple Notch1 target genes in hepatic vasculature, suggesting constitutive functional importance in vivo. The data suggest that Piezo1 is a mechanism conferring force sensitivity on ADAM10 and Notch1 with downstream consequences for sustained activation of Notch1 target genes and potentially other processes.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Células Endoteliais/metabolismo , Canais Iônicos/metabolismo , Proteínas de Membrana/metabolismo , Receptor Notch1/metabolismo , Animais , Células Cultivadas , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Canais Iônicos/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Domínios Proteicos , Estresse Mecânico , Fatores de Transcrição HES-1/genética
9.
Cardiovasc Res ; 78(2): 366-75, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18093989

RESUMO

AIMS: Currently, many potential cardiac revascularization therapies target the vascular endothelial growth factor (VEGF) pathway, with variable success. Knowledge regarding the role of the VEGF/Notch/ephrinB2 cascade in (ab)normal coronary development will provide information on the subtle balance of VEGF signalling in coronary maturation and might enhance our therapeutic possibilities. METHODS AND RESULTS: The effect of VEGF isoforms on coronary development was explored in vivo using immunohistochemistry and RT-qPCR on Vegf120/120 mouse embryos solely expressing VEGF120. In vitro, human arterial coronary endothelial cells were treated with VEGF121 or VEGF165 upon which RT-qPCR was performed. In vivo, mutant coronary arterial endothelium showed a decrease in protein expression of arterial markers such as cleaved Notch1, Delta-like4, and ephrinB2 concomitant with an increase of venous markers such as chicken ovalbumin upstream promoter transcription factor II. The venous endothelium showed the opposite effect, which was confirmed on the mRNA level. In vitro, mRNA expression of arterial markers highly depended on the VEGF isoform used, with VEGF165 having the strongest effect. Also, coronary arteriogenesis was anomalous in the mouse embryos with decreased arterial and increased venous medial development as shown by staining for smooth muscle alpha-actin, Delta-like1, and Notch3. CONCLUSION: We demonstrate that VEGF isoform-related spatiotemporal cardiac alterations in the VEGF/Notch/ephrinB2 cascade lead to disturbed coronary development. This knowledge can contribute to optimizing therapies targeting VEGF signalling by enabling balancing between angiogenesis and vascular maturation.


Assuntos
Vasos Coronários/metabolismo , Coração/embriologia , Miocárdio/metabolismo , Neovascularização Fisiológica , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Vasos Coronários/embriologia , Células Endoteliais/metabolismo , Efrina-B2/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Miocárdio/patologia , Neovascularização Fisiológica/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Receptores Notch/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular/genética , Veias/embriologia , Veias/metabolismo
10.
Sci Rep ; 8(1): 11922, 2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-30093686

RESUMO

Diabetic retinopathy (DR) is one of the major complications of diabetes, which eventually leads to blindness. Up to date, no animal model has yet shown all the co-morbidities often observed in DR patients. Here, we investigated whether obese 42 weeks old ZSF1 rat, which spontaneously develops diabetes, hypertension and obesity, would be a suitable model to study DR. Although arteriolar tortuosity increased in retinas from obese as compared to lean (hypertensive only) ZSF1 rats, vascular density pericyte coverage, microglia number, vascular morphology and retinal thickness were not affected by diabetes. These results show that, despite high glucose levels, obese ZSF1 rats did not develop DR. Such observations prompted us to investigate whether the expression of genes, possibly able to contain DR development, was affected. Accordingly, mRNA sequencing analysis showed that genes (i.e. Npy and crystallins), known to have a protective role, were upregulated in retinas from obese ZSF1 rats. Lack of retina damage, despite obesity, hypertension and diabetes, makes the 42 weeks of age ZSF1 rats a suitable animal model to identify genes with a protective function in DR. Further characterisation of the identified genes and downstream pathways could provide more therapeutic targets for the treat DR.


Assuntos
Nefropatias Diabéticas/genética , Retinopatia Diabética/genética , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Hipertensão/genética , Obesidade/genética , Animais , Glicemia/metabolismo , Cristalinas/genética , Cristalinas/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Retinopatia Diabética/metabolismo , Hipertensão/metabolismo , Masculino , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Obesidade/metabolismo , Ratos , Retina/metabolismo , Retina/patologia
11.
Sci Rep ; 7: 43817, 2017 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-28272478

RESUMO

Erythro-myeloid progenitors (EMPs) were recently described to arise from the yolk sac endothelium, just prior to vascular remodeling, and are the source of adult/post-natal tissue resident macrophages. Questions remain, however, concerning whether EMPs differentiate directly from the endothelium or merely pass through. We provide the first evidence in vivo that EMPs can emerge directly from endothelial cells (ECs) and demonstrate a role for these cells in vascular development. We find that EMPs express most EC markers but late EMPs and EMP-derived cells do not take up acetylated low-density lipoprotein (AcLDL), as ECs do. When the endothelium is labelled with AcLDL before EMPs differentiate, EMPs and EMP-derived cells arise that are AcLDL+. If AcLDL is injected after the onset of EMP differentiation, however, the majority of EMP-derived cells are not double labelled. We find that cell division precedes entry of EMPs into circulation, and that blood flow facilitates the transition of EMPs from the endothelium into circulation in a nitric oxide-dependent manner. In gain-of-function studies, we inject the CSF1-Fc ligand in embryos and found that this increases the number of CSF1R+ cells, which localize to the venous plexus and significantly disrupt venous remodeling. This is the first study to definitively establish that EMPs arise from the endothelium in vivo and show a role for early myeloid cells in vascular development.


Assuntos
Células Endoteliais/citologia , Células Precursoras Eritroides/citologia , Células-Tronco Embrionárias Murinas/citologia , Células Progenitoras Mieloides/citologia , Remodelação Vascular , Saco Vitelino/citologia , Animais , Células Endoteliais/metabolismo , Células Precursoras Eritroides/metabolismo , Feminino , Hematopoese , Lipoproteínas LDL/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Células-Tronco Embrionárias Murinas/metabolismo , Células Progenitoras Mieloides/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Saco Vitelino/irrigação sanguínea , Saco Vitelino/embriologia
12.
Cardiovasc Res ; 109(2): 196-203, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26645978

RESUMO

Hereditary haemorrhagic telangiectasia (HHT) is characterized by the development of arteriovenous malformations--enlarged shunts allowing arterial flow to bypass capillaries and enter directly into veins. HHT is caused by mutations in ALK1 or Endoglin; however, the majority of arteriovenous malformations are idiopathic and arise spontaneously. Idiopathic arteriovenous malformations differ from those due to loss of ALK1 in terms of both location and disease progression. Furthermore, while arteriovenous malformations in HHT and Alk1 knockout models have decreased NOTCH signalling, some idiopathic arteriovenous malformations have increased NOTCH signalling. The pathogenesis of these lesions also differs, with loss of ALK1 causing expansion of the shunt through proliferation, and NOTCH gain of function inducing initial shunt enlargement by cellular hypertrophy. Hence, we propose that idiopathic arteriovenous malformations are distinct from those of HHT. In this review, we explore the role of ALK1-NOTCH interactions in the development of arteriovenous malformations and examine a possible role of two signalling pathways downstream of ALK1, TMEM100 and IDs, in the development of arteriovenous malformations in HHT. A nuanced understanding of the precise molecular mechanisms underlying idiopathic and HHT-associated arteriovenous malformations will allow for development of targeted treatments for these lesions.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Malformações Arteriovenosas/genética , Capilares/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Telangiectasia Hemorrágica Hereditária/genética , Animais , Humanos , Receptores Notch/genética
13.
Drug Deliv ; 23(8): 2919-2926, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26651867

RESUMO

CONTEXT: After arterial occlusion, diametrical growth of pre-existing natural bypasses around the obstruction, i.e. arteriogenesis, is the body's main coping mechanism. We have shown before that continuous infusion of chemokine (C-X-C motif) ligand 1 (CXCL1) promotes arteriogenesis in a rodent hind limb ischemia model. OBJECTIVE: For clinical translation of these positive results, we developed a new administration strategy of local and sustained delivery. Here, we investigate the therapeutic potential of CXCL1 in a drug delivery system based on microspheres. MATERIALS AND METHODS: We generated poly(ester amide) (PEA) microspheres loaded with CXCL1 and evaluated them in vitro for cellular toxicity and chemokine release characteristics. In vivo, murine femoral arteries were ligated and CXCL1 was administered either intra-arterially via osmopump or intramuscularly encapsulated in biodegradable microspheres. Perfusion recovery was measured with Laser-Doppler. RESULTS: The developed microspheres were not cytotoxic and displayed a sustained chemokine release up to 28 d in vitro. The amount of released CXCL1 was 100-fold higher than levels in native ligated hind limb. Also, the CXCL1-loaded microspheres significantly enhanced perfusion recovery at day 7 after ligation compared with both saline and non-loaded conditions (55.4 ± 5.0% CXCL1-loaded microspheres versus 43.1 ± 4.5% non-loaded microspheres; n = 8-9; p < 0.05). On day 21 after ligation, the CXCL1-loaded microspheres performed even better than continuous CXCL1 administration (102.1 ± 4.4% CXCL1-loaded microspheres versus 85.7 ± 4.8% CXCL1 osmopump; n = 9; p < 0.05). CONCLUSION: Our results demonstrate a proof of concept that sustained, local delivery of CXCL1 encapsulated in PEA microspheres provides a new tool to stimulate arteriogenesis in vivo.


Assuntos
Quimiocina CXCL1/administração & dosagem , Artéria Femoral/efeitos dos fármacos , Animais , Preparações de Ação Retardada/administração & dosagem , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/métodos , Membro Posterior/irrigação sanguínea , Isquemia/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Poliaminas/química , Poliésteres/química
14.
Regen Med ; 8(6): 759-70, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24147531

RESUMO

Vascularization of engineered tissues is critical for success. Adequate and physiologically regulated blood supply is important for viability of the implanted tissue but even more important for the proper function of parenchymal cells, which is the desired clinical outcome for most applications in regenerative medicine. Several methods are being developed to stimulate revascularization of engineered tissue. Prevascularized scaffolds with a hierarchical vascular pattern, allowing surgical hook-up of the inflow and outflow tracts, that are already preseeded and cultured with primary vascular cells or precursors will be required for larger tissues or tissues with an immediate high metabolism, such as myocardium. The preimplantation presence of a mature vasculature will improve differentiation and maturation of the parenchyma, thus meeting the functional demands of the host. This may also be true for smaller or metabolically less-active tissues, yet for viability and immediate function they may rely on facilitated postimplantation ingrowth of the host vasculature.


Assuntos
Neovascularização Fisiológica , Engenharia Tecidual/métodos , Humanos , Alicerces Teciduais/química
15.
Stem Cells Int ; 2012: 805602, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22550518

RESUMO

The development of the vascular system begins with the formation of hemangioblastic cells, hemangioblasts, which organize in blood islands in the yolk sac. The hemangioblasts differentiate into hematopoietic and angioblastic cells. Subsequently, the hematopoietic line will generate blood cells, whereas the angioblastic cells will give rise to vascular endothelial cells (ECs). In response to specific molecular and hemodynamic stimuli, ECs will acquire either arterial or venous identity. Recruitment towards the endothelial tubes and subsequent differentiation of pericyte and/or vascular smooth muscle cells (vSMCs) takes place and the mature vessel is formed. The Notch signaling pathway is required for determining the arterial program of both endothelial and smooth muscle cells; however, it is simultaneously involved in the generation of hematopoietic stem cells (HSCs), which will give rise to hematopoietic cells. Notch signaling also regulates the function of endothelial progenitor cells (EPCs), which are bone-marrow-derived cells able to differentiate into ECs and which could be considered the adult correlate of the angioblast. In addition, Notch signaling has been reported to control sprouting angiogenesis during blood vessels formation in the adult. In this paper we discuss the physiological role of Notch in vascular development, providing an overview on the involvement of Notch in vascular biology from hematopoietic stem cell to adaptive neovascularization in the adult.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA