Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(5): e0302926, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38718095

RESUMO

Zinc Finger MIZ-Type Containing 1 (Zmiz1), also known as ZIMP10 or RAI17, is a transcription cofactor and member of the Protein Inhibitor of Activated STAT (PIAS) family of proteins. Zmiz1 is critical for a variety of biological processes including vascular development. However, its role in the lymphatic vasculature is unknown. In this study, we utilized human dermal lymphatic endothelial cells (HDLECs) and an inducible, lymphatic endothelial cell (LEC)-specific Zmiz1 knockout mouse model to investigate the role of Zmiz1 in LECs. Transcriptional profiling of ZMIZ1-deficient HDLECs revealed downregulation of genes crucial for lymphatic vessel development. Additionally, our findings demonstrated that loss of Zmiz1 results in reduced expression of proliferation and migration genes in HDLECs and reduced proliferation and migration in vitro. We also presented evidence that Zmiz1 regulates Prox1 expression in vitro and in vivo by modulating chromatin accessibility at Prox1 regulatory regions. Furthermore, we observed that loss of Zmiz1 in mesenteric lymphatic vessels significantly reduced valve density. Collectively, our results highlight a novel role of Zmiz1 in LECs and as a transcriptional regulator of Prox1, shedding light on a previously unknown regulatory factor in lymphatic vascular biology.


Assuntos
Proliferação de Células , Células Endoteliais , Proteínas de Homeodomínio , Vasos Linfáticos , Fatores de Transcrição , Proteínas Supressoras de Tumor , Animais , Humanos , Camundongos , Movimento Celular/genética , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Linfangiogênese/genética , Vasos Linfáticos/metabolismo , Vasos Linfáticos/citologia , Camundongos Knockout , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
2.
Front Psychiatry ; 15: 1375492, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38686122

RESUMO

Neurodevelopmental disorders (NDDs) are a class of pathologies arising from perturbations in brain circuit formation and maturation with complex etiological triggers often classified as environmental and genetic. Neuropsychiatric conditions such as autism spectrum disorders (ASD), intellectual disability (ID), and attention deficit hyperactivity disorders (ADHD) are common NDDs characterized by their hereditary underpinnings and inherent heterogeneity. Genetic risk factors for NDDs are increasingly being identified in non-coding regions and proteins bound to them, including transcriptional regulators and chromatin remodelers. Importantly, de novo mutations are emerging as important contributors to NDDs and neuropsychiatric disorders. Recently, de novo mutations in transcriptional co-factor Zmiz1 or its regulatory regions have been identified in unrelated patients with syndromic ID and ASD. However, the role of Zmiz1 in brain development is unknown. Here, using publicly available databases and a Zmiz1 mutant mouse model, we reveal that Zmiz1 is highly expressed during embryonic brain development in mice and humans, and though broadly expressed across the brain, Zmiz1 is enriched in areas prominently impacted in ID and ASD such as cortex, hippocampus, and cerebellum. We investigated the relationship between Zmiz1 structure and pathogenicity of protein variants, the epigenetic marks associated with Zmiz1 regulation, and protein interactions and signaling pathways regulated by Zmiz1. Our analysis reveals that Zmiz1 regulates multiple developmental processes, including neurogenesis, neuron connectivity, and synaptic signaling. This work paves the way for future studies on the functions of Zmiz1 and highlights the importance of combining analysis of mouse models and human data.

3.
bioRxiv ; 2023 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-37503058

RESUMO

Zinc Finger MIZ-Type Containing 1 (Zmiz1), also known as ZIMP10 or RAI17, is a transcription cofactor and member of the Protein Inhibitor of Activated STAT (PIAS) family of proteins. Zmiz1 is critical for a variety of biological processes including vascular development. However, its role in the lymphatic vasculature is unknown. In this study, we utilized human dermal lymphatic endothelial cells (HDLECs) and an inducible, lymphatic endothelial cell (LEC)-specific Zmiz1 knockout mouse model to investigate the role of Zmiz1 in LECs. Transcriptional profiling of Zmiz1-deficient HDLECs revealed downregulation of genes crucial for lymphatic vessel development. Additionally, our findings demonstrated that loss of Zmiz1 results in reduced expression of proliferation and migration genes in HDLECs and reduced proliferation and migration in vitro. We also presented evidence that Zmiz1 regulates Prox1 expression in vitro and in vivo by modulating chromatin accessibility at Prox1 regulatory regions. Furthermore, we observed that loss of Zmiz1 in mesenteric lymphatic vessels significantly reduced valve density. Collectively, our results highlight a novel role of Zmiz1 in LECs and as a transcriptional regulator of Prox1, shedding light on a previously unknown regulatory factor in lymphatic vascular biology.

4.
Front Physiol ; 14: 1165379, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37324380

RESUMO

Proper vascular formation is regulated by multiple signaling pathways. The vascular endothelial growth factor (VEGF) signaling promotes endothelial proliferation. Notch and its downstream targets act to lead endothelial cells toward an arterial fate through regulation of arterial gene expression. However, the mechanisms of how endothelial cells (ECs) in the artery maintain their arterial characteristics remain unclear. Here, we show that PRDM16 (positive regulatory domain-containing protein 16), a zinc finger transcription factor, is expressed in arterial ECs, but not venous ECs in developing embryos and neonatal retinas. Endothelial-specific deletion of Prdm16 induced ectopic venous marker expression in the arterial ECs and reduced vascular smooth muscle cell (vSMC) recruitment around arteries. Whole-genome transcriptome analysis using isolated brain ECs show that the expression of Angpt2 (encoding ANGIOPOIETIN2, which inhibits vSMC recruitment) is upregulated in the Prdm16 knockout ECs. Conversely, forced expression of PRDM16 in venous ECs is sufficient to induce arterial gene expression and repress the ANGPT2 level. Together, these results reveal an arterial cell-autonomous function for PRDM16 in suppressing venous characteristics in arterial ECs.

5.
Arterioscler Thromb Vasc Biol ; 43(8): 1384-1403, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37288572

RESUMO

BACKGROUND: Hereditary hemorrhagic telangiectasia (HHT) is a vascular disorder characterized by arteriovenous malformations and blood vessel enlargements. However, there are no effective drug therapies to combat arteriovenous malformation formation in patients with HHT. Here, we aimed to address whether elevated levels of ANG2 (angiopoietin-2) in the endothelium is a conserved feature in mouse models of the 3 major forms of HHT that could be neutralized to treat brain arteriovenous malformations and associated vascular defects. In addition, we sought to identify the angiogenic molecular signature linked to HHT. METHODS: Cerebrovascular defects, including arteriovenous malformations and increased vessel calibers, were characterized in mouse models of the 3 common forms of HHT using transcriptomic and dye injection labeling methods. RESULTS: Comparative RNA sequencing analyses of isolated brain endothelial cells revealed a common, but unique proangiogenic transcriptional program associated with HHT. This included a consistent upregulation in cerebrovascular expression of ANG2 and downregulation of its receptor Tyr kinase with Ig and EGF homology domains (TIE2/TEK) in HHT mice compared with controls. Furthermore, in vitro experiments revealed TEK signaling activity was hampered in an HHT setting. Pharmacological blockade of ANG2 improved brain vascular pathologies in all HHT models, albeit to varying degrees. Transcriptomic profiling further indicated that ANG2 inhibition normalized the brain vasculature by impacting a subset of genes involved in angiogenesis and cell migration processes. CONCLUSIONS: Elevation of ANG2 in the brain vasculature is a shared trait among the mouse models of the common forms of HHT. Inhibition of ANG2 activity can significantly limit or prevent brain arteriovenous malformation formation and blood vessel enlargement in HHT mice. Thus, ANG2-targeted therapies may represent a compelling approach to treat arteriovenous malformations and vascular pathologies related to all forms of HHT.


Assuntos
Malformações Arteriovenosas , Telangiectasia Hemorrágica Hereditária , Animais , Camundongos , Telangiectasia Hemorrágica Hereditária/tratamento farmacológico , Telangiectasia Hemorrágica Hereditária/genética , Células Endoteliais/metabolismo , Angiopoietina-2/genética , Angiopoietina-2/metabolismo , Malformações Arteriovenosas/metabolismo , Fenótipo
6.
JCI Insight ; 7(19)2022 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-35998033

RESUMO

The (Pro)renin receptor ([P]RR), also known as ATP6AP2, is a single-transmembrane protein that is implicated in a multitude of biological processes. However, the exact role of ATP6AP2 during blood vessel development remains largely undefined. Here, we use an inducible endothelial cell-specific (EC-specific) Atp6ap2-KO mouse model to investigate the role of ATP6AP2 during both physiological and pathological angiogenesis in vivo. We observed that postnatal deletion of Atp6ap2 in ECs results in cell migration defects, loss of tip cell polarity, and subsequent impairment of retinal angiogenesis. In vitro, Atp6ap2-deficient ECs similarly displayed reduced cell migration, impaired sprouting, and defective cell polarity. Transcriptional profiling of ECs isolated from Atp6ap2 mutant mice further indicated regulatory roles in angiogenesis, cell migration, and extracellular matrix composition. Mechanistically, we provided evidence that expression of various extracellular matrix components is controlled by ATP6AP2 via the ERK pathway. Furthermore, Atp6ap2-deficient retinas exhibited reduced revascularization in an oxygen-induced retinopathy model. Collectively, our results demonstrate a critical role of ATP6AP2 as a regulator of developmental and pathological angiogenesis.


Assuntos
Polaridade Celular , ATPases Translocadoras de Prótons , Receptores de Superfície Celular , Renina , Animais , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Oxigênio/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Receptores de Superfície Celular/metabolismo , Renina/metabolismo
7.
Microcirculation ; : e12740, 2021 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-34820962

RESUMO

OBJECTIVE: Despite the absolute requirement of Delta/Notch signaling to activate lateral inhibition during early blood vessel development, many mechanisms remain unclear about how this system is regulated. Our objective was to determine the involvement of Epsin 15 Homology Domain Containing 2 (EHD2) in delta-like ligand 4 (Dll4) endocytosis during Notch activation. APPROACH AND RESULTS: Using both in vivo and in vitro models, we demonstrate that EHD2 is a novel modulator of Notch activation in endothelial cells through controlling endocytosis of Dll4. In vitro, EHD2 localized to plasma membrane-bound Dll4 and caveolae. Chemical disruption of caveolae complexes resulted in EHD2 failing to organize around Dll4 as well as loss of Dll4 internalization. Reduced Dll4 internalization blunted Notch activation in endothelial cells. In vivo, EHD2 is primarily expressed in the vasculature, colocalizing with junctional marker VE-cadherin and Dll4. Knockout of EHD2 in zebrafish produced a significant increase in dysmorphic sprouts in zebrafish intersomitic vessels during development and a reduction in downstream Notch signaling. CONCLUSIONS: Overall, we demonstrate that EHD2 is necessary for Dll4 transcytosis and downstream Notch activation.

8.
Dev Dyn ; 249(5): 666-678, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32020697

RESUMO

BACKGROUND: Annexin A3 (Anxa3) is a member of the calcium-regulated, cell membrane-binding family of annexin proteins. We previously confirmed that Anxa3 is expressed in the endothelial lineage in vertebrates and that loss of anxa3 in Xenopus laevis leads to embryonic blood vessel defects. However, the biological function of Anxa3 in mammals is completely unknown. In order to investigate Anxa3 vascular function in mammals, we generated an endothelial cell-specific Anxa3 conditional knockout mouse model (Anxa3f/f ;Tie2-Cre). RESULTS: Anxa3f/f ;Tie2-Cre mice are born at Mendelian ratios and display morphologically normal blood vessels during development. However, loss of Anxa3 leads to artery-vein (AV) misalignment characterized by atypical AV crossovers in the postnatal and adult retina. CONCLUSIONS: Anxa3 is not essential for embryonic blood vessel formation but is required for proper parallel AV alignment in the murine retina. AV crossovers associated with Anxa3f/f ;Tie2-Cre mice are similar to AV intersections observed in patients with branch retinal vein occlusion (BRVO), although we did not observe occluded vessels. This new Anxa3 mouse model may provide a basis for understanding AV crossover formation associated with BRVO.


Assuntos
Anexina A3/metabolismo , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Retina/metabolismo , Veias/metabolismo , Animais , Anexina A3/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Masculino , Camundongos , Retina/fisiologia , Veias/fisiologia
9.
Circulation ; 139(17): 2049-2063, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30744395

RESUMO

BACKGROUND: Hereditary hemorrhagic telangiectasia is an autosomal dominant vascular disorder caused by heterozygous, loss-of-function mutations in 4 transforming growth factor beta (TGFß) pathway members, including the central transcriptional mediator of the TGFß pathway, Smad4. Loss of Smad4 causes the formation of inappropriate, fragile connections between arteries and veins called arteriovenous malformations (AVMs), which can hemorrhage leading to stroke, aneurysm, or death. Unfortunately, the molecular mechanisms underlying AVM pathogenesis remain poorly understood, and the TGFß downstream effectors responsible for hereditary hemorrhagic telangiectasia-associated AVM formation are currently unknown. METHODS: To identify potential biological targets of the TGFß pathway involved in AVM formation, we performed RNA- and chromatin immunoprecipitation-sequencing experiments on BMP9 (bone morphogenetic protein 9)-stimulated endothelial cells (ECs) and isolated ECs from a Smad4-inducible, EC-specific knockout ( Smad4-iECKO) mouse model that develops retinal AVMs. These sequencing studies identified the angiopoietin-Tek signaling pathway as a downstream target of SMAD4. We used monoclonal blocking antibodies to target a specific component in this pathway and assess its effects on AVM development. RESULTS: Sequencing studies uncovered 212 potential biological targets involved in AVM formation, including the EC surface receptor, TEK (TEK receptor tyrosine kinase) and its antagonistic ligand, ANGPT2 (angiopoietin-2). In Smad4-iECKO mice, Angpt2 expression is robustly increased, whereas Tek levels are decreased, resulting in an overall reduction in angiopoietin-Tek signaling. We provide evidence that SMAD4 directly represses Angpt2 transcription in ECs. Inhibition of ANGPT2 function in Smad4-deficient mice, either before or after AVMs form, prevents and alleviates AVM formation and normalizes vessel diameters. These rescue effects are attributed to a reversion in EC morphological changes, such as cell size and shape that are altered in the absence of Smad4. CONCLUSIONS: Our studies provide a novel mechanism whereby the loss of Smad4 causes increased Angpt2 transcription in ECs leading to AVM formation, increased blood vessel calibers, and changes in EC morphology in the retina. Blockade of ANGPT2 function in an in vivo Smad4 model of hereditary hemorrhagic telangiectasia alleviated these vascular phenotypes, further implicating ANGPT2 as an important TGFß downstream mediator of AVM formation. Therefore, alternative approaches that target ANGPT2 function may have therapeutic value for the alleviation of hereditary hemorrhagic telangiectasia symptoms, such as AVMs.


Assuntos
Angiopoietina-2/antagonistas & inibidores , Malformações Arteriovenosas/prevenção & controle , Proteína Smad4/deficiência , Telangiectasia Hemorrágica Hereditária/complicações , Angiopoietina-2/biossíntese , Angiopoietina-2/genética , Animais , Malformações Arteriovenosas/etiologia , Malformações Arteriovenosas/metabolismo , Malformações Arteriovenosas/patologia , Tamanho Celular , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Receptor TIE-2/fisiologia , Transdução de Sinais , Proteína Smad4/genética , Proteína Smad4/fisiologia , Telangiectasia Hemorrágica Hereditária/genética , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo
10.
J Vasc Res ; 55(3): 125-135, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29779031

RESUMO

BACKGROUND: The development of models that incorporate intact microvascular networks enables the investigation of multicellular dynamics during angiogenesis. Our laboratory introduced the rat mesentery culture model as such a tool, which would be enhanced with mouse tissue. Since mouse mesentery is avascular, an alternative is mouse mesometrium, the connective tissue of uterine horns. The study's objective was to demonstrate that mouse mesometrium contains microvascular networks that can be cultured to investigate multicellular dynamics during angiogenesis. METHODS: Harvested mesometrium tissues from C57Bl/6 female mice were cultured in media with serum for up to 7 days. PECAM, NG2, αSMA, and LYVE-1 labeling identified endothelial cells, pericytes, smooth muscle cells, and lymphatic endothelial cells, respectively. RESULTS: These cells comprised microvascular networks with arterioles, venules, and capillaries. Compared to day 0, capillary sprouts per vascular length were increased by 3 and 5 days in culture (day 0, 0.08 ± 0.01; day 3, 3.19 ± 0.78; day 5, 2.49 ± 0.05 sprouts/mm; p < 0.05). Time-lapse imaging of cultured tissues from FlkEGFP mice showcases the use of the model for lineage studies. The impact is supported by the identification of endothelial cell jumping from one sprout to another. CONCLUSION: These results introduce a novel culture model for investigating multicellular dynamics during angiogenesis in real-time ex vivo microvascular networks.


Assuntos
Microvasos/fisiologia , Neovascularização Fisiológica , Útero/irrigação sanguínea , Actinas/metabolismo , Animais , Antígenos/metabolismo , Biomarcadores/metabolismo , Feminino , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Membrana Transportadoras , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Modelos Animais , Neovascularização Fisiológica/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteoglicanas/metabolismo , Fatores de Tempo , Imagem com Lapso de Tempo , Técnicas de Cultura de Tecidos , Fator A de Crescimento do Endotélio Vascular/farmacologia
11.
Angiogenesis ; 21(2): 363-380, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29460088

RESUMO

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant vascular disorder that leads to abnormal connections between arteries and veins termed arteriovenous malformations (AVM). Mutations in TGFß pathway members ALK1, ENG and SMAD4 lead to HHT. However, a Smad4 mouse model of HHT does not currently exist. We aimed to create and characterize a Smad4 endothelial cell (EC)-specific, inducible knockout mouse (Smad4f/f;Cdh5-CreERT2) that could be used to study AVM development in HHT. We found that postnatal ablation of Smad4 caused various vascular defects, including the formation of distinct AVMs in the neonate retina. Our analyses demonstrated that increased EC proliferation and size, altered mural cell coverage and distorted artery-vein gene expression are associated with Smad4 deficiency in the vasculature. Furthermore, we show that depletion of Smad4 leads to decreased Vegfr2 expression, and concurrent loss of endothelial Smad4 and Vegfr2 in vivo leads to AVM enlargement. Our work provides a new model in which to study HHT-associated phenotypes and links the TGFß and VEGF signaling pathways in AVM pathogenesis.


Assuntos
Malformações Arteriovenosas , Células Endoteliais , Proteínas do Olho/metabolismo , Vasos Retinianos , Proteína Smad4/deficiência , Telangiectasia Hemorrágica Hereditária , Animais , Malformações Arteriovenosas/genética , Malformações Arteriovenosas/metabolismo , Malformações Arteriovenosas/patologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Proteínas do Olho/genética , Camundongos , Camundongos Knockout , Vasos Retinianos/anormalidades , Vasos Retinianos/metabolismo , Telangiectasia Hemorrágica Hereditária/genética , Telangiectasia Hemorrágica Hereditária/metabolismo , Telangiectasia Hemorrágica Hereditária/patologia
12.
Oncotarget ; 8(29): 47076-47089, 2017 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-28423364

RESUMO

Helicobacter pylori infection triggers a cascade of inflammatory stages that may lead to the appearance of non-atrophic gastritis, multifocal atrophic, intestinal metaplasia, dysplasia, and cancer. Deleted in malignant brain tumors 1 (DMBT1) belongs to the group of secreted scavenger receptor cysteine-rich proteins and is considered to be involved in host defense by binding to pathogens. Initial studies showed its deletion and loss of expression in a variety of tumors but the role of this gene in tumor development is not completely understood. Here, we examined the role of DMBT1 in gastric precancerous lesions in Caucasian, African American and Hispanic individuals as well as in the development of gastric pathology in a mouse model of H. pylori infection. We found that in 3 different populations, mucosal DMBT1 expression was significantly increased (2.5 fold) in individuals with dysplasia compared to multifocal atrophic gastritis without intestinal metaplasia; the increase was also observed in individuals with advanced gastritis and positive H. pylori infection. In our animal model, H. pylori infection of Dmbt1-/- mice resulted in significantly higher levels of gastritis, more extensive mucous metaplasia and reduced Il33 expression levels in the gastric mucosa compared to H. pylori-infected wild type mice. Our data in the animal model suggest that in response to H. pylori infection DMBT1 may mediate mucosal protection reducing the risk of developing gastric precancerous lesions. However, the increased expression in human gastric precancerous lesions points to a more complex role of DMBT1 in gastric carcinogenesis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Lesões Pré-Cancerosas/genética , Receptores de Superfície Celular/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Animais , Proteínas de Ligação ao Cálcio , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Etnicidade/genética , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Perfilação da Expressão Gênica , Estudos de Associação Genética , Infecções por Helicobacter/complicações , Humanos , Camundongos , Camundongos Knockout , Estadiamento de Neoplasias , Proteínas Supressoras de Tumor
13.
Gene Expr Patterns ; 23-24: 22-31, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28167138

RESUMO

The murine retina has become an ideal model to study blood vessel formation. Blood vessels in the retina undergo various processes, including remodeling and differentiation, to form a stereotypical network that consists of precisely patterned arteries and veins. This model presents a powerful tool for understanding many different aspects of angiogenesis including artery and vein (AV) cell fate acquisition and differentiation. However, characterization of AV differentiation has been largely unexplored in the mouse retinal model. In this study, we describe the expression of previously established AV markers and assess arteriovenous acquisition and identity in the murine neonatal retina. Using in situ hybridization and immunofluorescent antibody staining techniques, we analyzed numerous AV differentiation markers such as EphB4-EphrinB2 and members of the Notch pathway. We find that at postnatal day 3 (P3), when blood vessels are beginning to populate the retina, AV identity is not immediately established. However, by P5 expression of many molecular identifiers of arteries and veins become restricted to their respective vessel types. This molecular distinction is more obvious at P7 and remains unchanged through P9. Overall, these studies indicate that, similar to the embryo, acquisition of AV identity occurs in a step-wise process and is largely established by P7 during retina development.


Assuntos
Artérias/crescimento & desenvolvimento , Retina/crescimento & desenvolvimento , Veias/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Proteínas Sanguíneas/análise , Proteínas Sanguíneas/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas In Vitro , Camundongos , Transcriptoma
14.
Dev Biol ; 406(2): 222-34, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26321050

RESUMO

Wnt signaling is essential to many events during organogenesis, including the development of the mammalian lung. The Wnt family member Wnt4 has been shown to be required for the development of kidney, gonads, thymus, mammary and pituitary glands. Here, we show that Wnt4 is critical for proper morphogenesis and growth of the respiratory system. Using in situ hybridization in mouse embryos, we identify a previously uncharacterized site of Wnt4 expression in the anterior trunk mesoderm. This expression domain initiates as early as E8.25 in the mesoderm abutting the tracheoesophageal endoderm, between the fusing dorsal aortae and the heart. Analysis of Wnt4(-/-) embryos reveals severe lung hypoplasia and tracheal abnormalities; however, aortic fusion and esophageal development are unaffected. We find decreased cell proliferation in Wnt4(-/-) lung buds, particularly in tip domains. In addition, we observe reduction of the important lung growth factors Fgf9, Fgf10, Sox9 and Wnt2 in the lung bud during early stages of organogenesis, as well as decreased tracheal expression of the progenitor factor Sox9. Together, these data reveal a previously unknown role for the secreted protein Wnt4 in respiratory system development.


Assuntos
Proliferação de Células/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Pulmão/embriologia , Via de Sinalização Wnt/fisiologia , Proteína Wnt4/metabolismo , Animais , Primers do DNA/genética , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator 9 de Crescimento de Fibroblastos/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOX9/metabolismo , Proteína Wnt2/metabolismo , Proteína Wnt4/genética
15.
Development ; 142(17): 3058-70, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26253403

RESUMO

The Rho family of small GTPases has been shown to be required in endothelial cells (ECs) during blood vessel formation. However, the underlying cellular events controlled by different GTPases remain unclear. Here, we assess the cellular mechanisms by which Cdc42 regulates mammalian vascular morphogenesis and maintenance. In vivo deletion of Cdc42 in embryonic ECs (Cdc42(Tie2KO)) results in blocked lumen formation and endothelial tearing, leading to lethality of mutant embryos by E9-10 due to failed blood circulation. Similarly, inducible deletion of Cdc42 (Cdc42(Cad5KO)) at mid-gestation blocks angiogenic tubulogenesis. By contrast, deletion of Cdc42 in postnatal retinal vessels leads to aberrant vascular remodeling and sprouting, as well as markedly reduced filopodia formation. We find that Cdc42 is essential for organization of EC adhesion, as its loss results in disorganized cell-cell junctions and reduced focal adhesions. Endothelial polarity is also rapidly lost upon Cdc42 deletion, as seen by failed localization of apical podocalyxin (PODXL) and basal actin. We link observed failures to a defect in F-actin organization, both in vitro and in vivo, which secondarily impairs EC adhesion and polarity. We also identify Cdc42 effectors Pak2/4 and N-WASP, as well as the actomyosin machinery, to be crucial for EC actin organization. This work supports the notion of Cdc42 as a central regulator of the cellular machinery in ECs that drives blood vessel formation.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Citoesqueleto/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Neovascularização Fisiológica , Proteína cdc42 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Animais , Aorta/metabolismo , Apoptose , Vasos Sanguíneos/citologia , Adesão Celular , Polaridade Celular , Proliferação de Células , Sobrevivência Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Matriz Extracelular/metabolismo , Feminino , Deleção de Genes , Integrases/metabolismo , Camundongos Knockout , Modelos Biológicos , Gravidez , Pseudópodes/metabolismo , Receptor TIE-2/metabolismo , Vasos Retinianos/embriologia , Vasos Retinianos/metabolismo , Saco Vitelino/irrigação sanguínea , Saco Vitelino/metabolismo
16.
PLoS One ; 10(7): e0132580, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26182056

RESUMO

Annexins are a large family of calcium binding proteins that associate with cell membrane phospholipids and are involved in various cellular processes including endocytosis, exocytosis and membrane-cytoskeletal organization. Despite studies on numerous Annexin proteins, the function of Annexin A3 (Anxa3) is largely unknown. Our studies identify Anxa3 as a unique marker of the endothelial and myeloid cell lineages of Xenopus laevis during development. Anxa3 transcripts are also detected in endothelial cells (ECs) of zebrafish and mouse embryos, suggesting an important evolutionary function during formation of blood vessels. Indeed, Anxa3 loss-of-function experiments in frog embryos reveal its critical role during the morphogenesis of early blood vessels, as angioblasts in MO injected embryos fail to form vascular cords. Furthermore, in vitro experiments in mammalian cells identify a role for Anxa3 in EC migration. Our results are the first to reveal an in vivo function for Anxa3 during vascular development and represent a previously unexplored aspect of annexin biology.


Assuntos
Anexina A3/genética , Regulação da Expressão Gênica no Desenvolvimento , Neovascularização Fisiológica/genética , Animais , Anexina A3/antagonistas & inibidores , Anexina A3/metabolismo , Morte Celular , Linhagem Celular , Proliferação de Células , Embrião de Mamíferos , Embrião não Mamífero , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Microinjeções , Morfolinos/genética , Morfolinos/metabolismo , Xenopus laevis , Peixe-Zebra
17.
Curr Opin Genet Dev ; 32: 86-91, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25748252

RESUMO

The vascular system is a complex, largely stereotyped network of interconnecting and branching vessels. How thousands of vessels form at precise locations is a key question regarding vascular morphogenesis. In order to achieve this defined architecture, the embryo integrates a multitude of attractive and repulsive cues to guide and shape the developing vasculature. This review summarizes recent studies investigating the interactions between endothelial cells and signals from surrounding tissues that pattern the initial blood vessel network.


Assuntos
Vasos Sanguíneos/embriologia , Padronização Corporal/fisiologia , Células Endoteliais/fisiologia , Modelos Biológicos , Morfogênese/fisiologia , Transdução de Sinais/fisiologia , Vertebrados/embriologia , Animais , Movimento Celular/fisiologia , Microambiente Celular/fisiologia , Sinais (Psicologia) , Células Endoteliais/metabolismo , Mesoderma/embriologia , Notocorda/embriologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
PLoS One ; 8(9): e74686, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24023956

RESUMO

Vascular endothelial growth factor receptor 2 (VEGFR2) is highly expressed by lymphatic endothelial cells and has been shown to stimulate lymphangiogenesis in adult mice. However, the role VEGFR2 serves in the development of the lymphatic vascular system has not been defined. Here we use the Cre-lox system to show that the proper development of the lymphatic vasculature requires VEGFR2 expression by lymphatic endothelium. We show that Lyve-1(wt/Cre);Vegfr2(flox/flox) mice possess significantly fewer dermal lymphatic vessels than Vegfr2(flox/flox) mice. Although Lyve-1(wt/Cre);Vegfr2(flox/flox) mice exhibit lymphatic hypoplasia, the lymphatic network is functional and contains all of the key features of a normal lymphatic network (initial lymphatic vessels and valved collecting vessels surrounded by smooth muscle cells (SMCs)). We also show that Lyve-1(Cre) mice display robust Cre activity in macrophages and in blood vessels in the yolk sac, liver and lung. This activity dramatically impairs the development of blood vessels in these tissues in Lyve-1(wt/Cre);Vegfr2(flox/flox) embryos, most of which die after embryonic day14.5. Lastly, we show that inactivation of Vegfr2 in the myeloid lineage does not affect the development of the lymphatic vasculature. Therefore, the abnormal lymphatic phenotype of Lyve-1(wt/Cre);Vegfr2(flox/flox) mice is due to the deletion of Vegfr2 in the lymphatic vasculature not macrophages. Together, this work demonstrates that VEGFR2 directly promotes the expansion of the lymphatic network and further defines the molecular mechanisms controlling the development of the lymphatic vascular system.


Assuntos
Vasos Linfáticos/embriologia , Neovascularização Fisiológica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Endoteliais/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Fígado/irrigação sanguínea , Fígado/embriologia , Pulmão/irrigação sanguínea , Pulmão/embriologia , Vasos Linfáticos/metabolismo , Proteínas de Membrana Transportadoras , Camundongos , Sobrevivência de Tecidos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Saco Vitelino/irrigação sanguínea , Saco Vitelino/embriologia
19.
Dev Dyn ; 242(5): 580-90, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23444297

RESUMO

BACKGROUND: Neuronal guidance cues influence endothelial cell (EC) behavior to shape the embryonic vascular system. The repulsive neuronal guidance cue, Semaphorin 3E (Sema3E), is critical for creating avascular zones that instruct and subsequently pattern the first embryonic vessels, the paired dorsal aortae (DA). Sema3E(-) (/) (-) embryos develop highly branched plexus-like vessels during vasculogenesis, instead of smooth paired vessels. Unexpectedly, despite these severe DA patterning defects, mutant mice are viable throughout adulthood. RESULTS: Examination of Sema3E(-) (/) (-) mice reveals that the plexus-like DA resolve into single, unbranched vessels between embryonic day (E) E8.25 and E8.75. Although fusion of Sema3E(-) (/) (-) DA occurs slightly earlier than in heterozygotes, the DA are otherwise indistinguishable, suggesting a complete "rescue" in their development. Resolution of the DA null plexuses occurs by remodeling rather than by means of changes in cell proliferation or death. CONCLUSIONS: Normalization of Sema3E(-) (/) (-) DA patterning defects demonstrates resilience of embryonic vascular patterning programs. Additional repulsive guidance cues within the lateral plate mesoderm likely re-establish avascular zones lost in Sema3E(-) (/) (-) embryos and guide resolution of mutant plexus into branchless, parallel aortae. Our observations explain how Sema3E(-) (/) (-) mice survive throughout development and into adulthood, despite severe initial vascular defects.


Assuntos
Aorta/embriologia , Padronização Corporal/genética , Glicoproteínas/genética , Cardiopatias Congênitas/genética , Proteínas de Membrana/genética , Neovascularização Fisiológica/fisiologia , Animais , Aorta/anormalidades , Proteínas do Citoesqueleto , Embrião de Mamíferos , Endotélio Vascular/anormalidades , Endotélio Vascular/embriologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Idade Gestacional , Glicoproteínas/metabolismo , Glicoproteínas/fisiologia , Cardiopatias Congênitas/mortalidade , Cardiopatias Congênitas/fisiopatologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Neovascularização Fisiológica/genética , Semaforinas , Fatores de Tempo
20.
Circ Res ; 110(1): 34-46, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22076636

RESUMO

RATIONALE: Positive signals, such as vascular endothelial growth factor, direct endothelial cells (ECs) to specific locations during blood vessel formation. Less is known about repulsive signal contribution to shaping vessels. Recently, "neuronal guidance cues" have been shown to influence EC behavior, particularly in directing sprouting angiogenesis by repelling ECs. However, their role during de novo blood vessel formation remains unexplored. OBJECTIVE: To identify signals that guide and pattern the first mammalian blood vessels. METHODS AND RESULTS: Using genetic mouse models, we show that blood vessels are sculpted through the generation of stereotyped avascular zones by EC-repulsive cues. We demonstrate that Semaphorin3E (Sema3E) is a key factor that shapes the paired dorsal aortae in mouse, as sema3E(-/-) embryos develop an abnormally branched aortic plexus with a markedly narrowed avascular midline. In vitro cultures and avian grafting experiments show strong repulsion of ECs by Sema3E-expressing cells. We further identify the mouse notochord as a rich source of multiple redundant neuronal guidance cues. Mouse embryos that lack notochords fail to form cohesive aortic vessels because of loss of the avascular midline, yet maintain lateral avascular zones. We demonstrate that lateral avascular zones are directly generated by the lateral plate mesoderm, a critical source of Sema3E. CONCLUSIONS: These findings demonstrate that Sema3E-generated avascular zones are critical regulators of mammalian cardiovascular patterning and are the first to identify a repulsive role for the lateral plate mesoderm. Integration of multiple, and in some cases redundant, repulsive cues from various tissues is critical to patterning the first embryonic blood vessels.


Assuntos
Vasos Sanguíneos/embriologia , Embrião de Mamíferos/irrigação sanguínea , Endotélio Vascular/embriologia , Glicoproteínas/fisiologia , Proteínas de Membrana/fisiologia , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Animais , Aorta/citologia , Aorta/embriologia , Vasos Sanguíneos/citologia , Células Cultivadas , Proteínas do Citoesqueleto , Endotélio Vascular/citologia , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/fisiologia , Glicoproteínas/deficiência , Glicoproteínas/genética , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/fisiologia , Técnicas In Vitro , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Mesoderma/citologia , Mesoderma/embriologia , Camundongos , Camundongos Knockout , Modelos Animais , Notocorda/citologia , Notocorda/embriologia , Semaforinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA