Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Transl Med ; 21(1): 199, 2023 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-36927819

RESUMO

BACKGROUND: Increased circulating uric acid (UA) concentration may disrupt cardiac function in heart failure patients, but the specific mechanism remains unclear. Here, we postulate that hyperuremia induces sterol regulatory element binding protein 1 (SREBP1), which in turn activate hepatic fatty acid biosynthesis response, leading to cardiac dysfunction. METHODS AND RESULTS: Increased circulating uric acid was observed in heart failure patients and inversely correlated to cardiac function. Besides, uric acid correlated to circulating lipids profile based on metabolomics in heart failure patients. Using cultured human hepatoellular carcinomas (HepG2) and Tg(myl7:egfp) zebrafish, we demonstrated that UA regulated fatty acid synthase (FASN) via SREBP1 signaling pathway, leading to FFA accumulation and impaired energy metabolism, which could be rescued via SREBP1 knockdown. In ISO treated zebrafish, UA aggravated heart failure via increased cardiovascular cavity size, decreased heart beats, pericardial edema and long-stretched heart deformation. CONCLUSIONS: Our findings suggest that UA-SREBP1-FASN signaling exacerbates cardiac dysfunction during FFA accumulation. Identification of this mechanism may help in treatment and prevention of heart failure.


Assuntos
Cardiopatias , Insuficiência Cardíaca , Animais , Humanos , Ácido Úrico , Peixe-Zebra/metabolismo , Ácido Graxo Sintases/metabolismo , Ácidos Graxos/metabolismo , Insuficiência Cardíaca/complicações
2.
Front Chem ; 10: 869732, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35548679

RESUMO

Metabolic profiling harbors the potential to better understand various disease entities such as cancer, diabetes, Alzheimer's, Parkinson's disease or COVID-19. To better understand such diseases and their intricate metabolic pathways in human studies, model animals are regularly used. There, standardized rearing conditions and uniform sampling strategies are prerequisites towards a successful metabolomic study that can be achieved through model organisms. Although metabolomic approaches have been employed on model organisms before, no systematic assessment of different conditions to optimize metabolite extraction across several organisms and sample types has been conducted. We address this issue using a highly standardized metabolic profiling assay analyzing 630 metabolites across three commonly used model organisms (Drosophila, mouse, and zebrafish) to find an optimal extraction protocol for various matrices. Focusing on parameters such as metabolite coverage, concentration and variance between replicates we compared seven extraction protocols. We found that the application of a combination of 75% ethanol and methyl tertiary-butyl ether (MTBE), while not producing the broadest coverage and highest concentrations, was the most reproducible extraction protocol. We were able to determine up to 530 metabolites in mouse kidney samples, 509 in mouse liver, 422 in zebrafish and 388 in Drosophila and discovered a core overlap of 261 metabolites in these four matrices. To enable other scientists to search for the most suitable extraction protocol in their experimental context and interact with this comprehensive data, we have integrated our data set in the open-source shiny app "MetaboExtract". Hereby, scientists can search for metabolites or compound classes of interest, compare them across the different tested extraction protocols and sample types as well as find reference concentration values.

3.
Hepatology ; 75(2): 322-337, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34435364

RESUMO

BACKGROUND AND AIMS: In patients with acute liver failure (ALF) who suffer from massive hepatocyte loss, liver progenitor cells (LPCs) take over key hepatocyte functions, which ultimately determines survival. This study investigated how the expression of hepatocyte nuclear factor 4α (HNF4α), its regulators, and targets in LPCs determines clinical outcome of patients with ALF. APPROACH AND RESULTS: Clinicopathological associations were scrutinized in 19 patients with ALF (9 recovered and 10 receiving liver transplantation). Regulatory mechanisms between follistatin, activin, HNF4α, and coagulation factor expression in LPC were investigated in vitro and in metronidazole-treated zebrafish. A prospective clinical study followed up 186 patients with cirrhosis for 80 months to observe the relevance of follistatin levels in prevalence and mortality of acute-on-chronic liver failure. Recovered patients with ALF robustly express HNF4α in either LPCs or remaining hepatocytes. As in hepatocytes, HNF4α controls the expression of coagulation factors by binding to their promoters in LPC. HNF4α expression in LPCs requires the forkhead box protein H1-Sma and Mad homolog 2/3/4 transcription factor complex, which is promoted by the TGF-ß superfamily member activin. Activin signaling in LPCs is negatively regulated by follistatin, a hepatocyte-derived hormone controlled by insulin and glucagon. In contrast to patients requiring liver transplantation, recovered patients demonstrate a normal activin/follistatin ratio, robust abundance of the activin effectors phosphorylated Sma and Mad homolog 2 and HNF4α in LPCs, leading to significantly improved coagulation function. A follow-up study indicated that serum follistatin levels could predict the incidence and mortality of acute-on-chronic liver failure. CONCLUSIONS: These results highlight a crucial role of the follistatin-controlled activin-HNF4α-coagulation axis in determining the clinical outcome of massive hepatocyte loss-induced ALF. The effects of insulin and glucagon on follistatin suggest a key role of the systemic metabolic state in ALF.


Assuntos
Ativinas/genética , Folistatina/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Falência Hepática Aguda/metabolismo , Ativinas/metabolismo , Insuficiência Hepática Crônica Agudizada/sangue , Adulto , Idoso , Animais , Coagulação Sanguínea , Linhagem Celular , Fator V/genética , Feminino , Folistatina/sangue , Seguimentos , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica , Fator 4 Nuclear de Hepatócito/genética , Hepatócitos/metabolismo , Humanos , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/patologia , Falência Hepática Aguda/cirurgia , Regeneração Hepática , Transplante de Fígado , Masculino , Metronidazol , Camundongos , Pessoa de Meia-Idade , Prognóstico , Regiões Promotoras Genéticas , Estudos Prospectivos , Protrombina/genética , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Proteína Smad4/genética , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta1/genética , Peixe-Zebra
4.
Diabetes ; 69(5): 1020-1031, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32139597

RESUMO

Progression from the initial vascular response upon hyperglycemia to a proliferative stage with neovacularizations is the hallmark of proliferative diabetic retinopathy. Here, we report on the novel diabetic pdx1 -/- zebrafish mutant as a model for diabetic retinopathy that lacks the transcription factor pdx1 through CRISPR-Cas9-mediated gene knockout leading to disturbed pancreatic development and hyperglycemia. Larval pdx1 -/- mutants prominently show vasodilation of blood vessels through increased vascular thickness in the hyaloid network as direct developmental precursor of the adult retinal vasculature in zebrafish. In adult pdx1 -/- mutants, impaired glucose homeostasis induces increased hyperbranching and hypersprouting with new vessel formation in the retina and aggravation of the vascular alterations from the larval to the adult stage. Both vascular aspects respond to antiangiogenic and antihyperglycemic pharmacological interventions in the larval stage and are accompanied by alterations in the nitric oxide metabolism. Thus, the pdx1 -/- mutant represents a novel model to study mechanisms of hyperglycemia-induced retinopathy wherein extensive proangiogenic alterations in blood vessel morphology and metabolic alterations underlie the vascular phenotype.


Assuntos
Proteínas de Homeodomínio/metabolismo , Hiperglicemia , Neovascularização Patológica , Vasos Retinianos/fisiologia , Transativadores/metabolismo , Animais , Glicemia , Sistemas CRISPR-Cas , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Larva , Óxido Nítrico/metabolismo , Ftalazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Neovascularização Retiniana , Transativadores/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra
5.
PLoS One ; 15(3): e0231153, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32210446

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0068033.].

6.
Sci Rep ; 5: 15007, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26458334

RESUMO

JUNB, a subunit of the AP-1 transcription factor complex, mediates gene regulation in response to a plethora of extracellular stimuli. Previously, JUNB was shown to act as a critical positive regulator of blood vessel development and homeostasis as well as a negative regulator of proliferation, inflammation and tumour growth. Here, we demonstrate that the oncogenic miR-182 is a novel JUNB target. Loss-of-function studies by morpholino-mediated knockdown and the CRISPR/Cas9 technology identify a novel function for both JUNB and its target miR-182 in lymphatic vascular development in zebrafish. Furthermore, we show that miR-182 attenuates foxo1 expression indicating that strictly balanced Foxo1 levels are required for proper lymphatic vascular development in zebrafish. In conclusion, our findings uncover with the Junb/miR-182/Foxo1 regulatory axis a novel key player in governing lymphatic vascular morphogenesis in zebrafish.


Assuntos
Regulação da Expressão Gênica , Linfangiogênese , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Animais , Expressão Ectópica do Gene , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Técnicas de Silenciamento de Genes , Inativação Gênica , Fenótipo , Proteínas Proto-Oncogênicas c-jun/genética , Ducto Torácico/embriologia , Ducto Torácico/metabolismo , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética
7.
Cell Mol Life Sci ; 72(14): 2709-18, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25894695

RESUMO

Blood vessels have been described a long time ago as passive circuits providing sufficient blood supply to ensure proper distribution of oxygen and nutrition. Blood vessels are mainly formed during embryonic development and in the early postnatal period. In the adult, blood vessels are quiescent, but can be activated and subsequently induced under pathophysiological conditions, such as ischemia and tumor growth. Surprisingly, recent data have suggested an active function for blood vessels, named angiocrine signaling, releasing trophogens which regulate organ development and organ regeneration including in the pancreas, lung, tumor cells, liver and bone. Lung development is driven by hypoxia as well as an intense endothelial-epithelial interaction, and important mechanisms contributing to these processes have recently been identified. This review aims to summarize recent developments and concepts about embryonic pulmonary vascular development and lung regeneration. We discuss hypoxia-inducible factor HIF-2α and vascular endothelial growth factor VEGF as important mediators in lung development and focus on endothelial-epithelial interactions and angiocrine signaling mechanisms.


Assuntos
Pulmão/fisiologia , Regeneração , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Humanos , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Neovascularização Fisiológica , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia
8.
Rejuvenation Res ; 18(4): 341-346, 2015 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-25706901

RESUMO

Dehydroepiandrosterone (DHEA) and dehydroepiandrosterone-sulphate (DHEAS) are the most abundant circulating adrenal steroid hormones. The plasma level of DHEAS correlates with longevity in primates and varies during human development with a maximum in early adulthood and a marked decline during aging. DHEA promotes the expression of molecular chaperones which are housekeeping stress response proteins essential for the processes of folding, translocation, maintenance and repair of proteins, RNA and DNA, as well as for homeostasis, immune response and cancer resistance. The level of chaperone expression correlates with longevity and shows a decline during aging. DHEA-induced promotion of chaperone expression could contribute to the epigenetic evolution of primate longevity.

9.
Diabetes ; 64(1): 213-25, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25092676

RESUMO

Hyperglycemia causes micro- and macrovascular complications in diabetic patients. Elevated glucose concentrations lead to increased formation of the highly reactive dicarbonyl methylglyoxal (MG), yet the early consequences of MG for development of vascular complications in vivo are poorly understood. In this study, zebrafish were used as a model organism to analyze early vascular effects and mechanisms of MG in vivo. High tissue glucose increased MG concentrations in tg(fli:EGFP) zebrafish embryos and rapidly induced several additional malformed and uncoordinated blood vessel structures that originated out of existing intersomitic blood vessels (ISVs). However, larger blood vessels, including the dorsal aorta and common cardinal vein, were not affected. Expression silencing of MG-degrading enzyme glyoxalase (glo) 1 elevated MG concentrations and induced a similar vascular hyperbranching phenotype in zebrafish. MG enhanced phosphorylation of vascular endothelial growth factor (VEGF) receptor 2 and its downstream target Akt/protein kinase B (PKB). Pharmacological inhibitors for VEGF receptor 2 and Akt/PKB as well as MG scavenger aminoguanidine and glo1 activation prevented MG-induced hyperbranching of ISVs. Taken together, MG acts on smaller blood vessels in zebrafish via the VEGF receptor signaling cascade, thereby describing a new mechanism that can explain vascular complications under hyperglycemia and elevated MG concentrations.


Assuntos
Vasos Sanguíneos/metabolismo , Glucose/metabolismo , Aldeído Pirúvico/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Vasos Sanguíneos/anormalidades , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Masculino , Neovascularização Fisiológica/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
10.
Invest Ophthalmol Vis Sci ; 55(11): 7637-51, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25425566

RESUMO

Corneal neovascularization (NV) refers to the growth of blood vessels and/or lymphatics into the physiologically avascular cornea, which occurs in several pathological processes. In mouse models, corneal NV can be artificially induced to investigate mechanisms of corneal pathologies. However, mouse models of corneal NV are not restricted to cornea-specific research, but also are widely used to investigate general mechanisms of angiogenesis. Because the cornea is transparent and easily accessible, corneal NV models are among the most useful in vivo models in angiogenesis research. The three different approaches that are used to study corneal NV in mice are based on direct application of proangiogenic or antiangiogenic transmitters, external injury to the cornea, or genetically engineered mice, which spontaneously develop corneal NV. The aim of this review is to compare the scope and limitations of the different approaches for corneal NV in mice. Our main focus is to highlight the potential of transgenic spontaneous models of corneal NV. Transgenic models do not require any experimental interference and make it possible to investigate different interconnected proangiogenic signaling cascades. As a result, transgenic models are highly useful for disease-centered angiogenesis research. In summary, transgenic models of corneal NV will complement and advance existing ocular NV assays, and help to discover new angiogenesis-related treatment strategies for ocular and extraocular diseases.


Assuntos
Córnea/patologia , Neovascularização da Córnea/genética , Regulação da Expressão Gênica , RNA/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Biomarcadores/metabolismo , Western Blotting , Córnea/metabolismo , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Modelos Animais de Doenças , Predisposição Genética para Doença , Camundongos , Camundongos Transgênicos , Fator A de Crescimento do Endotélio Vascular/biossíntese
11.
Arterioscler Thromb Vasc Biol ; 34(10): 2292-300, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25147336

RESUMO

OBJECTIVE: Nucleoside diphosphate kinase B (NDPKB) participates in the activation of heterotrimeric and monomeric G proteins, which are pivotal mediators in angiogenic signaling. The role of NDPKB in angiogenesis has to date not been defined. Therefore, we analyzed the contribution of NDPKB to angiogenesis and its underlying mechanisms in well-characterized in vivo and in vitro models. APPROACH AND RESULTS: Zebrafish embryos were depleted of NDPKB by morpholino-mediated knockdown. These larvae displayed severe malformations specifically in vessels formed by angiogenesis. NDPKB-deficient (NDPKB(-/-)) mice were subjected to oxygen-induced retinopathy. In this model, the number of preretinal neovascularizations in NDPKB(-/-) mice was strongly reduced in comparison with wild-type littermates. In accordance, a delayed blood flow recovery was detected in the NDPKB(-/-) mice after hindlimb ligation. In in vitro studies, a small interfering RNA-mediated knockdown of NDPKB was performed in human umbilical endothelial cells. NDPKB depletion impaired vascular endothelial growth factor (VEGF)-induced sprouting and hampered the VEGF-induced spatial redistributions of the VEGF receptor type 2 and VE-cadherin at the plasma membrane. Concomitantly, NDPKB depletion increased the permeability of the human umbilical endothelial cell monolayer. CONCLUSIONS: This is the first report to show that NDPKB is required for VEGF-induced angiogenesis and contributes to the correct localization of VEGF receptor type 2 and VE-cadherin at the endothelial adherens junctions. Therefore, our data identify NDPKB as a novel molecular target to modulate VEGF-dependent angiogenesis.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Células Endoteliais/enzimologia , Músculo Esquelético/irrigação sanguínea , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neovascularização Fisiológica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Membro Posterior , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Isquemia/enzimologia , Isquemia/genética , Isquemia/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nucleosídeo NM23 Difosfato Quinases/deficiência , Nucleosídeo NM23 Difosfato Quinases/genética , Interferência de RNA , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Neovascularização Retiniana/enzimologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/fisiopatologia , Transdução de Sinais , Fatores de Tempo , Transfecção , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
12.
Invest Ophthalmol Vis Sci ; 55(7): 4295-303, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24917145

RESUMO

PURPOSE: Corneal neovascularization can cause loss of vision. The introduction of anti-VEGF therapy has been a major improvement in therapeutic options. Recently, we established Kelch-like Ect2-interacting protein (KLEIP/KLHL20) knockout mice as a model of spontaneous corneal neovascular dystrophy. The aim of the present study was to characterize corneal neovascularization in progressive corneal dystrophy in KLEIP(-/-) mice, to evaluate the efficacy of anti-VEGF therapy, and to identify novel molecular regulators in this experimental model. METHODS: Corneal neovascularization was assessed by immunohistochemistry. Vascular endothelial growth factor signaling was inhibited by injection of a blocking antibody. Microarrays were used to measure expression of mRNA and microRNA (miRNA) in dystrophic corneae. Results were validated by immunohistochemistry and Western blotting. RESULTS: Blood vessels and lymphatics grew from the limbus toward the dystrophic epithelium in corneae of KLEIP(-/-) mice. Blocking VEGF signaling did not reduce phenotype progression. Correspondingly, microarray analysis revealed no upregulation of canonical vascular growth factors in late dystrophy. During phenotype progression, angiopoietin-1 expression increased while miR-204 expression decreased. Bioinformatic analysis identified a binding site for miR-204 in the angiopoietin-1 gene. Validation by in vitro experiments confirmed regulation of angiopoietin-1 by miR-204. CONCLUSIONS: Vascular endothelial growth factor does not act as a major player in corneal neovascularization in KLEIP(-/-) mice. However, the proangiogenic factor angiopoietin-1 was strongly upregulated in late-stage phenotype, correlating with loss of miR-204 expression. Correspondingly, we identified miR-204 as a novel regulator of angiopoietin-1 in vitro. These findings may explain the incomplete efficacy of anti-VEGF therapy in the clinic and may provide new candidates for pharmaceutical intervention.


Assuntos
Angiopoietina-1/genética , Neovascularização da Córnea/genética , Proteínas de Ligação a DNA/deficiência , Regulação da Expressão Gênica , MicroRNAs/genética , RNA Mensageiro/genética , Inibidores da Angiogênese/administração & dosagem , Angiopoietina-1/biossíntese , Animais , Western Blotting , Neovascularização da Córnea/tratamento farmacológico , Neovascularização da Córnea/metabolismo , Modelos Animais de Doenças , Imuno-Histoquímica , Injeções Intravítreas , Camundongos , Camundongos Knockout , MicroRNAs/biossíntese , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
13.
Basic Res Cardiol ; 109(2): 404, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24522833

RESUMO

ß1-Integrins are essential for angiogenesis. The mechanisms regulating integrin function in endothelial cells (EC) and their contribution to angiogenesis remain elusive. Brag2 is a guanine nucleotide exchange factor for the small Arf-GTPases Arf5 and Arf6. The role of Brag2 in EC and angiogenesis and the underlying molecular mechanisms remain unclear. siRNA-mediated Brag2-silencing reduced EC angiogenic sprouting and migration. Brag2-siRNA transfection differentially affected α5ß1- and αVß3-integrin function: specifically, Brag2-silencing increased focal/fibrillar adhesions and adhesion on ß1-integrin ligands (fibronectin and collagen), while reducing the adhesion on the αVß3-integrin ligand, vitronectin. Consistent with these results, Brag2-silencing enhanced surface expression of α5ß1-integrin, while reducing surface expression of αVß3-integrin. Mechanistically, Brag2-mediated αVß3-integrin-recycling and ß1-integrin endocytosis and specifically of the active/matrix-bound α5ß1-integrin present in fibrillar/focal adhesions (FA), suggesting that Brag2 contributes to the disassembly of FA via ß1-integrin endocytosis. Arf5 and Arf6 are promoting downstream of Brag2 angiogenic sprouting, ß1-integrin endocytosis and the regulation of FA. In vivo silencing of the Brag2-orthologues in zebrafish embryos using morpholinos perturbed vascular development. Furthermore, in vivo intravitreal injection of plasmids containing Brag2-shRNA reduced pathological ischemia-induced retinal and choroidal neovascularization. These data reveal that Brag2 is essential for developmental and pathological angiogenesis by promoting EC sprouting through regulation of adhesion by mediating ß1-integrin internalization and link for the first time the process of ß1-integrin endocytosis with angiogenesis.


Assuntos
Adesão Celular/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Neovascularização Patológica/fisiopatologia , Retinopatia da Prematuridade/fisiopatologia , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/metabolismo , Animais , Animais Geneticamente Modificados , Células COS , Movimento Celular/fisiologia , Chlorocebus aethiops , Neovascularização de Coroide/genética , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/fisiopatologia , Modelos Animais de Doenças , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Camundongos Endogâmicos C57BL , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica/genética , Neovascularização Fisiológica/fisiologia , RNA Interferente Pequeno/genética , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra
14.
Rejuvenation Res ; 17(2): 157-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23992378

RESUMO

Evidence is given that replicative senescence--possibly as organismal aging--constitutes epigenetic phenomena, counteracted by homeostatic factors such as, e.g., the molecular chaperones, which are housekeeping molecules essential for the folding, repair, and transport of proteins, RNA, and DNA. Weakening of the chaperone defense with age probably contributes to the frailty in senescence. The present review presents evidence that the human fetal estrogen hormone estetrol, by promotion of chaperone functions, homeostasis, and cancer resistance, may prove useful as a supplement during human senescence.


Assuntos
Resistência à Doença/genética , Epigênese Genética/efeitos dos fármacos , Estetrol/farmacologia , Longevidade/genética , Chaperonas Moleculares/metabolismo , Neoplasias/genética , Humanos , Longevidade/efeitos dos fármacos
15.
PLoS One ; 8(7): e68033, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874489

RESUMO

Protein kinase D isoenzymes (PKDs, Prkds) are serine threonine kinases that belong to the CAMK superfamily. PKD1 is expressed in endothelial cells and is a major mediator of biological responses downstream of the VEGFRs that are relevant for angiogenesis such as endothelial cell migration, proliferation and tubulogenesis in vitro. PKDs also play a critical role in tumor development and progression, including tumor angiogenesis. However, given the plethora of signaling modules that drive angiogenesis, the precise role of PKD1 in both physiological and tumor angiogenesis in vivo has not been worked out so far. This study aimed at dissecting the contribution of PKD1 to physiological blood vessel formation, PKD1 was found to be widely expressed during zebrafish development. As far as physiological angiogenesis was concerned, morpholino-based silencing of PKD1 expression moderately reduced the formation of the intersomitic vessels and the dorsal longitudinal anastomotic vessel in tg(fli1:EGFP) zebrafish. In addition, silencing of PKD1 resulted in reduced formation of the parachordal lymphangioblasts that serves as a precursor for the developing thoracic duct. Interestingly, tumor angiogenesis was completely abolished in PKD1 morphants using the zebrafish/tumor xenograft angiogenesis assay. Our data in zebrafish demonstrate that PKD1 contributes to the regulation of physiological angiogenesis and lymphangiogenesis during zebrafish development and is essential for tumor angiogenesis.


Assuntos
Neoplasias/genética , Neovascularização Patológica/genética , Neovascularização Fisiológica/genética , Proteína Quinase C/genética , Peixe-Zebra/genética , Animais , Linhagem Celular Tumoral , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Humanos , Linfangiogênese/genética , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Proteína Quinase C/metabolismo , Peixe-Zebra/metabolismo
16.
Circ Res ; 111(11): 1421-33, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22955733

RESUMO

RATIONALE: Formation and remodeling of the vasculature during development and disease involve a highly conserved and precisely regulated network of attractants and repellants. Various signaling pathways control the behavior of endothelial cells, but their posttranscriptional dose titration by microRNAs is poorly understood. OBJECTIVE: To identify microRNAs that regulate angiogenesis. METHODS AND RESULTS: We show that the highly conserved microRNA family encoding miR-10 regulates the behavior of endothelial cells during angiogenesis by positively titrating proangiogenic signaling. Knockdown of miR-10 led to premature truncation of intersegmental vessel growth in the trunk of zebrafish larvae, whereas overexpression of miR-10 promoted angiogenic behavior in zebrafish and cultured human umbilical venous endothelial cells. We found that miR-10 functions, in part, by directly regulating the level of fms-related tyrosine kinase 1 (FLT1), a cell-surface protein that sequesters vascular endothelial growth factor, and its soluble splice variant sFLT1. The increase in FLT1/sFLT1 protein levels upon miR-10 knockdown in zebrafish and in human umbilical venous endothelial cells inhibited the angiogenic behavior of endothelial cells largely by antagonizing vascular endothelial growth factor receptor 2 signaling. CONCLUSIONS: Our study provides insights into how FLT1 and vascular endothelial growth factor receptor 2 signaling is titrated in a microRNA-mediated manner and establishes miR-10 as a potential new target for the selective modulation of angiogenesis.


Assuntos
Células Endoteliais/metabolismo , MicroRNAs/genética , Neovascularização Fisiológica/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Animais Geneticamente Modificados , Sequência de Bases , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Immunoblotting , Larva/genética , Larva/metabolismo , Masculino , Camundongos , Microscopia Confocal , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Transdução de Sinais/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra
17.
Trends Cardiovasc Med ; 22(1): 7-11, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22841837

RESUMO

The members of the HOX transcription factor family are important basic regulators of morphogenesis and development and several HOX proteins have also been identified as essential regulators of physiological and pathologic angiogenesis. HOXC9 is highly expressed in quiescent endothelial cells and keeps the vasculature in a resting state via inhibition of interleukin-8 production. HOXC9 overexpression in zebra-fish negatively regulated vascular development which can be rescued by exogenous interleukin-8. The further understanding of the HOXC9-IL-8 signaling axis and the identification of other HOXC9 targets in the vasculature will provide important insights into mechanisms promoting endothelial cell activation during physiological angiogenesis. It will also be beneficial to understand pathophysiological angiogenesis regulation and thus provide important new directions for the development of novel anti-angiogenic therapeutic strategies.


Assuntos
Endotélio Vascular/patologia , Proteínas de Homeodomínio/genética , Animais , Modelos Animais de Doenças , Expressão Gênica/genética , Proteínas de Homeodomínio/biossíntese , Humanos , Interleucina-8/genética , Neovascularização Fisiológica , Peixe-Zebra
18.
Rejuvenation Res ; 15(2): 210-2, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22533434

RESUMO

Bile acids are detergent molecules derived from cholesterol in the liver that are important for the metabolism and absorption of lipids in the intestine. Bile acids are also steroid hormones activating specific nuclear receptors and G protein-coupled receptors. Conjugated bile acids are cytoprotective and anticarcinogenic. Bile acid synthesis and bile flow decreases markedly during aging. The housekeeping molecular chaperones are stress response proteins, important for the processes of folding, maintenance, and repair of proteins, RNA, and DNA, as well as for the structure and function of the steroid hormone receptors. The level of expression of the molecular chaperones correlates with mammalian longevity as well as with the life span of differentiated cells. The functions of the chaperone machinery are progressively impaired during aging, and the progressive age-related impairment of these housekeeping mechanisms probably contributes to the phenotype of aging. This review presents evidence that the bile acids are chemical chaperones, improving the general chaperone defense, and thus serve to support an epigenetic mechanism of possible significance for the evolution of mammalian longevity, as well as for the attainment of healthy aging.


Assuntos
Ácidos e Sais Biliares/metabolismo , Longevidade , Chaperonas Moleculares/metabolismo , Envelhecimento , Animais , Ácidos e Sais Biliares/química , Diferenciação Celular , DNA/química , DNA/metabolismo , Detergentes/farmacologia , Humanos , Ligantes , Dobramento de Proteína , RNA/química , RNA/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo
19.
Invest Ophthalmol Vis Sci ; 53(6): 3260-8, 2012 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-22511632

RESUMO

PURPOSE: The BTB-kelch protein KLEIP/KLHL20 is an actin binding protein that regulates cell-cell contact formation and cell migration. The aim of our study was to characterize KLEIP's function in ocular health and disease in mice. METHODS: KLEIP(-/-) mice were generated, and corneas were examined histologically and stained for keratin-1, loricrin, keratin-12, keratin-14, CD31, LYVE-1, F4/80, E-cadherin, and Ki67. Corneal abrasions were performed after eyelid opening. RESULTS: Corneas of KLEIP(+/+) and KLEIP(-/-) mice were indistinguishable at birth. After eyelid opening corneal epithelial hyperplasia started to manifest in KLEIP(-/-) mice, showing a progressive epithelial metaplasia leading to total corneal opacity. In KLEIP(-/-) mice the initial stratified squamous corneal epithelium was altered to an epidermal histo-architecture showing several superficial keratinized cells, cell infiltrations into the stroma, and several apoptotic cells. Skin markers keratin 1 and loricrin were positive, and surface disease was accompanied by deep stromal vascularization. Expression analysis for E-cadherin in KLEIP(-/-) corneas showed acellular areas in the squamous epithelium, indicating a progressive fragile corneal integrity. Removal of the virgin epithelium accelerated strongly development of the epithelial and stromal alterations, identifying mechanical injuries as the major trigger for corneal dystrophy formation and scarification in KLEIP(-/-) mice. CONCLUSIONS: The data identify KLEIP as an important molecule regulating corneal epithelial integrity.


Assuntos
Proteínas de Transporte/genética , Córnea/metabolismo , Neovascularização da Córnea/complicações , Opacidade da Córnea/patologia , Regulação da Expressão Gênica , RNA Mensageiro/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/biossíntese , Córnea/patologia , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Opacidade da Córnea/etiologia , Opacidade da Córnea/genética , Modelos Animais de Doenças , Progressão da Doença , Genótipo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real
20.
Cell Signal ; 24(6): 1261-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22374305

RESUMO

Vascular endothelial growth factor (VEGF) is a main stimulator of pathological vessel formation. Nevertheless, increasing evidence suggests that Angiotensin II (Ang II) can play an augmentory role in this process. We thus analyzed the contribution of the two Ang II receptor types, AT(1)R and AT(2)R, in a mouse model of VEGF-driven angiogenesis, i.e. oxygen-induced proliferative retinopathy. Application of the AT(1)R antagonist telmisartan but not the AT(2)R antagonist PD123,319 largely attenuated the pathological response. A direct effect of Ang II on endothelial cells (EC) was analyzed by assessing angiogenic responses in primary bovine retinal and immortalized rat microvascular EC. Selective stimulation of the AT(1)R by Ang II in the presence of PD123,319 revealed a pro-angiogenic activity which further increased VEGF-driven EC sprouting and migration. In contrast, selective stimulation of the AT(2)R by either CGP42112A or Ang II in the presence of telmisartan inhibited the VEGF-driven angiogenic response. Using specific inhibitors (pertussis toxin, RGS proteins, kinase inhibitors) we identified G(12/13) and G(i) dependent signaling pathways as the mediators of the AT(1)R-induced angiogenesis and the AT(2)R-induced inhibition, respectively. As AT(1)R and AT(2)R stimulation displays opposing effects on the activity of the monomeric GTPase RhoA and pro-angiogenic responses to Ang II and VEGF requires activation of Rho-dependent kinase (ROCK), we conclude that the opposing effects of the Ang II receptors on VEGF-driven angiogenesis converge on the regulation of activity of RhoA-ROCK-dependent EC migration.


Assuntos
Angiotensina II/metabolismo , Neovascularização Fisiológica , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Retina/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Antagonistas de Receptores de Angiotensina/farmacologia , Animais , Bovinos , Movimento Celular , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/citologia , Microvasos/crescimento & desenvolvimento , Microvasos/metabolismo , Neovascularização Patológica , Ratos , Retina/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA