Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
NMR Biomed ; : e5148, 2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38556903

RESUMO

Intravoxel incoherent motion (IVIM) MRI has emerged as a valuable technique for the assessment of tissue characteristics and perfusion. However, there is limited knowledge about the relationship between IVIM-derived measures and changes at the level of the vascular network. In this study, we investigated the potential use of IVIM MRI as a noninvasive tool for measuring changes in cerebral vascular density. Variations in quantitative immunohistochemical measurements of the vascular density across different regions in the rat brain (cortex, corpus callosum, hippocampus, thalamus, and hypothalamus) were related to the pseudo-diffusion coefficient D* and the flowing blood fraction f in healthy Wistar rats. We assessed whether region-wise differences in the vascular density are reflected by variations in the IVIM measurements and found a significant positive relationship with the pseudo-diffusion coefficient (p < 0.05, ß = 0.24). The effect of cerebrovascular alterations, such as blood-brain barrier (BBB) disruption on the perfusion-related IVIM parameters, is not well understood. Therefore, we investigated the effect of BBB disruption on the IVIM measures in a rat model of metabolic and vascular comorbidities (ZSF1 obese rat) and assessed whether this affects the relationship between the cerebral vascular density and the noninvasive IVIM measurements. We observed increased vascular permeability without detecting any differences in diffusivity, suggesting that BBB leakage is present before changes in the tissue integrity. We observed no significant difference in the relationship between cerebral vascular density and the IVIM measurements in our model of comorbidities compared with healthy normotensive rats.

2.
Development ; 147(15)2020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32769200

RESUMO

Fluid flow is a powerful morphogenic force during embryonic development. The physical forces created by flowing fluids can either create morphogen gradients or be translated by mechanosensitive cells into biological changes in gene expression. In this Primer, we describe how fluid flow is created in different systems and highlight the important mechanosensitive signalling pathways involved for sensing and transducing flow during embryogenesis. Specifically, we describe how fluid flow helps establish left-right asymmetry in the early embryo and discuss the role of flow of blood, lymph and cerebrospinal fluid in sculpting the embryonic cardiovascular and nervous system.


Assuntos
Sistema Cardiovascular/embriologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário/fisiologia , Sistema Nervoso/embriologia , Neurogênese , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Transdução de Sinais
4.
Curr Heart Fail Rep ; 20(5): 451-460, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37526812

RESUMO

PURPOSE OF REVIEW: Though patient studies have been important for understanding the disease, research done in animals and cell culture complement our knowledge from patient data and provide insight into the mechanism of the disease. Understanding how COVID causes damage to the heart is essential to understanding possible long-term consequences. RECENT FINDINGS: COVID-19 is primarily a disease that attacks the lungs; however, it is known to have important consequences in many other tissues including the heart. Though myocarditis does occur in some patients, for most cases of cardiac damage, the injury arises from scarring either due to myocardial infarction or micro-infarction. The main focus is on how COVID affects blood flow through the coronaries. We review how endothelial activation leads to a hypercoagulative state in COVID-19. We also emphasize the effects that the cytokine storm can directly have on the regulation of coronary blood flow. Since the main two cell types that can be infected in the heart are pericytes and cardiomyocytes, we further describe the known effects on pericyte function and how that can further lead to microinfarcts within the heart. Though many of these effects are systemic, this review focuses on the consequences on cardiac tissue of this dysregulation and the role that it has in the formation of myocardial scarring.

5.
Circ Res ; 127(6): 707-723, 2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32527198

RESUMO

RATIONALE: How endothelial cells (ECs) migrate and form an immature vascular plexus has been extensively studied. Yet, mechanisms underlying vascular remodeling remain poorly established. A better understanding of these processes may lead to the design of novel therapeutic strategies complementary to current angiogenesis inhibitors. OBJECTIVE: Starting from our previous observations that PP2A (protein phosphatase 2) regulates the HIF (hypoxia-inducible factor)/PHD-2 (prolyl hydroxylase 2)-constituted oxygen machinery, we hypothesized that this axis could play an important role during blood vessel formation, tissue perfusion, and oxygen restoration. METHODS AND RESULTS: We show that the PP2A regulatory subunit B55α is at the crossroad between vessel pruning and vessel maturation. Blood vessels with high B55α counter cell stress conditions and thrive for stabilization and maturation. When B55α is inhibited, ECs cannot cope with cell stress and undergo apoptosis, leading to massive pruning of nascent blood vessels. Mechanistically, we found that the B55α/PP2A complex restrains PHD-2 activity, promoting EC survival in a HIF-dependent manner, and furthermore dephosphorylates p38, altogether protecting ECs against cell stress occurring, for example, during the onset of blood flow. In tumors, EC-specific B55α deficiency induces pruning of immature-like tumor blood vessels resulting in delayed tumor growth and metastasis, without affecting nonpathological vessels. Consistently, systemic administration of a pan-PP2A inhibitor disrupts vascular network formation and tumor progression in vivo without additional effects on B55α-deficient vessels. CONCLUSIONS: Our data underline a unique role of the B55α/PP2A phosphatase complex in vessel remodeling and suggest the use of PP2A-inhibitors as potent antiangiogenic drugs targeting specifically nascent blood vessels with a mode-of-action complementary to VEGF-R (vascular endothelial growth factor receptor)-targeted therapies. Graphical Abstract: A graphical abstract is available for this article.


Assuntos
Apoptose , Neoplasias da Mama/enzimologia , Carcinoma Pulmonar de Lewis/enzimologia , Células Endoteliais/enzimologia , Neovascularização Patológica , Proteína Fosfatase 2/metabolismo , Remodelação Vascular , Inibidores da Angiogênese/farmacologia , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patologia , Linhagem Celular Tumoral , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Inibidores Enzimáticos/farmacologia , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/genética , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35163400

RESUMO

Endothelial cells throughout the body are heterogeneous, and this is tightly linked to the specific functions of organs and tissues. Heterogeneity is already determined from development onwards and ranges from arterial/venous specification to microvascular fate determination in organ-specific differentiation. Acknowledging the different phenotypes of endothelial cells and the implications of this diversity is key for the development of more specialized tissue engineering and vascular repair approaches. However, although novel technologies in transcriptomics and proteomics are facilitating the unraveling of vascular bed-specific endothelial cell signatures, still much research is based on the use of insufficiently specialized endothelial cells. Endothelial cells are not only heterogeneous, but their specialized phenotypes are also dynamic and adapt to changes in their microenvironment. During the last decades, strong collaborations between molecular biology, mechanobiology, and computational disciplines have led to a better understanding of how endothelial cells are modulated by their mechanical and biochemical contexts. Yet, because of the use of insufficiently specialized endothelial cells, there is still a huge lack of knowledge in how tissue-specific biomechanical factors determine organ-specific phenotypes. With this review, we want to put the focus on how organ-specific endothelial cell signatures are determined from development onwards and conditioned by their microenvironments during adulthood. We discuss the latest research performed on endothelial cells, pointing out the important implications of mimicking tissue-specific biomechanical cues in culture.


Assuntos
Diferenciação Celular , Microambiente Celular , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Animais , Humanos , Especificidade de Órgãos , Engenharia Tecidual
7.
J Cell Mol Med ; 25(2): 729-741, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33295687

RESUMO

The metabolic syndrome (MetS) is an escalating problem worldwide, causing left ventricular stiffening, an early characteristic of diastolic dysfunction for which no treatment exists. As diastolic dysfunction and stiffening in MetS patients are associated with increased circulating dipeptidyl peptidase-4 (DPP-4) levels, we investigated whether the clinically approved DPP-4 inhibitor linagliptin reduces left ventricular stiffness in MetS-induced cardiac disease. Sixteen-week-old obese ZSF1 rats, displaying the MetS and left ventricular stiffness, received linagliptin-supplemented or placebo diet for four weeks. Linagliptin significantly reduced obesity, hyperlipidaemia, and hyperglycaemia and improved left ventricular relaxation. This improved relaxation was related to decreased cardiac fibrosis and cardiomyocyte passive stiffness (Fpassive ). The reduced Fpassive was the result of titin isoform switching from the stiff N2B to the more flexible N2BA and increased phosphorylation of total titin and specifically its N2Bus region (S4080 and S3391). Importantly, DPP-4 directly cleaved titin in vitro, resulting in an increased Fpassive , which was prevented by simultaneous administration of linagliptin. In conclusion, linagliptin improves left ventricular stiffness in obese ZSF1 rats by preventing direct DPP4-mediated titin cleavage, as well as by modulating both titin isoform levels and phosphorylation. Reducing left ventricular stiffness by administering linagliptin might prevent MetS-induced early diastolic dysfunction in human.


Assuntos
Linagliptina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Animais , Conectina/farmacologia , Cardiopatias/metabolismo , Masculino , Camundongos Obesos , Miocárdio/metabolismo , Obesidade/metabolismo , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional , Ratos
8.
PLoS Comput Biol ; 16(8): e1007874, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32822340

RESUMO

Shear stress induces directed endothelial cell (EC) migration in blood vessels leading to vessel diameter increase and induction of vascular maturation. Other factors, such as EC elongation and interaction between ECs and non-vascular areas are also important. Computational models have previously been used to study collective cell migration. These models can be used to predict EC migration and its effect on vascular remodelling during embryogenesis. We combined live time-lapse imaging of the remodelling vasculature of the quail embryo yolk sac with flow quantification using a combination of micro-Particle Image Velocimetry and computational fluid dynamics. We then used the flow and remodelling data to inform a model of EC migration during remodelling. To obtain the relation between shear stress and velocity in vitro for EC cells, we developed a flow chamber to assess how confluent sheets of ECs migrate in response to shear stress. Using these data as an input, we developed a multiphase, self-propelled particles (SPP) model where individual agents are driven to migrate based on the level of shear stress while maintaining appropriate spatial relationship to nearby agents. These agents elongate, interact with each other, and with avascular agents at each time-step of the model. We compared predicted vascular shape to real vascular shape after 4 hours from our time-lapse movies and performed sensitivity analysis on the various model parameters. Our model shows that shear stress has the largest effect on the remodelling process. Importantly, however, elongation played an especially important part in remodelling. This model provides a powerful tool to study the input of different biological processes on remodelling.


Assuntos
Hidrodinâmica , Remodelação Vascular , Animais , Circulação Sanguínea , Movimento Celular/fisiologia , Forma Celular , Biologia Computacional , Células Endoteliais/fisiologia , Codorniz/anatomia & histologia , Codorniz/embriologia , Estresse Mecânico
9.
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
10.
Basic Res Cardiol ; 115(4): 39, 2020 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-32451732

RESUMO

Heart failure with preserved ejection fraction (HFpEF) is a complex heterogeneous disease for which our pathophysiological understanding is still limited and specific prevention and treatment strategies are lacking. HFpEF is characterised by diastolic dysfunction and cardiac remodelling (fibrosis, inflammation, and hypertrophy). Recently, microvascular dysfunction and chronic low-grade inflammation have been proposed to participate in HFpEF development. Furthermore, several recent studies demonstrated the occurrence of generalized lymphatic dysfunction in experimental models of risk factors for HFpEF, including obesity, hypercholesterolaemia, type 2 diabetes mellitus (T2DM), hypertension, and aging. Here, we review the evidence for a combined role of coronary (micro)vascular dysfunction and lymphatic vessel alterations in mediating key pathological steps in HFpEF, including reduced cardiac perfusion, chronic low-grade inflammation, and myocardial oedema, and their impact on cardiac metabolic alterations (oxygen and nutrient supply/demand imbalance), fibrosis, and cardiomyocyte stiffness. We focus primarily on HFpEF caused by metabolic risk factors, such as obesity, T2DM, hypertension, and aging.


Assuntos
Endotélio Vascular/patologia , Insuficiência Cardíaca/fisiopatologia , Vasos Linfáticos/patologia , Envelhecimento/patologia , Animais , Diabetes Mellitus Tipo 2/complicações , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Humanos , Hipertensão/complicações , Microvasos/patologia , Obesidade/complicações
11.
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
12.
Development ; 142(23): 4158-67, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26443647

RESUMO

Normal vascular development requires blood flow. Time-lapse imaging techniques have revolutionised our understanding of developmental biology, but measuring changes in blood flow dynamics has met with limited success. Ultrasound biomicroscopy and optical coherence tomography can concurrently image vascular structure and blood flow velocity, but these techniques lack the resolution to accurately calculate fluid forces such as shear stress. This is important because hemodynamic forces are biologically active and induce changes in the expression of genes important for vascular development. Regional variations in shear stress, rather than the overall level, control processes such as vessel enlargement and regression during vascular remodelling. We present a technique to concurrently visualise vascular remodelling and blood flow dynamics. We use an avian embryonic model and inject an endothelial-specific dye and fluorescent microspheres. The motion of the microspheres is captured with a high-speed camera and the velocity of the blood flow in and out of the region of interest is quantified by micro-particle image velocitymetry (µPIV). The vessel geometry and flow are used to numerically solve the flow physics with computational fluid dynamics (CFD). Using this technique, we can analyse changes in shear stress, pressure drops and blood flow velocities over a period of 10 to 16 h. We apply this to study the relationship between shear stress and chronic changes in vessel diameter during embryonic development, both in normal development and after TGFß stimulation. This technique allows us to study the interaction of biomolecular and biomechanical signals during vascular remodelling using an in vivo developmental model.


Assuntos
Hemodinâmica/fisiologia , Remodelação Vascular/fisiologia , Animais , Fenômenos Biomecânicos , Velocidade do Fluxo Sanguíneo/fisiologia , Simulação por Computador , Coturnix , Hematócrito , Microesferas , Modelos Cardiovasculares , Reologia , Resistência ao Cisalhamento , Estresse Mecânico , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo
13.
Development ; 142(23): 4151-7, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26552886

RESUMO

Angiogenesis is tightly controlled by a number of signalling pathways. Although our understanding of the molecular mechanisms involved in angiogenesis has rapidly increased, the role that biomechanical signals play in this process is understudied. We recently developed a technique to simultaneously analyse flow dynamics and vascular remodelling by time-lapse microscopy in the capillary plexus of avian embryos and used this to study the hemodynamic environment present during angiogenic sprouting. We found that sprouts always form from a vessel at lower pressure towards a vessel at higher pressure, and that sprouts form at the location of a shear stress minimum, but avoid locations where two blood streams merge even if this point is at a lower level of shear stress than the sprouting location. Using these parameters, we were able to successfully predict sprout location in quail embryos. We also found that the pressure difference between two vessels is permissive to elongation, and that sprouts will either change direction or regress if the pressure difference becomes negative. Furthermore, the sprout elongation rate is proportional to the pressure difference between the two vessels. Our results show that flow dynamics are predictive of the location of sprout formation in perfused vascular networks and that pressure differences across the interstitium can guide sprout elongation.


Assuntos
Neovascularização Fisiológica/fisiologia , Codorniz/embriologia , Animais , Fenômenos Biomecânicos , Células Endoteliais/citologia , Hemodinâmica , Hidrodinâmica , Processamento de Imagem Assistida por Computador , Microscopia de Fluorescência , Morfogênese , Pressão , Resistência ao Cisalhamento , Estresse Mecânico , Imagem com Lapso de Tempo
14.
Angiogenesis ; 20(3): 373-384, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28374123

RESUMO

Growth factors, such as VEGF, promote the sprouting of new blood vessels. Growth factors are generally produced far from the endothelium, and the transport of these proteins is often assumed to occur through diffusion. When sprouting occurs in a perfused vascular bed, however, interstitial flow is present that can modify protein transport. We recently developed a technique to analyze flow dynamics and vascular remodeling simultaneously in avian embryos. In this study, we extend our technique to model interstitial flow through the porous matrix of the mesenchymal tissue and use this to investigate how flow in the blood vessels affects the distribution of growth factors in the mesenchyme, using VEGF as a prototypical angiogenic molecule. We find that flow controls sprouting location and elongation, both through the direct action of mechanical force and through indirect effects on growth factor distribution. Most importantly, we find that the distribution of VEGF is regulated by interstitial flow, and the effect of diffusion of VEGF is negligible.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mecanotransdução Celular , Neovascularização Fisiológica , Fluxo Sanguíneo Regional , Animais , Mecanotransdução Celular/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Fluxo Sanguíneo Regional/efeitos dos fármacos , Estresse Mecânico , Fator A de Crescimento do Endotélio Vascular/farmacologia
15.
Arterioscler Thromb Vasc Biol ; 34(7): 1468-76, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24764455

RESUMO

OBJECTIVE: The H2.0-like homeobox transcription factor (HLX) plays an essential role in visceral organogenesis in mice and has been shown to regulate angiogenic sprouting in vitro and in zebrafish embryos. We therefore examined the role of HLX in vascular development in mouse and avian embryos. APPROACH AND RESULTS: In situ hybridization showed that Hlx is expressed in a subset of sprouting blood vessels in postnatal mouse retinas and embryos. Hlx expression was conserved in quail embryos and upregulated in blood vessels at the onset of circulation. In vitro assays showed that Hlx is dynamically regulated by growth factors and shear stress alterations. Proangiogenic vascular endothelial growth factor induces Hlx expression in cultured endothelial cells, whereas signals that induce stalk cell identity lead to a reduction in Hlx expression. HLX was also downregulated in embryos in which flow was ablated, whereas injection of a starch solution, which increases blood viscosity and therefore shear stress, causes an upregulation in HLX. HLX knockdown in vitro resulted in a reduction in tip cell marker expression and in reduced angiogenic sprouting, but Hlx(-/-) embryos showed no defect in vascular sprouting at E8.5, E9.5, or E11.5 in vivo. Vascular remodeling of the capillary plexus was altered in Hlx(-/-) embryos, with a modestly enlarged venous plexus and reduction of the arterial plexus. CONCLUSIONS: Our findings indicate not only that Hlx regulates sprouting in vitro, but that its role in sprouting is nonessential in vivo. We find HLX is regulated by shear stress and a subtle defect in vascular remodeling is present in knockout embryos.


Assuntos
Vasos Sanguíneos/metabolismo , Proteínas de Homeodomínio/metabolismo , Neovascularização Fisiológica , Fatores de Transcrição/metabolismo , Saco Vitelino/irrigação sanguínea , Animais , Vasos Sanguíneos/embriologia , Viscosidade Sanguínea , Células Cultivadas , Embrião de Mamíferos/irrigação sanguínea , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Proteínas de Homeodomínio/genética , Humanos , Mecanotransdução Celular , Camundongos , Camundongos Knockout , Codorniz , Interferência de RNA , Fluxo Sanguíneo Regional , Estresse Mecânico , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transfecção
16.
J Biomech ; 171: 112180, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38906711

RESUMO

In the Ross procedure, a patient's pulmonary valve is transplanted in the aortic position. Despite advantages of this surgery, reoperation is still needed in many cases due to excessive dilatation of the pulmonary autograft. To further understand the failure mechanisms, we propose a multiscale model predicting adaptive processes in the autograft at the cell and tissue scale. The cell-scale model consists of a network model, that includes important signaling pathways and relations between relevant transcription factors and their target genes. The resulting gene activity leads to changes in the mechanical properties of the tissue, modeled as a constrained mixture of collagen, elastin and smooth muscle. The multiscale model is calibrated with findings from experiments in which seven sheep underwent the Ross procedure. The model is then validated against a different set of sheep experiments, for which a qualitative agreement between model and experiment is found. Model outcomes at the cell scale, including the activity of genes and transcription factors, also match experimentally obtained transcriptomics data.


Assuntos
Valva Pulmonar , Valva Pulmonar/cirurgia , Valva Pulmonar/transplante , Animais , Ovinos , Autoenxertos , Transdução de Sinais , Modelos Cardiovasculares , Simulação por Computador , Humanos , Valva Aórtica/cirurgia , Valva Aórtica/patologia
17.
Eur Heart J Open ; 4(1): oead129, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38174347

RESUMO

Aims: Microvascular dysfunction has been proposed to drive heart failure with preserved ejection fraction (HFpEF), but the initiating molecular and cellular events are largely unknown. Our objective was to determine when microvascular alterations in HFpEF begin, how they contribute to disease progression, and how pericyte dysfunction plays a role herein. Methods and results: Microvascular dysfunction, characterized by inflammatory activation, loss of junctional barrier function, and altered pericyte-endothelial crosstalk, was assessed with respect to the development of cardiac dysfunction, in the Zucker fatty and spontaneously hypertensive (ZSF1) obese rat model of HFpEF at three time points: 6, 14, and 21 weeks of age. Pericyte loss was the earliest and strongest microvascular change, occurring before prominent echocardiographic signs of diastolic dysfunction were present. Pericytes were shown to be less proliferative and had a disrupted morphology at 14 weeks in the obese ZSF1 animals, who also exhibited an increased capillary luminal diameter and disrupted endothelial junctions. Microvascular dysfunction was also studied in a mouse model of chronic reduction in capillary pericyte coverage (PDGF-Bret/ret), which spontaneously developed many aspects of diastolic dysfunction. Pericytes exposed to oxidative stress in vitro showed downregulation of cell cycle-associated pathways and induced a pro-inflammatory state in endothelial cells upon co-culture. Conclusion: We propose pericytes are important for maintaining endothelial cell function, where loss of pericytes enhances the reactivity of endothelial cells to inflammatory signals and promotes microvascular dysfunction, thereby accelerating the development of HFpEF.

18.
Semin Cell Dev Biol ; 22(9): 1028-35, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22001248

RESUMO

Within a day of gastrulation, the embryonic heart begins to beat and creates blood flow in the developing cardiovascular system. The onset of blood flow completely changes the environment in which the cardiovascular system is forming. Flow provides physiological feedback such that the developing network adapts to cue provided by the flow. Targeted inactivation of genes that alter early blood fluid dynamics induce secondary defects in the heart and vasculature and therefore proper blood flow is known to be essential for vascular development. Though hemodynamics, or blood fluid dynamics, are known to activate signaling pathways in the mature cardiovascular system in pathologies ranging from artherosclerosis to angiogenesis, the role in development has not been as intensively studied. The question arises how blood vessels in the embryos, which initially lack cells types such as smooth muscle cells, differ in their response to mechanical signals from blood flow as compared to the more mature cardiovascular system. Many genes known to be regulated by hemodynamics in the adult are important for developmental angiogenesis. Therefore the onset of blood flow is of primary importance to vascular development. This review will focus on how blood flow initiates and the effects of the mechanical signals created by blood flow on cardiovascular development.


Assuntos
Sistema Cardiovascular/crescimento & desenvolvimento , Animais , Sistema Cardiovascular/embriologia , Hemodinâmica/fisiologia , Humanos , Neovascularização Fisiológica/fisiologia , Fluxo Sanguíneo Regional/fisiologia
19.
Dev Biol ; 369(2): 330-9, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22820069

RESUMO

The glycocalyx, and the thicker endothelial surface layer (ESL), are necessary both for endothelial barrier function and for sensing mechanical forces in the adult. The goal of this study is to use a combination of imaging techniques to establish when the glycocalyx and endothelial surface layer form during embryonic development and to determine the biological significance of the glycocalyx layer during vascular development in quail embryos. Using transmission electron microscopy, we show that the glycocalyx layer is present as soon as blood flow starts (14 somites). The early endothelial glycocalyx (14 somites) lacks the distinct hair-like morphology that is present later in development (17 and 25 somites). The average thickness does not change significantly (14 somites, 182 nm ± 33 nm; 17 somites, 218 ± 30 nm; 25 somites, 212 ± 32 nm). The trapping of circulating fluorescent albumin was used to evaluate the development of the ESL. Trapped fluorescent albumin was first observed at 25 somites. In order to assess a functional role for the glycocalyx during development, we selectively degraded luminal glycosaminoglycans. Degradation of hyaluronan compromised endothelial barrier function and prevented vascular remodeling. Degradation of heparan sulfate down regulated the expression of shear-sensitive genes but does not inhibit vascular remodeling. Our findings show that the glycocalyx layer is present as soon as blood flow starts (14 somites). Selective degradations of major glycocalyx components were shown to inhibit normal vascular development, examined through morphology, vascular barrier function, and gene expression.


Assuntos
Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Coturnix/embriologia , Coturnix/metabolismo , Glicocálix/metabolismo , Animais , Permeabilidade Capilar , Coturnix/genética , Endotélio Vascular/embriologia , Endotélio Vascular/metabolismo , Corantes Fluorescentes , Regulação da Expressão Gênica no Desenvolvimento , Hemodinâmica , Ácido Hialurônico/metabolismo , Imuno-Histoquímica , Microscopia Confocal , Neovascularização Fisiológica/genética , Somitos/embriologia , Somitos/metabolismo
20.
Angiogenesis ; 16(1): 71-83, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22941228

RESUMO

Vascular development is believed to occur first by vasculogenesis followed by angiogenesis. Though angiogenesis is the formation of new vessels, we found that vascular density actually decreases during this second stage. The onset of the decrease coincided with the entry of erythroblasts into circulation. We therefore measured the level of shear stress at various developmental stages and found that it was inversely proportional to vascular density. To investigate whether shear stress was inhibitory to angiogenesis, we altered shear stress levels either by preventing erythroblasts from entering circulation ("low" shear stress) or by injection of a starch solution to increase the blood plasma viscosity ("high" shear stress). By time-lapse microscopy, we show that reverse intussusception (merging of two vessels) is inversely proportional to the level of shear stress. We also found that angiogenesis (both sprouting and splitting) was inversely proportional to shear stress levels. These effects were specific to the arterial or venous plexus however, such that the effect on reverse intussusception was present only in the arterial plexus and the effect on sprouting only in the venous plexus. We cultured embryos under altered shear stress in the presence of either DAPT, a Notch inhibitor, or DMH1, an inhibitor of the bone morphogenetic protein (BMP) pathway. DAPT treatment phenocopied the inhibition of erythroblast circulation ("low" shear stress) and the effect of DAPT treatment could be partially rescued by injection of starch. Inhibition of the BMP signaling prevented the reduction in vascular density that was observed when starch was injected to increase shear stress levels.


Assuntos
Artérias/embriologia , Artérias/fisiologia , Neovascularização Fisiológica , Estresse Mecânico , Veias/embriologia , Veias/fisiologia , Animais , Viscosidade Sanguínea , Proteínas Morfogenéticas Ósseas/metabolismo , Coturnix/embriologia , Coturnix/fisiologia , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/fisiologia , Embrião não Mamífero/irrigação sanguínea , Embrião não Mamífero/fisiologia , Intussuscepção/embriologia , Intussuscepção/fisiopatologia , Camundongos , Receptores Notch/metabolismo , Fluxo Sanguíneo Regional , Transdução de Sinais , Imagem com Lapso de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA