Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 608
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 322(1): H57-H65, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34797171

RESUMO

Cardiovascular disease risk increases with age regardless of sex. Some of this risk is attributable to alterations in natural hormones throughout the life span. The quintessential example of this being the dramatic increase in cardiovascular disease following the transition to menopause. Plasma levels of adiponectin, a "cardioprotective" adipokine released primarily by adipose tissue and regulated by hormones, also fluctuate throughout one's life. Plasma adiponectin levels increase with age in both men and women, with higher levels in both pre- and postmenopausal women compared with men. Younger cohorts seem to confer cardioprotective benefits from increased adiponectin levels yet elevated levels in the elderly and those with existing heart disease are associated with poor cardiovascular outcomes. Here, we review the most recent data regarding adiponectin signaling in the vasculature, highlight the differences observed between the sexes, and shed light on the apparent paradox regarding increased cardiovascular disease risk despite rising plasma adiponectin levels over time.


Assuntos
Adiponectina/metabolismo , Envelhecimento/metabolismo , Endotélio Vascular/metabolismo , Animais , Endotélio Vascular/crescimento & desenvolvimento , Humanos , Transdução de Sinais
2.
Am J Pathol ; 191(12): 2245-2264, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34563512

RESUMO

Whether alterations in the microtubule cytoskeleton affect the ability of endothelial cells (ECs) to sprout and form branching networks of tubes was investigated in this study. Bioassays of human EC tubulogenesis, where both sprouting behavior and lumen formation can be rigorously evaluated, were used to demonstrate that addition of the microtubule-stabilizing drugs, paclitaxel, docetaxel, ixabepilone, and epothilone B, completely interferes with EC tip cells and sprouting behavior, while allowing for EC lumen formation. In bioassays mimicking vasculogenesis using single or aggregated ECs, these drugs induce ring-like lumens from single cells or cyst-like spherical lumens from multicellular aggregates with no evidence of EC sprouting behavior. Remarkably, treatment of these cultures with a low dose of the microtubule-destabilizing drug, vinblastine, led to an identical result, with complete blockade of EC sprouting, but allowing for EC lumen formation. Administration of paclitaxel in vivo markedly interfered with angiogenic sprouting behavior in developing mouse retina, providing corroboration. These findings reveal novel biological activities for pharmacologic agents that are widely utilized in multidrug chemotherapeutic regimens for the treatment of human malignant cancers. Overall, this work demonstrates that manipulation of microtubule stability selectively interferes with the ability of ECs to sprout, a necessary step to initiate and form branched capillary tube networks.


Assuntos
Inibidores da Angiogênese/farmacologia , Neovascularização Patológica/tratamento farmacológico , Paclitaxel/farmacologia , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/crescimento & desenvolvimento , Células Cultivadas , Docetaxel/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/crescimento & desenvolvimento , Epotilonas/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/efeitos dos fármacos , Neovascularização Patológica/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Paclitaxel/análogos & derivados
3.
Arterioscler Thromb Vasc Biol ; 41(12): 2974-2989, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34615374

RESUMO

OBJECTIVE: Pathological angiogenesis is a hallmark of various diseases characterized by local hypoxia and inflammation. These disorders can be treated with inhibitors of angiogenesis, but current compounds display a variety of side effects and lose efficacy over time. This makes the identification of novel signaling pathways and pharmacological targets involved in angiogenesis a top priority. Approach and Results: Here, we show that inactivation of FAAH (fatty acid amide hydrolase), the enzyme responsible for degradation of the endocannabinoid anandamide, strongly impairs angiogenesis in vitro and in vivo. Both, the pharmacological FAAH inhibitor URB597 and anandamide induce downregulation of gene sets for cell cycle progression and DNA replication in endothelial cells. This is underscored by cell biological experiments, in which both compounds inhibit proliferation and migration and evoke cell cycle exit of endothelial cells. This prominent antiangiogenic effect is also of pathophysiological relevance in vivo, as laser-induced choroidal neovascularization in the eye of FAAH-/- mice is strongly reduced. CONCLUSIONS: Thus, elevation of endogenous anandamide levels by FAAH inhibition represents a novel antiangiogenic mechanism.


Assuntos
Amidoidrolases/farmacocinética , Ácidos Araquidônicos/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Endocanabinoides/farmacologia , Endotélio Vascular/crescimento & desenvolvimento , Músculo Liso Vascular/efeitos dos fármacos , Alcamidas Poli-Insaturadas/farmacologia , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/patologia , Agonistas de Receptores de Canabinoides/farmacologia , Bovinos , Linhagem Celular , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Humanos , Camundongos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Neovascularização Patológica
4.
Nature ; 529(7585): 216-20, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26735015

RESUMO

Endothelial cells (ECs) are plastic cells that can switch between growth states with different bioenergetic and biosynthetic requirements. Although quiescent in most healthy tissues, ECs divide and migrate rapidly upon proangiogenic stimulation. Adjusting endothelial metabolism to the growth state is central to normal vessel growth and function, yet it is poorly understood at the molecular level. Here we report that the forkhead box O (FOXO) transcription factor FOXO1 is an essential regulator of vascular growth that couples metabolic and proliferative activities in ECs. Endothelial-restricted deletion of FOXO1 in mice induces a profound increase in EC proliferation that interferes with coordinated sprouting, thereby causing hyperplasia and vessel enlargement. Conversely, forced expression of FOXO1 restricts vascular expansion and leads to vessel thinning and hypobranching. We find that FOXO1 acts as a gatekeeper of endothelial quiescence, which decelerates metabolic activity by reducing glycolysis and mitochondrial respiration. Mechanistically, FOXO1 suppresses signalling by MYC (also known as c-MYC), a powerful driver of anabolic metabolism and growth. MYC ablation impairs glycolysis, mitochondrial function and proliferation of ECs while its EC-specific overexpression fuels these processes. Moreover, restoration of MYC signalling in FOXO1-overexpressing endothelium normalizes metabolic activity and branching behaviour. Our findings identify FOXO1 as a critical rheostat of vascular expansion and define the FOXO1-MYC transcriptional network as a novel metabolic checkpoint during endothelial growth and proliferation.


Assuntos
Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Animais , Proliferação de Células , Respiração Celular , Endotélio Vascular/citologia , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Glicólise , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-myc/deficiência , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais
5.
Cell Mol Life Sci ; 77(5): 885-901, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31278420

RESUMO

Purinergic P2 receptors are critical regulators of several functions within the vascular system, including platelet aggregation, vascular inflammation, and vascular tone. However, a role for ATP release and P2Y receptor signalling in angiogenesis remains poorly defined. Here, we demonstrate that blood vessel growth is controlled by P2Y2 receptors. Endothelial sprouting and vascular tube formation were significantly dependent on P2Y2 expression and inhibition of P2Y2 using a selective antagonist blocked microvascular network generation. Mechanistically, overexpression of P2Y2 in endothelial cells induced the expression of the proangiogenic molecules CXCR4, CD34, and angiopoietin-2, while expression of VEGFR-2 was decreased. Interestingly, elevated P2Y2 expression caused constitutive phosphorylation of ERK1/2 and VEGFR-2. However, stimulation of cells with the P2Y2 agonist UTP did not influence sprouting unless P2Y2 was constitutively expressed. Finally, inhibition of VEGFR-2 impaired spontaneous vascular network formation induced by P2Y2 overexpression. Our data suggest that P2Y2 receptors have an essential function in angiogenesis, and that P2Y2 receptors present a therapeutic target to regulate blood vessel growth.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/crescimento & desenvolvimento , Neovascularização Fisiológica/fisiologia , Receptores Purinérgicos P2Y2/metabolismo , Angiopoietina-2/biossíntese , Antígenos CD34/biossíntese , Células Cultivadas , Humanos , Proteína Quinase 1 Ativada por Mitógeno/biossíntese , Proteína Quinase 3 Ativada por Mitógeno/biossíntese , Fosforilação/fisiologia , Agregação Plaquetária/fisiologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores CXCR4/biossíntese , Receptores Purinérgicos P2Y2/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese
6.
Cell Mol Life Sci ; 77(24): 5299-5320, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32166394

RESUMO

Stem/progenitor cells (SPCs) have been implicated to participate in vascular repair. However, the exact role of SPCs in endothelial repair of large vessels still remains controversial. This study aimed to delineate the cellular heterogeneity and possible functional role of endogenous vascular SPCs in large vessels. Using single-cell RNA-sequencing (scRNA-seq) and genetic lineage tracing mouse models, we uncovered the cellular heterogeneity of SPCs, i.e., c-Kit+ cells in the mouse aorta, and found that endogenous c-Kit+ cells acquire endothelial cell fate in the aorta under both physiological and pathological conditions. While c-Kit+ cells contribute to aortic endothelial turnover in the atheroprone regions during homeostasis, recipient c-Kit+ cells of nonbone marrow source replace both luminal and microvessel endothelial cells in transplant arteriosclerosis. Single-cell pseudotime analysis of scRNA-seq data and in vitro cell experiments suggest that vascular SPCs display endothelial differentiation potential and undergo metabolic reprogramming during cell differentiation, in which AKT/mTOR-dependent glycolysis is critical for endothelial gene expression. These findings demonstrate a critical role for c-Kit lineage cells in aortic endothelial turnover and replacement, and may provide insights into therapeutic strategies for vascular diseases.


Assuntos
Linhagem da Célula/genética , Endotélio Vascular/crescimento & desenvolvimento , Análise de Célula Única/métodos , Células-Tronco/metabolismo , Animais , Aorta/crescimento & desenvolvimento , Aorta/metabolismo , Diferenciação Celular/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-kit/genética , RNA-Seq , Células-Tronco/citologia , Serina-Treonina Quinases TOR/genética
7.
Int J Mol Sci ; 22(8)2021 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-33920100

RESUMO

Chitinase 3-like 1 (CHI3L1) is an enzymatically inactive mammalian chitinase that is associated with tumor inflammation. Previous research indicated that CHI3L1 is able to interact with different extracellular matrix components, such as heparan sulfate. In the present work, we investigated whether the interaction of CHI3L1 with the extracellular matrix of melanoma cells can trigger an inflammatory activation of endothelial cells. The analysis of the melanoma cell secretome indicated that CHI3L1 increases the abundance of various cytokines, such as CC-chemokine ligand 2 (CCL2), and growth factors, such as vascular endothelial growth factor A (VEGF-A). Using a solid-phase binding assay, we found that heparan sulfate-bound VEGF-A and CCL2 were displaced by recombinant CHI3L1 in a dose-dependent manner. Microfluidic experiments indicated that the CHI3L1 altered melanoma cell secretome promoted immune cell recruitment to the vascular endothelium. In line with the elevated VEGF-A levels, CHI3L1 was also able to promote angiogenesis through the release of extracellular matrix-bound pro-angiogenic factors. In conclusion, we showed that CHI3L1 is able to affect the tumor cell secretome, which in turn can regulate immune cell recruitment and blood vessel formation. Accordingly, our data suggest that the molecular targeting of CHI3L1 in the course of cancer immunotherapies can tune patients' response and antitumoral inflammation.


Assuntos
Quimiocina CCL2/genética , Proteína 1 Semelhante à Quitinase-3/genética , Melanoma/genética , Neovascularização Patológica/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/patologia , Linhagem Celular Tumoral , Células Endoteliais/imunologia , Células Endoteliais/patologia , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Matriz Extracelular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicosaminoglicanos/farmacologia , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Melanoma/imunologia , Melanoma/patologia , Técnicas Analíticas Microfluídicas , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Ligação Proteica/genética , Ligação Proteica/imunologia
8.
Angiogenesis ; 23(2): 75-77, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31993833

RESUMO

Vascular endothelial growth factor-A (VEGF-A/VEGF) interaction with VEGF receptor 2 (VEGFR2) is key for sprouting angiogenesis in health and disease. VEGF/VEGFR2 signaling promotes endothelial proliferation and migration, as well as the hierarchical organization into leader (tip) and follower (stalk) cells via a dynamic interplay with Notch. Recent studies reveal novel molecular mechanisms to fine-tune VEGF/Notch signaling and tip/stalk cell function during sprouting angiogenesis.


Assuntos
Diferenciação Celular , Células Endoteliais/fisiologia , Retroalimentação Fisiológica/fisiologia , Neovascularização Fisiológica/fisiologia , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências , Morfogênese/genética , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Cell Biol Int ; 44(12): 2588-2597, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32808723

RESUMO

Dermal microvascular endothelial cells (DMECs) play central roles in inflammation and angiogenesis and have become important cell models for studying various skin diseases. However, primary DMECs are difficult to culture and often contaminated by mesenchymal stem cells, fibroblasts, and other stromal cells. Surgically removed superfluous foreskin was first cut into pieces, digested with two types of enzymes, and dispersed into single cells. Cells obtained from the dermis were then subjected to Percoll density gradient centrifugation and cells located between densities 1.033 and 1.047 g/ml were further purified with endothelial growth medium containing decreasing concentrations of puromycin. Obtained HDMECs were identified by microscopy, flow cytometry, quantitative reverse-transcription polymerase chain reaction, western blot analysis, and immunofluorescent staining. The expression of CD31 (PECAM-1), CD34, VEGFR2, VWF (Von Willebrand Factor), VE-Cadherin (CD144), and NOS was positive. HDMECs were found to have abilities of angiogenesis and uptake of acetylated low-density lipoprotein. Growth curves and cell viability were analyzed, and a growth pattern consisting of the "latency phase-logarithmic growth phase-stagnation phase" was determined. In this study, a simple, rapid, effective, and low-cost method is established to isolate HDMECs from the foreskin with a purity of over 91% and high viability. The method showed good repeatability and allowed a stable passage. This study provides technical support and theoretical guidance for studying the physiological characteristics of HDMECs, the pathogenesis of the skin associated, and other microvascular diseases.


Assuntos
Separação Celular/métodos , Derme/citologia , Endotélio Vascular/crescimento & desenvolvimento , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , China , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Citometria de Fluxo , Humanos , Microvasos/metabolismo , Pele/metabolismo
10.
Artif Organs ; 44(4): E172-E180, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31736099

RESUMO

Engineering of functional vascularized pancreatic tissues offers an alternative way to solve the perpetual shortage of organs for transplantation. However, revascularization remains a major bottleneck in biological engineering, which limited the further clinical applications of this strategy. In this study, an efficient approach for enhancing re-endothelialization of rat decellularized pancreatic scaffolds (DPS) was presented, by conjugating with GRGDSPC peptide to maximize coverage of the vessel walls with human umbilical vein endothelial cells (HUVECs). First, pancreas was perfused with 1% Triton X-100 and 0.1% ammonium hydroxide to remove the cellular components. Subsequently, GRGDSPC was covalently coupled to the vasculature of DPS and re-seeded with HUVECs via perfusion of the portal vein in the bioreactor. After the re-endothelialized scaffolds were created, in vitro and in vivo experiments were undertaken to evaluate the angiogenesis. Our results demonstrated that GRGDSPC-conjugated scaffolds could support the survival and accelerated the proliferation of HUVECs; angiogenesis was also significantly improved over untreated scaffolds. In conclusion, GRGDSPC-conjugated scaffolds showed great potential for the generation of functional bioengineered pancreatic tissue suitable for long-term transplantation.


Assuntos
Endotélio Vascular/crescimento & desenvolvimento , Neovascularização Fisiológica/efeitos dos fármacos , Oligopeptídeos/farmacologia , Pâncreas/irrigação sanguínea , Alicerces Teciduais , Animais , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos Endogâmicos BALB C , Ratos Sprague-Dawley
11.
Growth Factors ; 37(1-2): 53-67, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31284789

RESUMO

Angiogenic blood vessel growth is essential to ensure organs receive adequate blood supply to support normal organ function and homeostasis. Angiogenesis involves a complex series of cellular events through which new vessels grow out from existing vasculature. Growth factor signaling, layered over a range of other signaling inputs, orchestrates this process. The response of endothelial cells (ECs) to growth factor signals must be carefully controlled through feedback mechanisms to prevent excessive vessel growth, remodeling or destabilization. In this article, we summarize recent findings describing how ECs respond to growth factor signals during blood vessel development and homeostasis and how perturbation of these responses can lead to disease.


Assuntos
Neovascularização Fisiológica , Transdução de Sinais , Doenças Vasculares/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Humanos , Doenças Vasculares/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
12.
Angiogenesis ; 22(2): 237-250, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30446855

RESUMO

The coronary vasculature is crucial for normal heart function, yet much remains to be learned about its development, especially the maturation of coronary arterial endothelium. Here, we show that endothelial inactivation of ADAM10, a key regulator of Notch signaling, leads to defects in coronary arterial differentiation, as evidenced by dysregulated genes related to Notch signaling and arterial identity. Moreover, transcriptome analysis indicated reduced EGFR signaling in A10ΔEC coronary endothelium. Further analysis revealed that A10ΔEC mice have enlarged dysfunctional hearts with abnormal myocardial compaction, and increased expression of venous and immature endothelium markers. These findings provide the first evidence for a potential role for endothelial ADAM10 in cardioprotective homeostatic EGFR signaling and implicate ADAM10/Notch signaling in coronary arterial cell specification, which is vital for normal heart development and function. The ADAM10/Notch signaling pathway thus emerges as a potential therapeutic target for improving the regenerative capacity and maturation of the coronary vasculature.


Assuntos
Proteína ADAM10/fisiologia , Secretases da Proteína Precursora do Amiloide/fisiologia , Diferenciação Celular/genética , Vasos Coronários/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Proteínas de Membrana/fisiologia , Animais , Vasos Coronários/citologia , Vasos Coronários/crescimento & desenvolvimento , Endotélio Vascular/crescimento & desenvolvimento , Feminino , Coração/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/genética
13.
Am J Physiol Heart Circ Physiol ; 316(1): H80-H88, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30289292

RESUMO

Homodimer formation is essential for the normal activity of endothelial nitric oxide synthase (eNOS). Structural uncoupling of eNOS, with generation of enzyme monomers, is thought to contribute to endothelial dysfunction in several vascular disorders, including aging. However, low-temperature SDS-PAGE of healthy arteries has revealed considerable variation between studies in the relative expression of eNOS dimers and monomers. While assessing structural uncoupling of eNOS in aging arteries, we identified methodological pitfalls that might contribute to such variation. Therefore, using human cultured aortic endothelial cells and aortas from young and aged Fischer-344 rats, we investigated optimal approaches for analyzing the expression of eNOS monomers and dimers. The results demonstrated that published differences in treatment of cell lysates can significantly impact the relative expression of several eNOS species, including denatured monomers, partially folded monomers, dimers, and higher-order oligomers. In aortas, experiments initially confirmed a large increase in eNOS monomers in aging arteries, consistent with structural uncoupling. However, these monomers were actually endogenous IgG, which, under these conditions, has mobility similar to eNOS monomers. Increased IgG levels in aged aortas likely reflect the aging-induced disruption of endothelial junctions and increased arterial penetration of IgG. After removal of the IgG signal, there were low levels of eNOS monomers in young arteries, which were not significantly different in aged arteries. Therefore, structural uncoupling of eNOS is not a prominent feature in young healthy arteries, and the process is not increased by aging. The study also identifies optimal approaches to analyze eNOS dimers and monomers. NEW & NOTEWORTHY Structural uncoupling of endothelial nitric oxide synthase (eNOS) is considered central to endothelial dysfunction. However, reported levels of eNOS dimers and monomers vary widely, even in healthy arteries. We demonstrate that sample processing can alter relative levels of eNOS species. Moreover, endothelial dysfunction in aging aortas results in IgG accumulation, which, because of similar mobility to eNOS monomers, could be misinterpreted as structural uncoupling. Indeed, enzyme monomerization is not prominent in young or aging arteries.


Assuntos
Envelhecimento/metabolismo , Artérias/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Multimerização Proteica , Animais , Artérias/crescimento & desenvolvimento , Artefatos , Células Cultivadas , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Humanos , Immunoblotting/normas , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Óxido Nítrico Sintase Tipo III/química , Óxido Nítrico Sintase Tipo III/genética , Dobramento de Proteína , Ratos , Ratos Endogâmicos F344
14.
Am J Physiol Heart Circ Physiol ; 316(2): H421-H429, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30499713

RESUMO

The heat shock response is an important cytoprotective mechanism for protein homeostasis and is an essential protective response to cellular stress and injury. Studies on changes in the heat shock response with aging have been mixed with regard to whether it is inhibited, and this, at least in part, reflects different tissues and different models. Cellular senescence is a key feature in aging, but work on the heat shock response in cultured senescent (SEN) cells has largely been limited to fibroblasts. Given the prevalence of oxidative injury in the aging cardiovascular system, we investigated whether SEN primary human coronary artery endothelial cells have a diminished heat shock response and impaired proteostasis. In addition, we tested whether this downregulation of heat shock response can be mitigated by 17ß-estradiol (E2), which has a critical cardioprotective role in women, as we have previously reported that E2 improves the heat shock response in endothelial cells (Hamilton KL, Mbai FN, Gupta S, Knowlton AA. Arterioscler Thromb Vasc Biol 24: 1628-1633, 2004). We found that SEN endothelial cells, despite their unexpectedly increased proteasome activity, had a diminished heat shock response and had more protein aggregation than early passage cells. SEN cells had increased oxidative stress, which promoted protein aggregation. E2 treatment did not decrease protein aggregation or improve the heat shock response in either early passage or SEN cells. In summary, cellular senescence in adult human endothelial cells is accompanied by increased oxidative stress and a blunting of proteostasis, and E2 did not mitigate these changes. NEW & NOTEWORTHY Senescent human endothelial cells have a diminished heat shock response and increased protein aggregates. Senescent human endothelial cells have increased basal oxidative stress, which increases protein aggregates. Physiological level of 17ß-estradiol did not improve proteostasis in endothelial cells.


Assuntos
Senescência Celular , Células Endoteliais/metabolismo , Endotélio Vascular/crescimento & desenvolvimento , Estradiol/farmacologia , Estrogênios/farmacologia , Estresse Oxidativo , Proteostase , Adolescente , Adulto , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Endotélio Vascular/metabolismo , Feminino , Resposta ao Choque Térmico , Humanos , Pessoa de Meia-Idade
15.
Am J Respir Crit Care Med ; 197(6): 776-787, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29268623

RESUMO

RATIONALE: Pregnancies complicated by antenatal stress, including preeclampsia (PE) and chorioamnionitis (CA), increase the risk for bronchopulmonary dysplasia (BPD) in preterm infants, but biologic mechanisms linking prenatal factors with BPD are uncertain. Levels of sFlt-1 (soluble fms-like tyrosine kinase 1), an endogenous antagonist to VEGF (vascular endothelial growth factor), are increased in amniotic fluid and maternal blood in PE and associated with CA. OBJECTIVES: Because impaired VEGF signaling has been implicated in the pathogenesis of BPD, we hypothesized that fetal exposure to sFlt-1 decreases lung growth and causes abnormal lung structure and pulmonary hypertension during infancy. METHODS: To test this hypothesis, we studied the effects of anti-sFlt-1 monoclonal antibody (mAb) treatment on lung growth in two established antenatal models of BPD that mimic PE and CA induced by intraamniotic (i.a.) injections of sFlt-1 or endotoxin, respectively. In experimental PE, mAb was administered by three different approaches, including antenatal treatment by either i.a. instillation or maternal uterine artery infusion, or by postnatal intraperitoneal injections. RESULTS: With each strategy, mAb therapy improved infant lung structure as assessed by radial alveolar count, vessel density, right ventricular hypertrophy, and lung function. As found in the PE model, the adverse lung effects of i.a. endotoxin were also reduced by antenatal or postnatal mAb therapy. CONCLUSIONS: We conclude that treatment with anti-sFlt-1 mAb preserves lung structure and function and prevents right ventricular hypertrophy in two rat models of BPD of antenatal stress and speculate that early mAb therapy may provide a novel strategy for the prevention of BPD.


Assuntos
Displasia Broncopulmonar/fisiopatologia , Endotélio Vascular/crescimento & desenvolvimento , Pulmão/crescimento & desenvolvimento , Alvéolos Pulmonares/crescimento & desenvolvimento , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/embriologia , Modelos Animais de Doenças , Endotélio Vascular/embriologia , Feminino , Humanos , Pulmão/embriologia , Gravidez , Alvéolos Pulmonares/embriologia , Ratos , Ratos Sprague-Dawley
16.
Int J Mol Sci ; 20(14)2019 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-31311132

RESUMO

Cardiovascular risk associated with fetal growth restriction (FGR) could result from an early impaired vascular function. However, whether this effect results in premature vascular aging has not been addressed. We studied the ex vivo reactivity of carotid and femoral arteries in fetal (near term), adults (eight months-old) and aged (16 months-old) guinea pigs in normal (control) and FGR offspring. Additionally, an epigenetic marker of vascular aging (i.e., LINE-1 DNA methylation) was evaluated in human umbilical artery endothelial cells (HUAEC) from control and FGR subjects. Control guinea pig arteries showed an increased contractile response (KCl-induced) and a progressive impairment of NO-mediated relaxing responses as animals get older. FGR was associated with an initial preserved carotid artery reactivity as well as a later significant impairment in NO-mediated responses. Femoral arteries from FGR fetuses showed an increased contractility but a decreased relaxing response compared with control fetuses, and both responses were impaired in FGR-adults. Finally, FGR-HUAEC showed decreased LINE-1 DNA methylation compared with control-HUAEC. These data suggest that the aging of vascular function occurs by changes in NO-mediated responses, with limited alterations in contractile capacity. Further, these effects are accelerated and imposed at early stages of development in subjects exposed to a suboptimal intrauterine environment.


Assuntos
Envelhecimento/patologia , Endotélio Vascular/crescimento & desenvolvimento , Retardo do Crescimento Fetal/patologia , Animais , Artérias Carótidas/crescimento & desenvolvimento , Artérias Carótidas/patologia , Artérias Carótidas/fisiopatologia , Células Cultivadas , Metilação de DNA , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Feminino , Artéria Femoral/crescimento & desenvolvimento , Artéria Femoral/patologia , Artéria Femoral/fisiopatologia , Retardo do Crescimento Fetal/genética , Cobaias , Humanos , Elementos Nucleotídeos Longos e Dispersos/genética , Óxido Nítrico/metabolismo , Vasoconstrição , Vasodilatação
17.
Am J Physiol Heart Circ Physiol ; 315(6): H1569-H1588, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30216121

RESUMO

Diseases of the cardiovascular system are the leading cause of morbidity and mortality in men and women in developed countries, and cardiovascular disease (CVD) is becoming more prevalent in developing countries. The prevalence of atherosclerotic CVD in men is greater than in women until menopause, when the prevalence of CVD increases in women until it exceeds that of men. Endothelial function is a barometer of vascular health and a predictor of atherosclerosis that may provide insights into sex differences in CVD as well as how and why the CVD risk drastically changes with menopause. Studies of sex differences in endothelial function are conflicting, with some studies showing earlier decrements in endothelial function in men compared with women, whereas others show similar age-related declines between the sexes. Because the increase in CVD risk coincides with menopause, it is generally thought that female hormones, estrogens in particular, are cardioprotective. Moreover, it is often proposed that androgens are detrimental. In truth, the relationships are more complex. This review first addresses female and male sex hormones and their receptors and how these interact with the cardiovascular system, particularly the endothelium, in healthy young women and men. Second, we address sex differences in sex steroid receptor-independent mechanisms controlling endothelial function, focusing on vascular endothelin and the renin-angiotensin systems, in healthy young women and men. Finally, we discuss sex differences in age-associated endothelial dysfunction, focusing on the role of attenuated circulating sex hormones in these effects.


Assuntos
Envelhecimento/fisiologia , Doenças Cardiovasculares/epidemiologia , Endotélio Vascular/metabolismo , Hormônios Gonadais/metabolismo , Animais , Doenças Cardiovasculares/etiologia , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/fisiopatologia , Humanos , Fatores Sexuais
18.
Am J Physiol Heart Circ Physiol ; 315(3): H531-H539, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29750566

RESUMO

Age-related microvascular dysfunction is well characterized in rodents and humans, but little is known about the properties of the microvascular endothelial glycocalyx in advanced age. We examined the glycocalyx in microvessels of young and old male C57BL6 mice (young: 6.1 ± 0.1 mo vs. old: 24.6 ± 0.2 mo) using intravital microscopy and transmission electron microscopy and in human participants (young: 29 ± 1 yr vs. old: 60 ± 2 yr) using intravital microscopy. Glycocalyx thickness in mesenteric and skeletal muscle microvessels was 51-54% lower in old compared with young mice. We also observed 33% lower glycocalyx thickness in the sublingual microcirculation of humans in advanced age. The perfused boundary region, a marker of glycocalyx barrier function, was also obtained using an automated capture and analysis system. In advanced age, we observed a 10-22% greater perfused boundary region in mice and humans, indicating a more penetrable glycocalyx. Finally, using this automated analysis system, we examined perfused microvascular density and red blood cell (RBC) fraction. Perfused microvascular density is a marker of microvascular function that reflects the length of perfused microvessel segments in a given area; RBC fraction represents the heterogeneity in RBC presence between microvessel segments. Compared with young, the perfused microvascular density was 16-21% lower and RBC fraction was 5-14% lower in older mice and in older humans. These data provide novel evidence that, across mammalian species, a diminished glycocalyx is present in advanced age and is accompanied by markers of impaired microvascular perfusion. Age-related glycocalyx deterioration may be an important contributor to microvascular dysfunction in older adults and subsequent pathophysiology. NEW & NOTEWORTHY Advanced age is characterized by microvascular dysfunction that contributes to age-related cardiovascular diseases, but little is known about endothelial glycocalyx properties in advanced age. This study reveals, for the first time, lower glycocalyx thickness and barrier function that is accompanied by impaired microvascular perfusion in both mice and humans in advanced age.


Assuntos
Envelhecimento/metabolismo , Endotélio Vascular/citologia , Glicocálix/ultraestrutura , Adulto , Animais , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Feminino , Glicocálix/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade
19.
Circ Res ; 119(5): 607-20, 2016 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-27354211

RESUMO

RATIONALE: Arterial endothelial cells are morphologically, functionally, and molecularly distinct from those found in veins and lymphatic vessels. How arterial fate is acquired during development and maintained in adult vessels is incompletely understood. OBJECTIVE: We set out to identify factors that promote arterial endothelial cell fate in vivo. METHODS AND RESULTS: We developed a functional assay, allowing us to monitor and manipulate arterial fate in vivo, using arteries isolated from quails that are grafted into the coelom of chick embryos. Endothelial cells migrate out from the grafted artery, and their colonization of host arteries and veins is quantified. Here we show that sympathetic innervation promotes arterial endothelial cell fate in vivo. Removal of sympathetic nerves decreases arterial fate and leads to colonization of veins, whereas exposure to sympathetic nerves or norepinephrine imposes arterial fate. Mechanistically, sympathetic nerves increase endothelial ERK (extracellular signal-regulated kinase) activity via adrenergic α1 and α2 receptors. CONCLUSIONS: These findings show that sympathetic innervation promotes arterial endothelial fate and may lead to novel approaches to improve arterialization in human disease.


Assuntos
Fibras Adrenérgicas/enzimologia , Artérias/enzimologia , Artérias/inervação , Endotélio Vascular/enzimologia , Endotélio Vascular/inervação , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Animais , Artérias/crescimento & desenvolvimento , Movimento Celular/fisiologia , Embrião de Galinha , Membrana Corioalantoide/enzimologia , Membrana Corioalantoide/crescimento & desenvolvimento , Membrana Corioalantoide/inervação , Coturnix , Endotélio Vascular/crescimento & desenvolvimento , Ativação Enzimática/fisiologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Técnicas de Cultura de Órgãos , Sistema Nervoso Periférico/enzimologia , Sistema Nervoso Periférico/crescimento & desenvolvimento , Transplante de Tecidos/métodos , Artérias Umbilicais/enzimologia , Artérias Umbilicais/crescimento & desenvolvimento
20.
Circ Res ; 119(8): e110-26, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27582371

RESUMO

RATIONALE: Sphingosine-1-phosphate (S1P) signaling is essential for vascular development and postnatal vascular homeostasis. The relative importance of S1P sources sustaining these processes remains unclear. OBJECTIVE: To address the level of redundancy in bioactive S1P provision to the developing and mature vasculature. METHODS AND RESULTS: S1P production was selectively impaired in mouse platelets, erythrocytes, endothelium, or smooth muscle cells by targeted deletion of genes encoding sphingosine kinases -1 and -2. S1P deficiency impaired aggregation and spreading of washed platelets and profoundly reduced their capacity to promote endothelial barrier function ex vivo. However, and in contrast to recent reports, neither platelets nor any other source of S1P was essential for vascular development, vascular integrity, or hemostasis/thrombosis. Yet rapid and profound depletion of plasma S1P during systemic anaphylaxis rendered both platelet- and erythrocyte-derived S1P essential for survival, with a contribution from blood endothelium observed only in the absence of circulating sources. Recovery was sensitive to aspirin in mice with but not without platelet S1P, suggesting that platelet activation and stimulus-response coupling is needed. S1P deficiency aggravated vasoplegia in this model, arguing a vital role for S1P in maintaining vascular resistance during recovery from circulatory shock. Accordingly, the S1P2 receptor mediated most of the survival benefit of S1P, whereas the endothelial S1P1 receptor was dispensable for survival despite its importance for maintaining vascular integrity. CONCLUSIONS: Although source redundancy normally secures essential S1P signaling in developing and mature blood vessels, profound depletion of plasma S1P renders both erythrocyte and platelet S1P pools necessary for recovery and high basal plasma S1P levels protective during anaphylactic shock.


Assuntos
Anafilaxia/metabolismo , Plaquetas/metabolismo , Endotélio Vascular/metabolismo , Eritrócitos/metabolismo , Homeostase/fisiologia , Lisofosfolipídeos/deficiência , Esfingosina/análogos & derivados , Anafilaxia/patologia , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esfingosina/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA