Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 43(7): 1199-1218, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37199159

RESUMEN

BACKGROUND: Endothelial cells (ECs) are sensitive to physical forces created by blood flow, especially to laminar shear stress. Among the cell responses to laminar flow, EC polarization against the flow direction emerges as a key event, particularly during the development and remodeling of the vascular network. EC adopt an elongated planar cell shape with an asymmetrical distribution of intracellular organelles along the axis of blood flow. This study aimed to investigate the involvement of planar cell polarity via the receptor ROR2 (receptor tyrosine kinase-like orphan receptor 2) in endothelial responses to laminar shear stress. METHODS: We generated a genetic mouse model with EC-specific deletion of Ror2, in combination with in vitro approaches involving loss- and gain-of-function experiments. RESULTS: During the first 2 weeks of life, the endothelium of the mouse aorta undergoes a rapid remodeling associated with a loss of EC polarization against the flow direction. Notably, we found a correlation between ROR2 expression and endothelial polarization levels. Our findings demonstrate that deletion of Ror2 in murine ECs impaired their polarization during the postnatal development of the aorta. In vitro experiments further validated the essential role of ROR2 in both EC collective polarization and directed migration under laminar flow conditions. Exposure to laminar shear stress triggered the relocalization of ROR2 to cell-cell junctions where it formed a complex with VE-Cadherin and ß-catenin, thereby regulating adherens junctions remodeling at the rear and front poles of ECs. Finally, we showed that adherens junctions remodeling and cell polarity induced by ROR2 were dependent on the activation of the small GTPase Cdc42. CONCLUSIONS: This study identified ROR2/planar cell polarity pathway as a new mechanism controlling and coordinating collective polarity patterns of EC during shear stress response.


Asunto(s)
Células Endoteliales , Receptores Huérfanos Similares al Receptor Tirosina Quinasa , Ratones , Animales , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Polaridad Celular/fisiología , Endotelio Vascular/metabolismo , Uniones Intercelulares , Estrés Mecánico
2.
Development ; 147(24)2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33144399

RESUMEN

Sense organs acquire their distinctive shapes concomitantly with the differentiation of sensory cells and neurons necessary for their function. Although our understanding of the mechanisms controlling morphogenesis and neurogenesis in these structures has grown, how these processes are coordinated remains largely unexplored. Neurogenesis in the zebrafish olfactory epithelium requires the bHLH proneural transcription factor Neurogenin 1 (Neurog1). To address whether Neurog1 also controls morphogenesis, we analysed the migratory behaviour of early olfactory neural progenitors in neurog1 mutant embryos. Our results indicate that the oriented movements of these progenitors are disrupted in this context. Morphogenesis is similarly affected by mutations in the chemokine receptor gene, cxcr4b, suggesting it is a potential Neurog1 target gene. We find that Neurog1 directly regulates cxcr4b through an E-box cluster located just upstream of the cxcr4b transcription start site. Our results suggest that proneural transcription factors, such as Neurog1, directly couple distinct aspects of nervous system development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Morfogénesis/genética , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Mucosa Olfatoria/crecimiento & desarrollo , Receptores CXCR4/genética , Proteínas de Pez Cebra/genética , Animales , Elementos E-Box/genética , Embrión no Mamífero , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica/genética , Mutación/genética , Neuronas/metabolismo , Sitio de Iniciación de la Transcripción , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
3.
Annu Rev Genet ; 49: 647-72, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26442849

RESUMEN

Although the left and right hemispheres of our brains develop with a high degree of symmetry at both the anatomical and functional levels, it has become clear that subtle structural differences exist between the two sides and that each is dominant in processing specific cognitive tasks. As the result of evolutionary conservation or convergence, lateralization of the brain is found in both vertebrates and invertebrates, suggesting that it provides significant fitness for animal life. This widespread feature of hemispheric specialization has allowed the emergence of model systems to study its development and, in some cases, to link anatomical asymmetries to brain function and behavior. Here, we present some of what is known about brain asymmetry in humans and model organisms as well as what is known about the impact of environmental and genetic factors on brain asymmetry development. We specifically highlight the progress made in understanding the development of epithalamic asymmetries in zebrafish and how this model provides an exciting opportunity to address brain asymmetry at different levels of complexity.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/fisiología , Lateralidad Funcional/fisiología , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Epitálamo/anatomía & histología , Epitálamo/fisiología , Lateralidad Funcional/genética , Hormonas/metabolismo , Humanos , Lenguaje , Postura , Pez Cebra/fisiología
4.
Arterioscler Thromb Vasc Biol ; 42(6): 745-763, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35510550

RESUMEN

BACKGROUND: While endothelial dysfunction is suggested to contribute to heart failure with preserved ejection fraction pathophysiology, understanding the importance of the endothelium alone, in the pathogenesis of diastolic abnormalities has not yet been fully elucidated. Here, we investigated the consequences of specific endothelial dysfunction on cardiac function, independently of any comorbidity or risk factor (diabetes or obesity) and their potential effect on cardiomyocyte. METHODS: The ubiquitine ligase Pdzrn3, expressed in endothelial cells (ECs), was shown to destabilize tight junction. A genetic mouse model in which Pdzrn3 is overexpressed in EC (iEC-Pdzrn3) in adults was developed. RESULTS: EC-specific Pdzrn3 expression increased cardiac leakage of IgG and fibrinogen blood-born molecules. The induced edema demonstrated features of diastolic dysfunction, with increased end-diastolic pressure, alteration of dP/dt min, increased natriuretic peptides, in addition to limited exercise capacity, without major signs of cardiac fibrosis and inflammation. Electron microscopic images showed edema with disrupted EC-cardiomyocyte interactions. RNA sequencing analysis of gene expression in cardiac EC demonstrated a decrease in genes coding for endothelial extracellular matrix proteins, which could be related to the fragile blood vessel phenotype. Irregularly shaped capillaries with hemorrhages were found in heart sections of iEC-Pdzrn3 mice. We also found that a high-fat diet was not sufficient to provoke diastolic dysfunction; high-fat diet aggravated cardiac inflammation, associated with an altered cardiac metabolic signature in EC-Pdzrn3 mice, reminiscent of heart failure with preserved ejection fraction features. CONCLUSIONS: An increase of endothelial permeability is responsible for mediating diastolic dysfunction pathophysiology and for aggravating detrimental effects of a high-fat diet on cardiac inflammation and metabolism.


Asunto(s)
Cardiomiopatías , Insuficiencia Cardíaca , Animales , Permeabilidad Capilar , Células Endoteliales/metabolismo , Fibrosis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Inflamación/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Volumen Sistólico/fisiología , Ubiquitina-Proteína Ligasas
5.
Arterioscler Thromb Vasc Biol ; 42(5): 597-609, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35387477

RESUMEN

BACKGROUND: Genome-wide association studies have revealed robust associations of common genetic polymorphisms in an intron of the PHACTR-1 (phosphatase and actin regulator 1) gene (chr6p24), with cervical artery dissection, spontaneous coronary artery dissection, and fibromuscular dysplasia. The aim was to assess its role in the pathogenesis of cervical artery dissection or fibromuscular dysplasia. METHODS: Using various tissue-specific Cre-driver mouse lines, Phactr1 was deleted either in endothelial cells using 2 tissue-specific Cre-driver (PDGFB [platelet-derived growth factor B]-CreERT2 mice and Tie2 [tyrosine kinase with immunoglobulin and EGF homology domains]-Cre) and smooth muscle cells (smooth muscle actin-CreERT2) with a third tissue-specific Cre-driver. RESULTS: To test the efficacy of the Phactr1 deletion after cre-induction, we confirmed first, a decrease in Phactr1 transcription and Phactr1 expression in endothelial cell and smooth muscle cell isolated from Phactr1iPDGFB and Phactr1iSMA mice. Irrespective to the tissue or the duration of the deletion, mice did not spontaneously display pathological phenotype or vascular impairment: mouse survival, growth, blood pressure, large vessel morphology, or actin organization were not different in knockout mice than their comparatives littermates. Challenging vascular function and repair either by angiotensin II-induced hypertension or limb ischemia did not lead to vascular morphology or function impairment in Phactr1-deleted mice. Similarly, there were no more consequences of Phactr1 deletion during embryogenesis in endothelial cells. CONCLUSIONS: Loss of PHACTR-1 function in the cells involved in vascular physiology does not appear to induce a pathological vascular phenotype. The in vivo effect of the intronic variation described in genome-wide association studies is unlikely to involve downregulation in PHACTR-1 expression.


Asunto(s)
Actinas , Arteriopatías Oclusivas/metabolismo , Displasia Fibromuscular , Proteínas de Microfilamentos/metabolismo , Actinas/metabolismo , Animales , Células Endoteliales/metabolismo , Displasia Fibromuscular/genética , Estudio de Asociación del Genoma Completo , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Miocitos del Músculo Liso/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo
6.
FASEB J ; 34(1): 1288-1303, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914666

RESUMEN

Retinopathies remain major causes of visual impairment in diabetic patients and premature infants. Introduction of anti-angiogenic drugs targeting vascular endothelial growth factor (VEGF) has transformed therapy for these proliferative retinopathies. However, limitations associated with anti-VEGF medications require to unravel new pathways of vessel growth to identify potential drug targets. Here, we investigated the role of Wnt/Frizzled-7 (Fzd7) pathway in a mouse model of oxygen-induced retinopathy (OIR). Using transgenic mice, which enabled endothelium-specific and time-specific Fzd7 deletion, we demonstrated that Fzd7 controls both vaso-obliteration and neovascular phases (NV). Deletion of Fzd7 at P12, after the ischemic phase of OIR, prevented formation of aberrant neovessels into the vitreous by suppressing proliferation of endothelial cells (EC) in tufts. Next we validated in vitro two Frd7 blocking strategies: a monoclonal antibody (mAbFzd7) against Fzd7 and a soluble Fzd7 receptor (CRD). In vivo a single intravitreal microinjection of mAbFzd7 or CRD significantly attenuated retinal neovascularization (NV) in mice with OIR. Molecular analysis revealed that Fzd7 may act through the activation of Wnt/ß-catenin and Jagged1 expression to control EC proliferation in extra-retinal neovessels. We identified Fzd7/ß-catenin signaling as new regulator of pathological retinal NV. Fzd7 appears to be a potent pharmacological target to prevent or treat aberrant angiogenesis of ischemic retinopathies.


Asunto(s)
Retinopatía Diabética/metabolismo , Isquemia/metabolismo , Proteínas Represoras/metabolismo , Neovascularización Retiniana/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Eliminación de Gen , Isquemia/genética , Isquemia/patología , Proteína Jagged-1/biosíntesis , Proteína Jagged-1/genética , Ratones , Ratones Mutantes , Proteínas Represoras/genética , Neovascularización Retiniana/genética , Neovascularización Retiniana/patología , beta Catenina/genética
7.
Development ; 141(7): 1572-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24598158

RESUMEN

Left-right (L/R) asymmetries in the brain are thought to underlie lateralised cognitive functions. Understanding how neuroanatomical asymmetries are established has been achieved through the study of the zebrafish epithalamus. Morphological symmetry in the epithalamus is broken by leftward migration of the parapineal, which is required for the subsequent elaboration of left habenular identity; the habenular nuclei flank the midline and show L/R asymmetries in marker expression and connectivity. The Nodal target pitx2c is expressed in the left epithalamus, but nothing is known about its role during the establishment of asymmetry in the brain. We show that abrogating Pitx2c function leads to the right habenula adopting aspects of left character, and to an increase in parapineal cell numbers. Parapineal ablation in Pitx2c loss of function results in right habenular isomerism, indicating that the parapineal is required for the left character detected in the right habenula in this context. Partial parapineal ablation in the absence of Pitx2c, however, reduces the number of parapineal cells to wild-type levels and restores habenular asymmetry. We provide evidence suggesting that antagonism between Nodal and Pitx2c activities sets an upper limit on parapineal cell numbers. We conclude that restricting parapineal cell number is crucial for the correct elaboration of epithalamic asymmetry.


Asunto(s)
Tipificación del Cuerpo/genética , Habénula/embriología , Glándula Pineal/embriología , Factores de Transcripción/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Recuento de Células , Embrión no Mamífero , Epitálamo/citología , Epitálamo/embriología , Habénula/citología , Proteína Nodal/fisiología , Tamaño de los Órganos/genética , Glándula Pineal/citología , Transducción de Señal/fisiología , Factores de Transcripción/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
8.
Arterioscler Thromb Vasc Biol ; 36(12): 2369-2380, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27758766

RESUMEN

OBJECTIVE: Vessel formation requires precise orchestration of a series of morphometric and molecular events controlled by a multitude of angiogenic factors and morphogens. Wnt/frizzled signaling is required for proper vascular formation. In this study, we investigated the role of the Fzd7 (frizzled-7) receptor in retinal vascular development and its relationship with the Wnt/ß-catenin canonical pathway and Notch signaling. APPROACH AND RESULTS: Using transgenic mice, we demonstrated that Fzd7 is required for postnatal vascular formation. Endothelial cell (EC) deletion of fzd7 (fzd7ECKO) delayed retinal plexus formation because of an impairment in tip cell phenotype and a decrease in stalk cell proliferation. Dvl (dishevelled) proteins are a main component of Wnt signaling and play a functionally redundant role. We found that Dvl3 depletion in dvl1-/- mice mimicked the fzd7ECKO vascular phenotype and demonstrated that Fzd7 acted via ß-catenin activation by showing that LiCl treatment rescued impairment in tip and stalk cell phenotypes induced in fzd7 mutants. Deletion of fzd7 or Dvl1/3 induced a strong decrease in Wnt canonical genes and Notch partners' expression. Genetic and pharmacological rescue strategies demonstrated that Fzd7 acted via ß-catenin activation, upstream of Notch signaling to control Dll4 and Jagged1 EC expression. CONCLUSIONS: Fzd7 expressed by EC drives postnatal angiogenesis via activation of Dvl/ß-catenin signaling and can control the integrative interaction of Wnt and Notch signaling during postnatal angiogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Fisiológica , Receptores Acoplados a Proteínas G/metabolismo , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio , Proliferación Celular , Células Cultivadas , Proteínas Dishevelled/deficiencia , Proteínas Dishevelled/genética , Células Endoteliales/efectos de los fármacos , Receptores Frizzled , Genotipo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Jagged-1/metabolismo , Cloruro de Litio/farmacología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Fenotipo , Interferencia de ARN , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Notch/metabolismo , Neovascularización Retiniana/genética , Neovascularización Retiniana/fisiopatología , Vasos Retinianos/efectos de los fármacos , Transfección , Vía de Señalización Wnt/efectos de los fármacos
9.
Circ Res ; 110(1): 47-58, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22076635

RESUMEN

RATIONALE: A growing body of evidence supports the hypothesis that the Wnt/planar cell polarity (PCP) pathway regulates endothelial cell proliferation and angiogenesis, but the components that mediate this regulation remain elusive. OBJECTIVE: We investigated the involvement of one of the receptors, Frizzled4 (Fzd4), in this process because its role has been implicated in retinal vascular development. METHODS AND RESULTS: We found that loss of fzd4 function in mice results in a striking reduction and impairment of the distal small artery network in the heart and kidney. We report that loss of fzd4 decreases vascular cell proliferation and migration and decreases the ability of the endothelial cells to form tubes. We show that fzd4 deletion induces defects in the expression level of stable acetylated tubulin and in Golgi organization during migration. Deletion of fzd4 favors Wnt noncanonical AP1-dependent signaling, indicating that Fzd4 plays a pivotal role favoring PCP signaling. Our data further demonstrate that Fzd4 is predominantly localized on the top of the plasma membrane, where it preferentially induces Dvl3 relocalization to promote its activation and α-tubulin recruitment during migration. In a pathological mouse angiogenic model, deletion of fzd4 impairs the angiogenic response and leads to the formation of a disorganized arterial network. CONCLUSIONS: These results suggest that Fzd4 is a major receptor involved in arterial formation and organization through a Wnt/PCP pathway.


Asunto(s)
Arterias/citología , Polaridad Celular/fisiología , Proliferación Celular , Receptores Frizzled/fisiología , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Proteínas Wnt/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Arterias/fisiología , Arteriolas/citología , Arteriolas/fisiología , Movimiento Celular/fisiología , Proteínas Dishevelled , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Receptores Frizzled/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas de Sustitución del Gen , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Microtúbulos/fisiología , Modelos Animales , Fosfoproteínas/fisiología
10.
Arterioscler Thromb Vasc Biol ; 31(11): e80-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21836067

RESUMEN

OBJECTIVE: The inflammatory response after myocardial infarction plays a crucial role in the healing process. Lately, there is accumulating evidence that the Wnt/Frizzled pathway may play a distinct role in inflammation. We have shown that secreted frizzled-related protein-1 (sFRP-1) overexpression reduced postinfarction scar size, and we noticed a decrease in neutrophil infiltration in the ischemic tissue. We aimed to further elucidate the role of sFRP-1 in the postischemic inflammatory process. METHODS AND RESULTS: We found that in vitro, sFRP-1 was able to block leukocyte activation and cytokine production. We transplanted bone marrow cells (BMCs) from transgenic mice overexpressing sFRP-1 into wild-type recipient mice and compared myocardial healing with that of mice transplanted with wild-type BMCs. These results were compared with those obtained in transgenic mice overexpressing sFRP-1 specifically in endothelial cells or in cardiomyocytes to better understand the spatiotemporal mechanism of the sFRP-1 effect. Our findings indicate that when overexpressed in the BMCs, but not in endothelial cells or cardiomyocytes, sFRP-1 was able to reduce neutrophil infiltration after ischemia, by switching the balance of pro- and antiinflammatory cytokine expression, leading to a reduction in scar formation and better cardiac hemodynamic parameters. CONCLUSION: sFRP-1 impaired the loop of cytokine amplification and decreased neutrophil activation and recruitment into the scar, without altering the neutrophil properties. These data support the notion that sFRP-1 may be a novel antiinflammatory factor protecting the heart from damage after myocardial infarction.


Asunto(s)
Cicatriz/etiología , Cicatriz/metabolismo , Inflamación/metabolismo , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Proteínas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Línea Celular , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Cicatriz/patología , Citocinas/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Transgénicos , Modelos Animales , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Proteínas/farmacología
11.
Artículo en Inglés | MEDLINE | ID: mdl-35074794

RESUMEN

The Wnt/frizzled signaling pathway is one of the major regulators of endothelial biology, controlling key cellular activities. Many secreted Wnt ligands have been identified and can initiate diverse signaling via binding to a complex set of Frizzled (Fzd) transmembrane receptors and coreceptors. Roughly, Wnt signaling is subdivided into two pathways: the canonical Wnt/ß-catenin signaling pathway whose main downstream effector is the transcriptional coactivator ß-catenin, and the noncanonical Wnt signaling pathway, which is subdivided into the Wnt/Ca2+ pathway and the planar cell polarity pathway. Here, we will focus on its cross talk with other angiogenic pathways and on its role in blood-retinal- and blood-brain-barrier formation and its maintenance in a differentiated state. We will unravel how retinal vascular pathologies and neurovascular degenerative diseases result from disruption of the Wnt pathway related to vascular instability, and highlight current research into therapeutic options.


Asunto(s)
Barrera Hematoencefálica , Vía de Señalización Wnt , Endotelio , Receptores Frizzled , Humanos , Ligandos
12.
J Cereb Blood Flow Metab ; 42(4): 613-629, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34644209

RESUMEN

Blood brain barrier (BBB) disruption is a critical component of the pathophysiology of cognitive impairment of vascular etiology (VCI) and associated with Alzheimer's disease (AD). The Wnt pathway plays a crucial role in BBB maintenance, but there is limited data on its role in cognitive pathologies. The E3 ubiquitin ligase PDZRN3 is a regulator of the Wnt pathway. In a murine model of VCI, overexpressing Pdzrn3 in endothelial cell (EC) exacerbated BBB hyperpermeability and accelerated cognitive decline. We extended these observations, in both VCI and AD models, showing that EC-specific depletion of Pdzrn3, reinforced the BBB, with a decrease in vascular permeability and a subsequent spare in cognitive decline. We found that in cerebral vessels, Pdzrn3 depletion protects against AD-induced Wnt target gene alterations and enhances endothelial tight junctional proteins. Our results provide evidence that Wnt signaling could be a molecular link regulating BBB integrity and cognitive decline under VCI and AD pathologies.


Asunto(s)
Enfermedad de Alzheimer , Barrera Hematoencefálica , Ubiquitina-Proteína Ligasas , Enfermedad de Alzheimer/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Homeostasis , Ratones , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
13.
Sci Rep ; 12(1): 8, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34996942

RESUMEN

Heart failure is the final common stage of most cardiopathies. Cardiomyocytes (CM) connect with others via their extremities by intercalated disk protein complexes. This planar and directional organization of myocytes is crucial for mechanical coupling and anisotropic conduction of the electric signal in the heart. One of the hallmarks of heart failure is alterations in the contact sites between CM. Yet no factor on its own is known to coordinate CM polarized organization. We have previously shown that PDZRN3, an ubiquitine ligase E3 expressed in various tissues including the heart, mediates a branch of the Planar cell polarity (PCP) signaling involved in tissue patterning, instructing cell polarity and cell polar organization within a tissue. PDZRN3 is expressed in the embryonic mouse heart then its expression dropped significantly postnatally corresponding with heart maturation and CM polarized elongation. A moderate CM overexpression of Pdzrn3 (Pdzrn3 OE) during the first week of life, induced a severe eccentric hypertrophic phenotype with heart failure. In models of pressure-overload stress heart failure, CM-specific Pdzrn3 knockout showed complete protection against degradation of heart function. We reported that Pdzrn3 signaling induced PKC ζ expression, c-Jun nuclear translocation and a reduced nuclear ß catenin level, consistent markers of the planar non-canonical Wnt signaling in CM. We then show that subcellular localization (intercalated disk) of junction proteins as Cx43, ZO1 and Desmoglein 2 was altered in Pdzrn3 OE mice, which provides a molecular explanation for impaired CM polarization in these mice. Our results reveal a novel signaling pathway that controls a genetic program essential for heart maturation and maintenance of overall geometry, as well as the contractile function of CM, and implicates PDZRN3 as a potential therapeutic target for the prevention of human heart failure.


Asunto(s)
Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/prevención & control , Corazón/crecimiento & desarrollo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Ratones , Ratones Noqueados , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , beta Catenina/genética , beta Catenina/metabolismo
14.
Mol Ther ; 18(8): 1545-52, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20551912

RESUMEN

Mesenchymal stem cells (MSC) are multipotent postnatal stem cells, involved in the treatment of ischemic vascular diseases. We investigate the ability of MSC, exposed to short-term hypoxic conditions, to participate in vascular and tissue regeneration in an in vivo model of hindlimb ischemia. Transplantation of hypoxic preconditioned murine MSC (HypMSC) enhanced skeletal muscle regeneration at day 7, improved blood flow and vascular formation compared to injected nonpreconditioned MSC (NormMSC). These observed effects were correlated with an increase in HypMSC engraftment and a putative role in necrotic skeletal muscle fiber clearance. Moreover, HypMSC transplantation resulted in a large increase in Wnt4 (wingless-related MMTV integration site 4) expression and we demonstrate its functional significance on MSC proliferation and migration, endothelial cell (EC) migration, as well as myoblast differentiation. Furthermore, suppression of Wnt4 expression in HypMSC, abrogated the hypoxia-induced vascular regenerative properties of these cells in the mouse hindlimb ischemia model. Our data suggest that hypoxic preconditioning plays a critical role in the functional capabilities of MSC, shifting MSC location in situ to enhance ischemic tissue recovery, facilitating vascular cell mobilization, and skeletal muscle fiber regeneration via a paracrine Wnt-dependent mechanism.


Asunto(s)
Miembro Posterior/metabolismo , Miembro Posterior/patología , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Proteínas Wnt/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/metabolismo , Isquemia/metabolismo , Isquemia/patología , Ratones , Ratones Noqueados , Mioblastos/citología , Mioblastos/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Wnt/genética , Proteína Wnt4
15.
FASEB J ; 23(12): 4181-92, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19723704

RESUMEN

The translocator protein (18 kDa) (TSPO), also known as peripheral-type benzodiazepine receptor, is directly or indirectly associated with many biological processes. Although extensively characterized, the specific function of TSPO during development remains unclear. It has been reported that TSPO is involved in a variety of mechanisms, including cell proliferation, apoptosis, regulation of mitochondrial functions, cholesterol transport and steroidogenesis, and porphyrin transport and heme synthesis. Although the literature has reported a murine knockout model, the experiment did not generate information because of early lethality. We then used the zebrafish model to address the function of tspo during development. Information about spatiotemporal expression showed that tspo has a maternal and a zygotic contribution which, during somatogenesis, seems to be erythroid restricted to the intermediate cell mass. Genetic and pharmacological approaches used to invalidate Tspo function resulted in embryos with specific erythropoietic cell depletion. Although unexpected, this lack of blood cells is independent of the Tspo cholesterol binding site and reveals a new in vivo key role for Tspo during erythropoiesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Eritropoyesis/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Receptores de GABA/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Evolución Biológica , Proteínas Portadoras/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Isoquinolinas/farmacología , Datos de Secuencia Molecular , Receptores de GABA/genética , Proteínas de Pez Cebra/genética
16.
Arterioscler Thromb Vasc Biol ; 29(12): 2090-2, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19745199

RESUMEN

OBJECTIVE: Studying the mechanisms of neovascularization and evaluating the effects of proangiogenic strategies require accurate analysis of the neovascular network. We sought to evaluate the contribution of the microcomputed tomography (mCT) providing high-resolution 3-dimensional (3D) structural data, to a better comprehension of the well-studied mouse hindlimb postischemic neovascularization. METHODS AND RESULTS: We showed a predominant arteriogenesis process in the thigh and a predominant angiogenesis-related process in the tibiofibular region, in response to ischemia during the first 15 days. After 15 days, mCT quantitative analysis reveals a remodeling of arterial neovessels and a regression depending on the restoration of the blood flow. We provided also new mCT data on the rapid and potent angiogenic effects of mesenchymal stem cell therapy on vessel formation and organization. We discussed the contribution of this technique compared with or in addition to data generated by the more conventional approaches. CONCLUSIONS: This study demonstrated that optimized mCT is a robust method for providing new insights into the 3D understanding of postischemic vessel formation.


Asunto(s)
Miembro Posterior/irrigación sanguínea , Isquemia/patología , Neovascularización Fisiológica , Tomografía Computarizada por Rayos X/métodos , Animales , Bario , Medios de Contraste , Modelos Animales de Enfermedad , Imagenología Tridimensional , Isquemia/cirugía , Trasplante de Células Madre Mesenquimatosas , Ratones , Neopreno , Enfermedades Vasculares Periféricas/patología , Enfermedades Vasculares Periféricas/cirugía
17.
Front Physiol ; 11: 861, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32848833

RESUMEN

Blood flow produces mechanical frictional forces, parallel to the blood flow exerted on the endothelial wall of the vessel, the so-called wall shear stress (WSS). WSS sensing is associated with several vascular pathologies, but it is first a physiological phenomenon. Endothelial cell sensitivity to WSS is involved in several developmental and physiological vascular processes such as angiogenesis and vascular morphogenesis, vascular remodeling, and vascular tone. Local conditions of blood flow determine the characteristics of WSS, i.e., intensity, direction, pulsatility, sensed by the endothelial cells that, through their effect of the vascular network, impact WSS. All these processes generate a local-global retroactive loop that determines the ability of the vascular system to ensure the perfusion of the tissues. In order to account for the physiological role of WSS, the so-called shear stress set point theory has been proposed, according to which WSS sensing acts locally on vessel remodeling so that WSS is maintained close to a set point value, with local and distant effects of vascular blood flow. The aim of this article is (1) to review the existing literature on WSS sensing involvement on the behavior of endothelial cells and its short-term (vasoreactivity) and long-term (vascular morphogenesis and remodeling) effects on vascular functioning in physiological condition; (2) to present the various hypotheses about WSS sensors and analyze the conceptual background of these representations, in particular the concept of tensional prestress or biotensegrity; and (3) to analyze the relevance, explanatory value, and limitations of the WSS set point theory, that should be viewed as dynamical, and not algorithmic, processes, acting in a self-organized way. We conclude that this dynamic set point theory and the biotensegrity concept provide a relevant explanatory framework to analyze the physiological mechanisms of WSS sensing and their possible shift toward pathological situations.

18.
Stem Cells ; 26(11): 2991-3001, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18757297

RESUMEN

Mesenchymal stem cell (MSC) transplantation offers a great angiogenic opportunity in vascular regenerative medicine. The canonical Wnt/beta-catenin signaling pathway has been demonstrated to play an essential role in stem cell fate. Recently, genetic studies have implicated the Wnt/Frizzled (Fz) molecular pathway, namely Wnt7B and Fz4, in blood growth regulation. Here, we investigated whether MSC could be required in shaping a functional vasculature and whether secreted Frizzled-related protein-1 (sFRP1), a modulator of the Wnt/Fz pathway, could modify MSC capacities, endowing MSC to increase vessel maturation. In the engraftment model, we show that murine bone marrow-derived MSC induced a beneficial vascular effect through a direct cellular contribution to vascular cells. MSC quickly organized into primitive immature vessel tubes connected to host circulation; this organization preceded host endothelial cell (EC) and smooth muscle cell (SMC) recruitment to later form mature neovessel. MSC sustained neovessel organization and maturation. We report here that sFRP1 forced expression enhanced MSC surrounding neovessel, which was correlated with an increase in vessel maturation and functionality. In vitro, sFRP1 strongly increased platelet-derived growth factor-BB (PDGF-BB) expression in MSC and enhanced beta-catenin-dependent cell-cell contacts between MSC themselves and EC or SMC. In vivo, sFRP1 increased their functional integration around neovessels and vessel maturation through a glycogen synthase kinase 3 beta (GSK3beta)-dependent pathway. sFRP1-overexpressing MSC compared with control MSC were well elongated and in a closer contact with the vascular wall, conditions required to achieve an organized mature vessel wall. We propose that genetically modifying MSC to overexpress sFRP1 may be potentially effective in promoting therapeutic angiogenesis/arteriogenesis processes. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica/fisiología , Proteínas/metabolismo , Animales , Becaplermina , Bovinos , Adhesión Celular/fisiología , Células Cultivadas , Colágeno , Combinación de Medicamentos , Células Endoteliales/citología , Células Endoteliales/fisiología , Péptidos y Proteínas de Señalización Intracelular , Laminina , Lentivirus/genética , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas/genética , Proteoglicanos , Proteínas Proto-Oncogénicas c-sis , Trasplante Heterólogo , beta Catenina/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 28(12): 2131-6, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18772499

RESUMEN

OBJECTIVE: Estradiol (E(2)) is known to accelerate reendothelialization and thus prevent intimal thickening and in-stent restenosis after angioplasty. Transplantation experiments with ERalpha(-/-) mice have previously shown that E(2) acts through local and bone marrow cell compartments to enhance endothelial healing. However, the downstream mechanisms induced by E(2) to mediate endothelial repair are still poorly understood. METHODS AND RESULTS: We show here that after endovascular carotid artery injury, E(2)-enhanced endothelial repair is lost in osteopontin-deficient mice (OPN(-/-)). Transplantation of OPN(-/-) bone marrow into wild-type lethally irradiated mice, and vice versa, suggested that osteopontin plays a crucial role in both the local and the bone marrow actions of E(2). In the vascular compartment, using transgenic mice expressing doxycyclin regulatable-osteopontin, we show that endothelial cell specific osteopontin overexpression mimics E(2)-enhanced endothelial cell migration and proliferation in the regenerating endothelium. In the bone marrow cell compartment, we demonstrate that E(2) enhances bone marrow-derived mononuclear cell adhesion to regenerating endothelium in vivo, and that this effect is dependent on osteopontin. CONCLUSIONS: We demonstrate here that E(2) acceleration of the endothelial repair requires osteopontin, both for bone marrow-derived cell recruitment and for endothelial cell migration and proliferation.


Asunto(s)
Traumatismos de las Arterias Carótidas/fisiopatología , Células Endoteliales/citología , Células Endoteliales/fisiología , Estradiol/farmacología , Osteopontina/fisiología , Animales , Trasplante de Médula Ósea , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Ratones , Ratones Noqueados , Ratones Transgénicos , Osteopontina/deficiencia , Osteopontina/genética , Regeneración/efectos de los fármacos , Regeneración/fisiología
20.
Eur J Cardiothorac Surg ; 34(2): 248-54, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18457957

RESUMEN

OBJECTIVES: To assess, using an in vivo engraftment strategy combining bone marrow cell (BMC) transplantation and tissue cardiomyoplasty, the functional outcome of distinct vascular progenitor cell therapy (endothelial progenitor (EPC) and mesenchymal stem (MSC) cells) at distance of myocardium infarction (MI). The study was also designed to test whether scaffold mixing progenitors with unfractionated BMC could improve progenitor recruitment in the damaged myocardium. METHODS: To track engrafted progenitor cells in vivo, cultured murine MSC and EPC were transduced with eGFP lentiviruses. Thirty days after cryogenical induction of MI, C57BL/6J mice were randomized to receive muscle patch placement coated or not (control group), labeled EPC or MSC mixed to the ration of 1:10, or not with unfractionated BMC. Two weeks after transplantation, cardiac function was recorded and heart sections were examined to detect GFP-labeled progenitor cells and analyze cell differentiation. RESULTS: This study showed that either type of mono cell therapy improved angiogenesis and cell survival in the scar but only MSC exhibited the capacity to invade the scar. We found no evidence of myocardial or vascular regeneration from progenitor cells. Engraftment of the progenitors/unfractionated BMC mix increased repopulation and thickness of the scar. CONCLUSION: Combined therapy with unfractionated BMC and expanded MSC appeared thus promising for scar repopulation.


Asunto(s)
Infarto del Miocardio/terapia , Pericardio/patología , Trasplante de Células Madre/métodos , Músculos Abdominales/trasplante , Animales , Diferenciación Celular , Movimiento Celular , Supervivencia Celular , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Corazón/fisiología , Inmunofenotipificación , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Músculo Liso/fisiología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Neovascularización Fisiológica , Regeneración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA