Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 296: 100541, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33722606

RESUMO

Vascular smooth muscle cells (VSMCs) contribute to the deposition of extracellular matrix proteins (ECMs), including Type IV collagen, in the vessel wall. ECMs coordinate communication among different cell types, but mechanisms underlying this communication remain unclear. Our previous studies have demonstrated that X-box binding protein 1 (XBP1) is activated and contributes to VSMC phenotypic transition in response to vascular injury. In this study, we investigated the participation of XBP1 in the communication between VSMCs and vascular progenitor cells (VPCs). Immunofluorescence and immunohistology staining revealed that Xbp1 gene was essential for type IV collagen alpha 1 (COL4A1) expression during mouse embryonic development and vessel wall ECM deposition and stem cell antigen 1-positive (Sca1+)-VPC recruitment in response to vascular injury. The Western blot analysis elucidated an Xbp1 gene dose-dependent effect on COL4A1 expression and that the spliced XBP1 protein (XBP1s) increased protease-mediated COL4A1 degradation as revealed by Zymography. RT-PCR analysis revealed that XBP1s in VSMCs not only upregulated COL4A1/2 transcription but also induced the occurrence of a novel transcript variant, soluble type IV collagen alpha 1 (COL4A1s), in which the front part of exon 4 is joined with the rear part of exon 42. Chromatin-immunoprecipitation, DNA/protein pulldown and in vitro transcription demonstrated that XBP1s binds to exon 4 and exon 42, directing the transcription from exon 4 to exon 42. This leads to transcription complex bypassing the internal sequences, producing a shortened COL4A1s protein that increased Sca1+-VPC migration. Taken together, these results suggest that activated VSMCs may recruit Sca1+-VPCs via XBP1s-mediated COL4A1s secretion, leading to vascular injury repair or neointima formation.


Assuntos
Comunicação Celular , Movimento Celular , Colágeno Tipo IV/metabolismo , Músculo Liso Vascular/fisiologia , Células-Tronco/fisiologia , Proteína 1 de Ligação a X-Box/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Colágeno Tipo IV/genética , Humanos , Camundongos , Músculo Liso Vascular/citologia , Transdução de Sinais , Células-Tronco/citologia , Proteína 1 de Ligação a X-Box/genética
2.
Cancer Sci ; 112(5): 1839-1852, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33205567

RESUMO

Angiogenesis is closely associated with tumorigenesis, invasion, and metastasis by providing oxygen and nutrients. Recently, increasing evidence indicates that cancer-derived exosomes which contain proteins, coding, and noncoding RNAs (ncRNAs) were shown to have proangiogenic function in cancer. A 26-nt-long ncRNA (X26nt) is generated in the process of inositol-requiring enzyme 1 alpha (IRE1α)-induced unspliced XBP1 splicing. However, the role of X26nt in the angiogenesis of gastric cancer (GC) remains largely unknown. In the present study, we found that X26nt was significantly elevated in GC and GC exosomes. Then, we verified that X26nt could be delivered into human umbilical vein endothelial cells (HUVECs) via GC cell exosomes and promote the proliferation, migration, and tube formation of HUVECs. We revealed that exosomal X26nt decreased vascular endothelial cadherin (VE-cadherin) by directly combining the 3'UTR of VE-cadherin mRNA in HUVECs, thereby increasing vascular permeability. We further demonstrated that X26nt accelerates the tumor growth and angiogenesis in a mouse subcutaneous tumor model. Our findings investigate a unique intercellular communication mediated by cancer-derived exosomes and reveal a novel mechanism of exosomal X26nt in the regulation of tumor vasculature.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar , Exossomos/metabolismo , Neovascularização Patológica/etiologia , RNA Longo não Codificante/metabolismo , Neoplasias Gástricas/irrigação sanguínea , Regiões 3' não Traduzidas , Animais , Antígenos CD/genética , Caderinas/genética , Comunicação Celular , Movimento Celular , Proliferação de Células , Endorribonucleases/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Serina-Treonina Quinases/metabolismo , Processamento de Proteína , RNA Mensageiro/metabolismo , Neoplasias Gástricas/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo
3.
J Cell Sci ; 132(16)2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31331967

RESUMO

Dysfunction of endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) leads to ischaemia, the central pathology of cardiovascular disease. Stem cell technology will revolutionise regenerative medicine, but a need remains to understand key mechanisms of vascular differentiation. RNA-binding proteins have emerged as novel post-transcriptional regulators of alternative splicing and we have previously shown that the RNA-binding protein Quaking (QKI) plays roles in EC differentiation. In this study, we decipher the role of the alternative splicing isoform Quaking 6 (QKI-6) to induce VSMC differentiation from induced pluripotent stem cells (iPSCs). PDGF-BB stimulation induced QKI-6, which bound to HDAC7 intron 1 via the QKI-binding motif, promoting HDAC7 splicing and iPS-VSMC differentiation. Overexpression of QKI-6 transcriptionally activated SM22 (also known as TAGLN), while QKI-6 knockdown diminished differentiation capability. VSMCs overexpressing QKI-6 demonstrated greater contractile ability, and upon combination with iPS-ECs-overexpressing the alternative splicing isoform Quaking 5 (QKI-5), exhibited higher angiogenic potential in vivo than control cells alone. This study demonstrates that QKI-6 is critical for modulation of HDAC7 splicing, regulating phenotypically and functionally robust iPS-VSMCs. These findings also highlight that the QKI isoforms hold key roles in alternative splicing, giving rise to cells which can be used in vascular therapy or for disease modelling.This article has an associated First Person interview with the first author of the paper.


Assuntos
Processamento Alternativo , Células Endoteliais/metabolismo , Modelos Cardiovasculares , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Linhagem Celular , Células Endoteliais/patologia , Células HEK293 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Isquemia/genética , Isquemia/metabolismo , Isquemia/patologia , Isquemia/terapia , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Proteínas de Ligação a RNA/genética
4.
Stem Cells ; 38(4): 556-573, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31721359

RESUMO

Histone deacetylase 7 (HDAC7) plays a pivotal role in the maintenance of the endothelium integrity. In this study, we demonstrated that the intron-containing Hdac7 mRNA existed in the cytosol and that ribosomes bound to a short open reading frame (sORF) within the 5'-terminal noncoding area of this Hdac7 mRNA in response to vascular endothelial growth factor (VEGF) stimulation in the isolated stem cell antigen-1 positive (Sca1+ ) vascular progenitor cells (VPCs). A 7-amino acid (7A) peptide has been demonstrated to be translated from the sORF in Sca1+ -VPCs in vitro and in vivo. The 7A peptide was shown to receive phosphate group from the activated mitogen-activated protein kinase MEKK1 and transfer it to 14-3-3 gamma protein, forming an MEKK1-7A-14-3-3γ signal pathway downstream VEGF. The exogenous synthetic 7A peptide could increase Sca1+ -VPCs cell migration, re-endothelialization in the femoral artery injury, and angiogenesis in hind limb ischemia. A Hd7-7sFLAG transgenic mice line was generated as the loss-of-function model, in which the 7A peptide was replaced by a FLAG-tagged scrabbled peptide. Loss of the endogenous 7A impaired Sca1+ -VPCs cell migration, re-endothelialization of the injured femoral artery, and angiogenesis in ischemic tissues, which could be partially rescued by the addition of the exogenous 7A/7Ap peptide. This study provides evidence that sORFs can be alternatively translated and the derived peptides may play an important role in physiological processes including vascular remodeling.


Assuntos
Histona Desacetilases/metabolismo , Neovascularização Fisiológica/genética , Animais , Proliferação de Células , Humanos , Masculino , Camundongos , Fosforilação , Transdução de Sinais
5.
Stem Cells ; 37(2): 226-239, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30372556

RESUMO

The mortality rate for (cardio)-vascular disease is one of the highest in the world, so a healthy functional endothelium is of outmost importance against vascular disease. In this study, human induced pluripotent stem (iPS) cells were reprogrammed from 1 ml blood of healthy donors and subsequently differentiated into endothelial cells (iPS-ECs) with typical EC characteristics. This research combined iPS cell technologies and next-generation sequencing to acquire an insight into the transcriptional regulation of iPS-ECs. We identified endothelial cell-specific molecule 1 (ESM1) as one of the highest expressed genes during EC differentiation, playing a key role in EC enrichment and function by regulating connexin 40 (CX40) and eNOS. Importantly, ESM1 enhanced the iPS-ECs potential to improve angiogenesis and neovascularisation in in vivo models of angiogenesis and hind limb ischemia. These findings demonstrated for the first time that enriched functional ECs are derived through cell reprogramming and ESM1 signaling, opening the horizon for drug screening and cell-based therapies for vascular diseases. Therefore, this study showcases a new approach for enriching and enhancing the function of induced pluripotent stem (iPS) cell-derived ECs from a very small amount of blood through ESM1 signaling, which greatly enhances their functionality and increases their therapeutic potential. Stem Cells 2019;37:226-239.


Assuntos
Células Endoteliais/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas de Neoplasias/metabolismo , Proteoglicanas/metabolismo , Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Células Endoteliais/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Proteínas de Neoplasias/genética , Proteoglicanas/genética , Transdução de Sinais
6.
Stem Cells ; 36(7): 1033-1044, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29569797

RESUMO

The fight against vascular disease requires functional endothelial cells (ECs) which could be provided by differentiation of induced Pluripotent Stem Cells (iPS Cells) in great numbers for use in the clinic. However, the great promise of the generated ECs (iPS-ECs) in therapy is often restricted due to the challenge in iPS-ECs preserving their phenotype and function. We identified that Follistatin-Like 3 (FSTL3) is highly expressed in iPS-ECs, and, as such, we sought to clarify its possible role in retaining and improving iPS-ECs function and phenotype, which are crucial in increasing the cells' potential as a therapeutic tool. We overexpressed FSTL3 in iPS-ECs and found that FSTL3 could induce and enhance endothelial features by facilitating ß-catenin nuclear translocation through inhibition of glycogen synthase kinase-3ß activity and induction of Endothelin-1. The angiogenic potential of FSTL3 was also confirmed both in vitro and in vivo. When iPS-ECs overexpressing FSTL3 were subcutaneously injected in in vivo angiogenic model or intramuscularly injected in a hind limb ischemia NOD.CB17-Prkdcscid/NcrCrl SCID mice model, FSTL3 significantly induced angiogenesis and blood flow recovery, respectively. This study, for the first time, demonstrates that FSTL3 can greatly enhance the function and maturity of iPS-ECs. It advances our understanding of iPS-ECs and identifies a novel pathway that can be applied in cell therapy. These findings could therefore help improve efficiency and generation of therapeutically relevant numbers of ECs for use in patient-specific cell-based therapies. In addition, it can be particularly useful toward the treatment of vascular diseases instigated by EC dysfunction. Stem Cells 2018;36:1033-1044.


Assuntos
Reprogramação Celular/genética , Proteínas Relacionadas à Folistatina/genética , Quinases da Glicogênio Sintase/antagonistas & inibidores , Células-Tronco Pluripotentes Induzidas/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular , Proteínas Relacionadas à Folistatina/metabolismo , Quinases da Glicogênio Sintase/metabolismo , Humanos , Camundongos
7.
Circ Res ; 121(12): 1331-1345, 2017 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-29089350

RESUMO

RATIONALE: Although not fully understood, the phenotypic transition of vascular smooth muscle cells exhibits at the early onset of the pathology of aortic aneurysms. Exploring the key regulators that are responsible for maintaining the contractile phenotype of vascular smooth muscle cells (VSMCs) may confer vascular homeostasis and prevent aneurysmal disease. XBP1 (X-box binding protein 1), which exists in a transcriptionally inactive unspliced form (XBP1u) and a spliced active form (XBP1s), is a key component in response to endoplasmic reticular stress. Compared with XBP1s, little is known about the role of XBP1u in vascular homeostasis and disease. OBJECTIVE: We aim to investigate the role of XBP1u in VSMC phenotypic switching and the pathogenesis of aortic aneurysms. METHODS AND RESULTS: XBP1u, but not XBP1s, was markedly repressed in the aorta during the early onset of aortic aneurysm in both angiotensin II-infused apolipoprotein E knockout (ApoE-/-) and CaPO4 (calcium phosphate)-induced C57BL/6J murine models, in parallel with a decrease in smooth muscle cell contractile apparatus proteins. In vivo studies revealed that XBP1 deficiency in smooth muscle cells caused VSMC dedifferentiation, enhanced vascular inflammation and proteolytic activity, and significantly aggravated both thoracic and abdominal aortic aneurysms in mice. XBP1 deficiency, but not an inhibition of XBP1 splicing, induced VSMC switching from the contractile phenotype to a proinflammatory and proteolytic phenotype. Mechanically, in the cytoplasm, XBP1u directly associated with the N terminus of FoxO4 (Forkhead box protein O 4), a recognized repressor of VSMC differentiation via the interaction and inhibition of myocardin. Blocking the XBP1u-FoxO4 interaction facilitated nuclear translocation of FoxO4, repressed smooth muscle cell marker genes expression, promoted proinflammatory and proteolytic phenotypic transitioning in vitro, and stimulated aortic aneurysm formation in vivo. CONCLUSIONS: Our study revealed the pivotal role of the XBP1u-FoxO4-myocardin axis in maintaining the VSMC contractile phenotype and providing protection from aortic aneurysm formation.


Assuntos
Aneurisma Aórtico/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Splicing de RNA , Proteína 1 de Ligação a X-Box/metabolismo , Animais , Aneurisma Aórtico/genética , Sítios de Ligação , Células COS , Proteínas de Ciclo Celular , Chlorocebus aethiops , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/genética , Células HEK293 , Homeostase , Humanos , Masculino , Camundongos , Músculo Liso Vascular/patologia , Mutação , Proteínas Nucleares/metabolismo , Ligação Proteica , Transativadores/metabolismo , Proteína 1 de Ligação a X-Box/genética
8.
Exp Cell Res ; 363(2): 262-270, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29352987

RESUMO

The X-box binding protein 1 (XBP1) is a pivotal transcription factor in the endoplasmic reticulum stress response. Our previous studies have proven that XBP1 is involved in vascular endothelial growth factor (VEGF)-mediated endothelial cell (EC) proliferation and angiogenesis. In this study, we used EC monolayer wound healing, tube formation and transwell migration models to explore the role of XBP1splicing in EC migration. We found that scratching on EC monolayer triggered XBP1splicing, which was attenuated by the presence of SU5416and LY294002, suggesting that VEGF signalling pathways may be involved. Over-expression of the spliced XBP1 (XBP1s) via Ad-XBP1s gene transfer increased while knockdown of IRE1αor XBP1 by ShRNA lentivirus suppressed EC migration. Over-expression of XBP1s up-regulated the nitric oxide synthase 3 (NOS3)mRNA through the 3'UTR-mediated stabilisation and increased eNOS protein translation. Further experiments demonstrated that miR-24 participated in the XBP1s-induced eNOSup-regulation and EC migration. Further co-IP and immunofluorescence staining assays revealed that protein kinase B (Akt), eNOS andXBP1s form a complex, resulting in Akt and eNOS nucleus relocation. These results suggest that XBP1 splicing can regulate eNOS expression and cellular location, leading to EC migration and therefore contributing to wound healing and angiogenesis.


Assuntos
Movimento Celular/fisiologia , Células Endoteliais/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Proliferação de Células/fisiologia , Células Endoteliais/citologia , Endotélio Vascular/metabolismo , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
J Mol Cell Cardiol ; 122: 98-113, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30098321

RESUMO

Atherosclerosis, a chronic inflammatory condition that is characterized by the accumulation of lipid-loaded macrophages, occurs preferentially at the arterial branching points where disturbed flow is prominent. The pathogenesis of atherosclerotic lesion formation is a multistage process involving multiple cell types, inflammatory mediators and hemodynamic forces in the vessel wall in response to atherogenic stimuli. Researches from the past decade have uncovered the critical roles of microRNAs (miRNAs) in regulating multiple pathophysiological effects and signaling pathways in endothelial cells (ECs), vascular smooth muscle cells (VSMCs), macrophages and lipid homeostasis, which are key in atherosclerotic lesion formation. The expression of these miRNAs are either in response to biomechanical (flow-responsive) or biochemical (non-flow-responsive) stimuli. Recent evidences also indicate an important role for long non-coding RNAs (lncRNAs) in mediating several atherosclerotic processes. In this review, we provide a detailed summary on the current paradigms in miRNA-dependent regulation, the emerging role of lncRNAs in the initiation and progression of atherosclerosis, and clinical interventions targeting these in an attempt to develop novel diagnostics and treatments for atherosclerosis.


Assuntos
Aterosclerose/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Biomarcadores/metabolismo , Colesterol/metabolismo , Células Endoteliais/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica/patologia , Ratos
10.
Stem Cells ; 35(4): 952-966, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28207177

RESUMO

The capability to derive endothelial cell (ECs) from induced pluripotent stem cells (iPSCs) holds huge therapeutic potential for cardiovascular disease. This study elucidates the precise role of the RNA-binding protein Quaking isoform 5 (QKI-5) during EC differentiation from both mouse and human iPSCs (hiPSCs) and dissects how RNA-binding proteins can improve differentiation efficiency toward cell therapy for important vascular diseases. iPSCs represent an attractive cellular approach for regenerative medicine today as they can be used to generate patient-specific therapeutic cells toward autologous cell therapy. In this study, using the model of iPSCs differentiation toward ECs, the QKI-5 was found to be an important regulator of STAT3 stabilization and vascular endothelial growth factor receptor 2 (VEGFR2) activation during the EC differentiation process. QKI-5 was induced during EC differentiation, resulting in stabilization of STAT3 expression and modulation of VEGFR2 transcriptional activation as well as VEGF secretion through direct binding to the 3' UTR of STAT3. Importantly, mouse iPS-ECs overexpressing QKI-5 significantly improved angiogenesis and neovascularization and blood flow recovery in experimental hind limb ischemia. Notably, hiPSCs overexpressing QKI-5, induced angiogenesis on Matrigel plug assays in vivo only 7 days after subcutaneous injection in SCID mice. These results highlight a clear functional benefit of QKI-5 in neovascularization, blood flow recovery, and angiogenesis. Thus, they provide support to the growing consensus that elucidation of the molecular mechanisms underlying EC differentiation will ultimately advance stem cell regenerative therapy and eventually make the treatment of cardiovascular disease a reality. The RNA binding protein QKI-5 is induced during EC differentiation from iPSCs. RNA binding protein QKI-5 was induced during EC differentiation in parallel with the EC marker CD144. Immunofluorescence staining showing that QKI-5 is localized in the nucleus and stained in parallel with CD144 in differentiated ECs (scale bar = 50 µm). Stem Cells 2017 Stem Cells 2017;35:952-966.


Assuntos
Diferenciação Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Neovascularização Fisiológica , Proteínas de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Antígenos CD , Caderinas , Modelos Animais de Doenças , Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Isquemia/patologia , Camundongos Endogâmicos C57BL , Ligação Proteica , Fluxo Sanguíneo Regional , Fator de Transcrição STAT3/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Stem Cells ; 34(9): 2368-80, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27300479

RESUMO

Recent studies have shown that Sca-1(+) (stem cell antigen-1) stem/progenitor cells within blood vessel walls may contribute to neointima formation, but the mechanism behind their recruitment has not been explored. In this work Sca-1(+) progenitor cells were cultivated from mouse vein graft tissue and found to exhibit increased migration when cocultured with smooth muscle cells (SMCs) or when treated with SMC-derived conditioned medium. This migration was associated with elevated levels of chemokines, CCL2 (chemokine (C-C motif) ligand 2) and CXCL1 (chemokine (C-X-C motif) ligand 1), and their corresponding receptors on Sca-1(+) progenitors, CCR2 (chemokine (C-C motif) receptor 2) and CXCR2 (chemokine (C-X-C motif) receptor 2), which were also upregulated following SMC conditioned medium treatment. Knockdown of either receptor in Sca-1(+) progenitors significantly inhibited cell migration. The GTPases Cdc42 and Rac1 were activated by both CCL2 and CXCL1 stimulation and p38 phosphorylation was increased. However, only Rac1 inhibition significantly reduced migration and p38 phosphorylation. After Sca-1(+) progenitors labeled with green fluorescent protein (GFP) were applied to the adventitial side of wire-injured mouse femoral arteries, a large proportion of GFP-Sca-1(+) -cells were observed in neointimal lesions, and a marked increase in neointimal lesion formation was seen 1 week post-operation. Interestingly, Sca-1(+) progenitor migration from the adventitia to the neointima was abrogated and neointima formation diminished in a wire injury model using CCL2(-/-) mice. These findings suggest vascular stem/progenitor cell migration from the adventitia to the neointima can be induced by SMC release of chemokines which act via CCR2/Rac1/p38 and CXCR2/Rac1/p38 signaling pathways. Stem Cells 2016;34:2368-2380.


Assuntos
Movimento Celular , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Miócitos de Músculo Liso/metabolismo , Neointima/patologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Antígenos Ly/metabolismo , Movimento Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Receptores CCR2 , Receptores de Interleucina-8B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 35(10): 2134-44, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26315405

RESUMO

OBJECTIVE: Smooth muscle cell (SMC) migration and proliferation play an essential role in neointimal formation after vascular injury. In this study, we intended to investigate whether the X-box-binding protein 1 (XBP1) was involved in these processes. APPROACH AND RESULTS: In vivo studies on femoral artery injury models revealed that vascular injury triggered an immediate upregulation of XBP1 expression and splicing in vascular SMCs and that XBP1 deficiency in SMCs significantly abrogated neointimal formation in the injured vessels. In vitro studies indicated that platelet-derived growth factor-BB triggered XBP1 splicing in SMCs via the interaction between platelet-derived growth factor receptor ß and the inositol-requiring enzyme 1α. The spliced XBP1 (XBP1s) increased SMC migration via PI3K/Akt activation and proliferation via downregulating calponin h1 (CNN1). XBP1s directed the transcription of mir-1274B that targeted CNN1 mRNA degradation. Proteomic analysis of culture media revealed that XBP1s decreased transforming growth factor (TGF)-ß family proteins secretion via transcriptional suppression. TGF-ß3 but not TGF-ß1 or TGF-ß2 attenuated XBP1s-induced CNN1 decrease and SMC proliferation. CONCLUSIONS: This study demonstrates for the first time that XBP1 is crucial for SMC proliferation via modulating the platelet-derived growth factor/TGF-ß pathways, leading to neointimal formation.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Neointima/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Transcrição/genética , Remodelação Vascular/genética , Lesões do Sistema Vascular/genética , Animais , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Artéria Femoral/lesões , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , RNA Mensageiro/metabolismo , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real/métodos , Receptor Cross-Talk , Fatores de Transcrição de Fator Regulador X , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/metabolismo , Remodelação Vascular/fisiologia , Lesões do Sistema Vascular/fisiopatologia , Proteína 1 de Ligação a X-Box
13.
Curr Opin Lipidol ; 26(5): 449-56, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26218417

RESUMO

PURPOSE OF REVIEW: Physiologically, endothelial integrity and smooth muscle homeostasis play key roles in the maintenance of vascular structure and functions. Under pathological conditions, endothelial and smooth muscle cells display great plasticity by transdifferentiating into other cell phenotypes. This review aims to update the progress in endothelial and smooth muscle cell transformation and to discuss their underlying mechanisms. RECENT FINDINGS: At the early stage of atherosclerosis, it was traditionally believed that smooth muscle cells from the media migrate into the intima in which they proliferate to form neointimal lesions. Recently, endothelial cells were shown to undergo transformation to form smooth muscle-like cells that contribute to neointimal formation. Furthermore, not only can medial smooth muscle cells migrate and proliferate, they also have the ability to differentiate into macrophages in the intima in which they form foam cells by uptaking lipids. Finally, the discovery of stem/progenitor cells in the vessel wall that can differentiate into all types of vascular cells has complicated the research field even further. SUMMARY: Based on the current progress in the research field, it is worthy to explore the contributions of cell transformation to the pathogenesis of atherosclerosis to understand the mechanisms on how they are regulated in order to develop novel therapeutic application targeting these processes to reverse the disease progression.


Assuntos
Aterosclerose/patologia , Células Endoteliais/fisiologia , Miócitos de Músculo Liso/fisiologia , Animais , Transdiferenciação Celular , Endotélio Vascular/patologia , Humanos , Músculo Liso Vascular/patologia
14.
J Biol Chem ; 289(44): 30625-30634, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25190803

RESUMO

It is well known that atherosclerosis occurs geographically at branch points where disturbed flow predisposes to the development of plaque via triggering of oxidative stress and inflammatory reactions. In this study, we found that disturbed flow activated anti-oxidative reactions via up-regulating heme oxygenase 1 (HO-1) in an X-box-binding protein 1 (XBP1) and histone deacetylase 3 (HDAC3)-dependent manner. Disturbed flow concomitantly up-regulated the unspliced XBP1 (XBP1u) and HDAC3 in a VEGF receptor and PI3K/Akt-dependent manner. The presence of XBP1 was essential for the up-regulation of HDAC3 protein. Overexpression of XBP1u and/or HDAC3 activated Akt1 phosphorylation, Nrf2 protein stabilization and nuclear translocation, and HO-1 expression. Knockdown of XBP1u decreased the basal level and disturbed flow-induced Akt1 phosphorylation, Nrf2 stabilization, and HO-1 expression. Knockdown of HDAC3 ablated XBP1u-mediated effects. The mammalian target of rapamycin complex 2 (mTORC2) inhibitor, AZD2014, ablated XBP1u or HDAC3 or disturbed flow-mediated Akt1 phosphorylation, Nrf2 nuclear translocation, and HO-1 expression. Neither actinomycin D nor cycloheximide affected disturbed flow-induced up-regulation of Nrf2 protein. Knockdown of Nrf2 abolished XBP1u or HDAC3 or disturbed flow-induced HO-1 up-regulation. Co-immunoprecipitation assays demonstrated that XBP1u physically bound to HDAC3 and Akt1. The region of amino acids 201 to 323 of the HDAC3 protein was responsible for the binding to XBP1u. Double immunofluorescence staining revealed that the interactions between Akt1 and mTORC2, Akt1 and HDAC3, Akt1 and XBP1u, HDAC3, and XBP1u occurred in the cytosol. Thus, we demonstrate that XBP1u and HDAC3 exert a protective effect on disturbed flow-induced oxidative stress via up-regulation of mTORC2-dependent Akt1 phosphorylation and Nrf2-mediated HO-1 expression.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Histona Desacetilases/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Estresse Oxidativo , Fatores de Transcrição/fisiologia , Processamento Alternativo , Animais , Artérias/patologia , Aterosclerose/metabolismo , Sobrevivência Celular , Células Cultivadas , Endotélio Vascular/patologia , Ativação Enzimática , Heme Oxigenase-1/metabolismo , Humanos , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fosforilação , Ligação Proteica , Isoformas de Proteínas/fisiologia , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fluxo Sanguíneo Regional/fisiologia , Fatores de Transcrição de Fator Regulador X , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima , Proteína 1 de Ligação a X-Box
15.
Arterioscler Thromb Vasc Biol ; 34(10): 2184-90, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25012135

RESUMO

Mechanical forces have long been known to play a role in the maintenance of vascular homeostasis in the mature animal and in developmental regulation in the fetus. More recently, it has been shown that stem cells play a role in vascular repair and remodeling in response to biomechanical stress. Laminar shear stress can directly activate growth factor receptors on stem/progenitor cells, initiating signaling pathways leading toward endothelial cell differentiation. Cyclic strain can stimulate stem cell differentiation toward smooth muscle lineages through different mechanisms. In vivo, blood flow in the coronary artery is significantly altered after stenting, leading to changes in biomechanical forces on the vessel wall. This disruption may activate stem cell differentiation into a variety of cells and cause delayed re-endothelialization. Based on progress in the research field, the present review aims to explore the role of mechanical forces in stem cell differentiation both in vivo and in vitro and to examine what this means for the application of stem cells in the clinic, in tissue engineering, and for the management of aberrant stem cell contribution to disease.


Assuntos
Aterosclerose/patologia , Vasos Sanguíneos/patologia , Diferenciação Celular , Linhagem da Célula , Mecanotransdução Celular , Células-Tronco/patologia , Animais , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Aterosclerose/terapia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiopatologia , Proliferação de Células , Humanos , Fenótipo , Regeneração , Fluxo Sanguíneo Regional , Células-Tronco/metabolismo , Estresse Mecânico
16.
Proc Natl Acad Sci U S A ; 109(34): 13793-8, 2012 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-22869753

RESUMO

The generation of induced pluripotent stem (iPS) cells is an important tool for regenerative medicine. However, the main restriction is the risk of tumor development. In this study we found that during the early stages of somatic cell reprogramming toward a pluripotent state, specific gene expression patterns are altered. Therefore, we developed a method to generate partial-iPS (PiPS) cells by transferring four reprogramming factors (OCT4, SOX2, KLF4, and c-MYC) to human fibroblasts for 4 d. PiPS cells did not form tumors in vivo and clearly displayed the potential to differentiate into endothelial cells (ECs) in response to defined media and culture conditions. To clarify the mechanism of PiPS cell differentiation into ECs, SET translocation (myeloid leukemia-associated) (SET) similar protein (SETSIP) was indentified to be induced during somatic cell reprogramming. Importantly, when PiPS cells were treated with VEGF, SETSIP was translocated to the cell nucleus, directly bound to the VE-cadherin promoter, increasing vascular endothelial-cadherin (VE-cadherin) expression levels and EC differentiation. Functionally, PiPS-ECs improved neovascularization and blood flow recovery in a hindlimb ischemic model. Furthermore, PiPS-ECs displayed good attachment, stabilization, patency, and typical vascular structure when seeded on decellularized vessel scaffolds. These findings indicate that reprogramming of fibroblasts into ECs via SETSIP and VEGF has a potential clinical application.


Assuntos
Reprogramação Celular , Células Endoteliais/citologia , Fibroblastos/metabolismo , Neovascularização Patológica , Engenharia Tecidual/métodos , Animais , Antígenos CD/genética , Aorta/patologia , Caderinas/genética , Diferenciação Celular , Células Cultivadas , Fibroblastos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos SCID , Modelos Genéticos , Regiões Promotoras Genéticas , Células-Tronco/citologia , Estresse Mecânico
17.
J Biol Chem ; 288(2): 859-72, 2013 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23184933

RESUMO

Sustained activation of X-box-binding protein 1 (XBP1) results in endothelial cell (EC) apoptosis and atherosclerosis development. The present study provides evidence that XBP1 mRNA splicing triggered an autophagic response in ECs by inducing autophagic vesicle formation and markers of autophagy BECLIN-1 and microtubule-associated protein 1 light chain 3ß (LC3-ßII). Endostatin activated autophagic gene expression through XBP1 mRNA splicing in an inositol-requiring enzyme 1α (IRE1α)-dependent manner. Knockdown of XBP1 or IRE1α by shRNA in ECs ablated endostatin-induced autophagosome formation. Importantly, data from arterial vessels from XBP1 EC conditional knock-out (XBP1eko) mice demonstrated that XBP1 deficiency in ECs reduced the basal level of LC3ß expression and ablated response to endostatin. Chromatin immunoprecipitation assays further revealed that the spliced XBP1 isoform bound directly to the BECLIN-1 promoter at the region from nt -537 to -755. BECLIN-1 deficiency in ECs abolished the XBP1-induced autophagy response, whereas spliced XBP1 did not induce transcriptional activation of a truncated BECLIN-1 promoter. These results suggest that XBP1 mRNA splicing triggers an autophagic signal pathway through transcriptional regulation of BECLIN-1.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Autofagia/genética , Proteínas de Ligação a DNA/genética , Endotélio Vascular/metabolismo , Proteínas de Membrana/genética , Splicing de RNA , RNA Mensageiro/genética , Fatores de Transcrição/genética , Ativação Transcricional/genética , Animais , Sequência de Bases , Proteína Beclina-1 , Células Cultivadas , Imunoprecipitação da Cromatina , Primers do DNA , Endotélio Vascular/citologia , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição de Fator Regulador X , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína 1 de Ligação a X-Box
18.
J Biol Chem ; 288(44): 31853-66, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24045946

RESUMO

Histone deacetylase 3 (HDAC3) plays a critical role in the maintenance of endothelial integrity and other physiological processes. In this study, we demonstrated that HDAC3 undergoes unconventional splicing during stem cell differentiation. Four different splicing variants have been identified, designated as HD3α, -ß, -γ, and -δ, respectively. HD3α was confirmed in stem cell differentiation by specific antibody against the sequences from intron 12. Immunofluorescence staining indicated that the HD3α isoform co-localized with CD31-positive or α-smooth muscle actin-positive cells at different developmental stages of mouse embryos. Overexpression of HD3α reprogrammed human aortic endothelial cells into mesenchymal cells featuring an endothelial-to-mesenchymal transition (EndMT) phenotype. HD3α directly interacts with HDAC3 and Akt1 and selectively activates transforming growth factor ß2 (TGFß2) secretion and cleavage. TGFß2 functioned as an autocrine and/or paracrine EndMT factor. The HD3α-induced EndMT was both PI3K/Akt- and TGFß2-dependent. This study provides the first evidence of the role of HDAC3 splicing in the maintenance of endothelial integrity.


Assuntos
Processamento Alternativo/fisiologia , Comunicação Autócrina/fisiologia , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Histona Desacetilases/biossíntese , Comunicação Parácrina/fisiologia , Fator de Crescimento Transformador beta2/metabolismo , Animais , Linhagem Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células Endoteliais/citologia , Histona Desacetilases/genética , Humanos , Isoenzimas/biossíntese , Isoenzimas/genética , Camundongos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Transformador beta2/genética
19.
Circulation ; 127(16): 1712-22, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23529610

RESUMO

BACKGROUND: Vascular endothelial cell growth factor plays a pivotal role in angiogenesis via regulating endothelial cell proliferation. The X-box binding protein 1 (XBP1) is believed to be a signal transducer in the endoplasmic reticulum stress response. It is unknown whether there is crosstalk between vascular endothelial cell growth factor signaling and XBP1 pathway. METHODS AND RESULTS: We found that vascular endothelial cell growth factor induced the kinase insert domain receptor internalization and interaction through C-terminal domain with the unspliced XBP1 and the inositol requiring enzyme 1 α in the endoplasmic reticulum, leading to inositol requiring enzyme 1 α phosphorylation and XBP1 mRNA splicing, which was abolished by siRNA-mediated knockdown of kinase insert domain receptor. Spliced XBP1 regulated endothelial cell proliferation in a PI3K/Akt/GSK3ß/ß-catenin/E2F2-dependent manner and modulated the cell size increase in a PI3K/Akt/GSK3ß/ß-catenin/E2F2-independent manner. Knockdown of XBP1 or inositol requiring enzyme 1 α decreased endothelial cell proliferation via suppression of Akt/GSK3ß phosphorylation, ß-catenin nuclear translocation, and E2F2 expression. Endothelial cell-specific knockout of XBP1 (XBP1ecko) in mice retarded the retinal vasculogenesis in the first 2 postnatal weeks and impaired the angiogenesis triggered by ischemia. Reconstitution of XBP1 by Ad-XBP1s gene transfer significantly improved angiogenesis in ischemic tissue in XBP1ecko mice. Transplantation of bone marrow from wild-type o XBP1ecko mice could also slightly improve the foot blood reperfusion in ischemic XBP1ecko mice. CONCLUSIONS: These results suggest that XBP1 can function via growth factor signaling pathways to regulate endothelial proliferation and angiogenesis.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/agonistas , Animais , Aorta/citologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/irrigação sanguínea , Estresse do Retículo Endoplasmático/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Terapia Genética , Transplante de Células-Tronco Hematopoéticas , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Isquemia/fisiopatologia , Isquemia/terapia , Perna (Membro)/irrigação sanguínea , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Splicing de RNA/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Quimera por Radiação , Fatores de Transcrição de Fator Regulador X , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/crescimento & desenvolvimento , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Proteína 1 de Ligação a X-Box
20.
Biomater Sci ; 12(7): 1750-1760, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38375548

RESUMO

Diabetes mellitus (DM) is characterized by prolonged hyperglycemia, impaired vascularization, and serious complications, such as blindness and chronic diabetic wounds. About 25% of patients with DM are estimated to encounter impaired healing of diabetic wounds, often leading to lower limb amputation. Multiple factors are attributed to the non-healing of diabetic wounds, including hyperglycaemia, chronic inflammation, and impaired angiogenesis. It is imperative to develop more efficient treatment strategies to tackle healing difficulties in diabetic wounds. Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) are promising for diabetic wound healing considering their anti-inflammatory, pro-angiogenic and pro-proliferative activities. A histone deacetylase 7 (HDAC7)-derived 7-amino-acid peptide (7A) was shown to be highly effective for angiogenesis. However, it has never been investigated whether MSC-EVs are synergistic with 7A for the healing of diabetic wounds. Herein, we propose that MSC-EVs can be combined with 7A to greatly promote diabetic wound healing. The combination of EVs and 7A significantly improved the migration and proliferation of skin fibroblasts. Moreover, EVs alone significantly suppressed LPS-induced inflammation in macrophages, and notably, the combination treatment showed an even better suppression effect. Importantly, the in vivo study revealed that the combination therapy consisting of EVs and 7A in an alginate hydrogel was more efficient for the healing of diabetic wounds in rats than monotherapy using either EV or 7A hydrogels. The underlying mechanisms include suppression of inflammation, improvement of skin cell proliferation and migration, and enhanced collagen fiber disposition and angiogenesis in wounds. In summary, the MSC-EV-7A hydrogel potentially constitutes a novel therapy for efficient healing of chronic diabetic wounds.


Assuntos
Diabetes Mellitus , Células-Tronco Mesenquimais , Humanos , Ratos , Animais , Hidrogéis/química , Angiogênese , Cicatrização , Inflamação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA