Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Allergol Immunopathol (Madr) ; 50(5): 47-56, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36086963

RESUMO

It has been found that angiopoietin-like 4 (ANGPTL4) expression is increased in the serum of patients with chronic obstructive pulmonary disease (COPD). Herein, cigarette smoke extract (CSE) was used to stimulate oxidative stress in bronchial epithelial cells BEAS-2B, and the role and potential mechanism of ANGPTL4 in smoking-induced lung dysfunction were explored. The roles of different concentrations of CSE (0, 1, 2.5, 5, or 10%) in cell viability and ANGPTL4 levels were evaluated. Following ANGPTL4 being knocked down, the effects of ANGPTL4 knockdown on oxidative stress and apoptosis were determined. Moreover, the level of NADPH oxidase 2 (NOX2) was upregulated to assess the mediated role of NOX in the regulation of ANGPTL4, along with JNK/p38 MAPK signaling. CSE treatment elevated the level of ANGPTL4, and ANGPTL4 knockdown reduced CSE-induced oxidative stress, apoptosis, and NOX level in BEAS-2B cells. The greatest degree of alteration was found in NOX2, and additional NOX2 overexpression broke the inhibitory influences of ANGPTL4 knockdown on oxidative stress and apoptosis. Otherwise, ANGPTL4 knockdown hindered the activation of JNK/p38 MAPK signaling, whereas NOX2 overexpression activated this signaling pathway. Together, ANGPTL4 knockdown attenuated CSE-induced oxidative stress, apoptosis, and activation of JNK/MAPK signaling by inhibiting NOX.


Assuntos
Fumar Cigarros , NADPH Oxidases , Angiopoietinas/metabolismo , Angiopoietinas/farmacologia , Apoptose , Linhagem Celular , Fumar Cigarros/efeitos adversos , Células Epiteliais/metabolismo , Humanos , Pulmão , NADPH Oxidases/genética , Estresse Oxidativo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Mol Biol Rep ; 48(1): 941-950, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33393005

RESUMO

Tissue engineering is a rapidly developing field with many potential clinical applications in tissue and organ regeneration. The development of a mature and stable vasculature within these engineered tissues (ET) remains a significant obstacle. Currently, several growth factors (GFs) have been identified to play key roles within in vivo angiogenesis, including vascular endothelial growth factor (VEGF), platelet derived growth factor (PDGF), FGF and angiopoietins. In this article we attempt to build on in vivo principles to review the single, dual and multiple GF release systems and their effects on promoting angiogenesis. We conclude that multiple GF release systems offer superior results compared to single and dual systems with more stable, mature and larger vessels produced. However, with more complex release systems this raises other problems such as increased cost and significant GF-GF interactions. Upstream regulators and pericyte-coated scaffolds could provide viable alternative to circumnavigate these issues.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/farmacologia , Neovascularização Fisiológica/genética , Engenharia Tecidual/métodos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Angiopoietinas/genética , Angiopoietinas/metabolismo , Angiopoietinas/farmacologia , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Metaloproteinases da Matriz/farmacologia , Pericitos/citologia , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Regeneração/efeitos dos fármacos , Regeneração/genética , Alicerces Teciduais , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Biochim Biophys Acta ; 1861(2): 130-137, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26569053

RESUMO

AIM: Atypical angiopoietin-like 8 (ANGPTL8), also known as betatrophin, is known to regulate lipid metabolism. However, its mechanism of action remains elusive. METHODS: HepG2, 3T3-L1, and NIT-1 cells were cultured in amino acid-complete MEM or histidine-free MEM to detect ANGPTL8 expression. The three cell types were treated with or without recombinant ANGPTL8 to investigate its role in lipid metabolism. Hydrodynamic tail vein gene delivery was also used to examine the role of ANGPTL8 in mice. RESULTS: ANGPTL8 is significantly up-regulated in amino acid-deprived cultured cells in vitro. The activation of ANGPTL8 gene transcription was mediated through the RAS/c-RAF/MAPK signaling pathway rather than the general GCN2/ATF4 pathways. ANGPTL8 activated the ERK signal transduction pathway in hepatocytes, adipocytes, and pancreatic ß-cells, up-regulating early growth response transcription factor (Egr1) and down-regulating adipose triglyceride lipase (ATGL). CONCLUSION: ANGPTL8 is a stress-response protein that regulates fat metabolism by suppressing ATGL expression, revealing a mechanistic connection between ANGPTL8 and lipid homeostasis in mammalian cells.


Assuntos
Adipócitos/metabolismo , Angiopoietinas/genética , Lipase/genética , Triglicerídeos/metabolismo , Células 3T3-L1 , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/metabolismo , Angiopoietinas/farmacologia , Animais , Diferenciação Celular , Linhagem Celular , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Meios de Cultura/química , Meios de Cultura/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Glicerol/metabolismo , Células Hep G2 , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Lipase/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Proteínas ras/genética , Proteínas ras/metabolismo
4.
J Cell Biochem ; 116(1): 45-57, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25145474

RESUMO

A key feature in the induction of pathological angiogenesis is that inflammation precedes and accompanies the formation of neovessels as evidenced by increased vascular permeability and the recruitment of inflammatory cells. Previously, we and other groups have shown that selected growth factors, namely vascular endothelial growth factor (VEGF) and angiopoietins (Ang1 and Ang2) do not only promote angiogenesis, but can also induce inflammatory response. Herein, given a pro-inflammatory environment, we addressed the individual capacity of VEGF and angiopoietins to promote the formation of mature neovessels and to identify the different types of inflammatory cells accompanying the angiogenic process over time. Sterilized polyvinyl alcohol (PVA) sponges soaked in growth factor-depleted Matrigel mixed with PBS, VEGF, Ang1, or Ang2 (200 ng/200 µl) were subcutaneously inserted into anesthetized mice. Sponges were removed at day 4, 7, 14, or 21 post-procedure for histological, immunohistological (IHC), and flow cytometry analyses. As compared to PBS-treated sponges, the three growth factors promoted the recruitment of inflammatory cells, mainly neutrophils and macrophages, and to a lesser extent, T- and B-cells. In addition, they were more potent and more rapid in the recruitment of endothelial cells (ECs) and in the formation and maturation (ensheating of smooth muscle cells around ECs) of neovessels. Thus, the autocrine/paracrine interaction among the different inflammatory cells in combination with VEGF, Ang1, or Ang2 provides a suitable microenvironment for the formation and maturation of blood vessels.


Assuntos
Angiopoietinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Angiopoietina-1/farmacologia , Angiopoietina-2/farmacologia , Animais , Colágeno/química , Combinação de Medicamentos , Citometria de Fluxo , Imuno-Histoquímica , Laminina/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Fagocitose/genética , Fagocitose/fisiologia , Proteoglicanas/química
5.
Haematologica ; 100(5): 585-94, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25637050

RESUMO

Successful expansion of hematopoietic stem cells would benefit the use of hematopoietic stem cell transplants in the clinic. Several angiopoietin-like proteins, including angiopoietin-like 7, can support the activity of hematopoietic stem cells. However, effects of ANGPTL7 on human hematopoietic stem cells and the downstream signaling cascade activated by ANGPTL7 are poorly understood. Here, we established a human hematopoietic stem and progenitor cell-supportive mouse fetal liver cell line that specifically expressed the Angptl7 protein. Furthermore, we found ANGPTL7 is capable of stimulating human hematopoietic stem and progenitor cell expansion and increasing the repopulation activities of human hematopoietic progenitors in xenografts. RNA-sequencing analysis showed that ANGPTL7 activated the expression of CXCR4, HOXB4 and Wnt downstream targets in human hematopoietic progenitors. In addition, chemical manipulation of Wnt signaling diminished the effects of ANGPTL7 on human hematopoietic stem and progenitor cells in culture. In summary, we identify the secreted growth factor ANGPTL7 as a regulator of both human hematopoietic stem and progenitor cell expansion and regeneration.


Assuntos
Angiopoietinas/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteína 7 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/farmacologia , Animais , Comunicação Celular , Análise por Conglomerados , Técnicas de Cocultura , Sangue Fetal/citologia , Expressão Gênica , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Camundongos , Camundongos Knockout , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Células Estromais/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
6.
Mol Ther ; 22(9): 1593-604, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24903577

RESUMO

Impaired wound healing is a major source of morbidity in diabetic patients. Poor outcome has, in part, been related to increased inflammation, poor angiogenesis, and deficiencies in extracellular matrix components. Despite the enormous impact of these chronic wounds, effective therapies are lacking. Here, we showed that the topical application of recombinant matricellular protein angiopoietin-like 4 (ANGPTL4) accelerated wound reepithelialization in diabetic mice, in part, by improving angiogenesis. ANGPTL4 expression is markedly elevated upon normal wound injury. In contrast, ANGPTL4 expression remains low throughout the healing period in diabetic wounds. Exogenous ANGPTL4 modulated several regulatory networks involved in cell migration, angiogenesis, and inflammation, as evidenced by an altered gene expression signature. ANGPTL4 influenced the expression profile of endothelial-specific CD31 in diabetic wounds, returning its profile to that observed in wild-type wounds. We showed ANGPTL4-induced nitric oxide production through an integrin/JAK/STAT3-mediated upregulation of inducible nitric oxide synthase (iNOS) expression in wound epithelia, thus revealing a hitherto unknown mechanism by which ANGPTL4 regulated angiogenesis via keratinocyte-to-endothelial-cell communication. These data show that the replacement of ANGPTL4 may be an effective adjunctive or new therapeutic avenue for treating poor healing wounds. The present finding also confirms that therapeutic angiogenesis remains an attractive treatment modality for diabetic wound healing.


Assuntos
Angiopoietinas/administração & dosagem , Diabetes Mellitus Experimental/complicações , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Transcrição STAT3/metabolismo , Angiopoietinas/farmacologia , Animais , Comunicação Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Queratinócitos/metabolismo , Camundongos , Reepitelização , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Transdução de Sinais
7.
Angiogenesis ; 17(3): 675-83, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24563071

RESUMO

Angiopoietin-like 2 (ANGPTL2) has been reported to induce sprouting angiogenesis; however, its role in vasculogenesis, the de novo lumenization of endothelial cells (EC), remains unexplored. We sought to investigate the potential role of ANGPTL2 in regulating human cord blood derived endothelial colony forming cell (ECFC) vasculogenesis through siRNA mediated inhibition of ANGPTL2 gene expression. We found that ECFCs in which ANGPTL2 was diminished displayed a threefold decrease in in vitro lumenal area whereas addition of exogenous ANGPTL2 protein domains to ECFCs lead to increased lumen formation within a 3 dimensional (3D) collagen assay of vasculogenesis. ECFC migration was attenuated by 36 % via ANGPTL2 knockdown (KD) although proliferation and apoptosis were not affected. We subsequently found that c-Jun NH2-terminal kinase (JNK), but not ERK1/2, phosphorylation was decreased upon ANGPTL2 KD, and expression of membrane type 1 matrix metalloproteinase (MT1-MMP), known to be regulated by JNK and a critical regulator of EC migration and 3D lumen formation, was decreased in lumenized structures in vitro derived from ANGPTL2 silenced ECFCs. Treatment of ECFCs in 3D collagen matrices with either a JNK inhibitor or exogenous rhTIMP-3 (an inhibitor of MT1-MMP activity) resulted in a similar phenotype of decreased vascular lumen formation as observed with ANGPTL2 KD, whereas stimulation of JNK activity increased vasculogenesis. Based on gene silencing, pharmacologic, cellular, and biochemical approaches, we conclude that ANGPTL2 positively regulates ECFC vascular lumen formation likely through its effects on migration and in part by activating JNK and increasing MT1-MMP expression.


Assuntos
Angiopoietinas/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Neovascularização Fisiológica , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/genética , Angiopoietinas/farmacologia , Ensaios de Migração Celular , Movimento Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
8.
Eur Heart J ; 34(47): 3657-68, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23674618

RESUMO

AIMS: Given the impact of vascular injuries and oedema on brain damage caused during stroke, vascular protection represents a major medical need. We hypothesized that angiopoietin-like 4 (ANGPTL4), a regulator of endothelial barrier integrity, might exert a protective effect during ischaemic stroke. METHODS AND RESULTS: Using a murine transient ischaemic stroke model, treatment with recombinant ANGPTL4 led to significantly decreased infarct size and improved behaviour. Quantitative characteristics of the vascular network (density and branchpoints) were preserved in ANGPTL4-treated mice. Integrity of tight and adherens junctions was also quantified and ANGPTL4-treated mice displayed increased VE-cadherin and claudin-5-positive areas. Brain oedema was thus significantly decreased in ANGPTL4-treated mice. In accordance, vascular damage and infarct severity were increased in angptl4-deficient mice thus providing genetic evidence that ANGPTL4 preserves brain tissue from ischaemia-induced alterations. Altogether, these data show that ANGPTL4 protects not only the global vascular network, but also interendothelial junctions and controls both deleterious inflammatory response and oedema. Mechanistically, ANGPTL4 counteracted VEGF signalling and thereby diminished Src-signalling downstream from VEGFR2. This led to decreased VEGFR2-VE-cadherin complex disruption, increased stability of junctions and thus increased endothelial cell barrier integrity of the cerebral microcirculation. In addition, ANGPTL4 prevented neuronal loss in the ischaemic area. CONCLUSION: These results, therefore, show ANGPTL4 counteracts the loss of vascular integrity in ischaemic stroke, by restricting Src kinase signalling downstream from VEGFR2. ANGPTL4 treatment thus reduces oedema, infarct size, neuronal loss, and improves mice behaviour. These results suggest that ANGPTL4 constitutes a relevant target for vasculoprotection and cerebral protection during stroke.


Assuntos
Angiopoietinas/farmacologia , Isquemia Encefálica/prevenção & controle , Nootrópicos/farmacologia , Acidente Vascular Cerebral/prevenção & controle , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/deficiência , Animais , Barreira Hematoencefálica/fisiologia , Encéfalo/irrigação sanguínea , Edema Encefálico/prevenção & controle , Isquemia Encefálica/fisiopatologia , Caderinas/fisiologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Encefalite/fisiopatologia , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Quinases da Família src/fisiologia
9.
J Biol Chem ; 287(11): 8444-56, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22267746

RESUMO

Intracellular triacylglycerol (TG) hydrolysis and fatty acid release by the white adipose tissue (WAT) during a fast is stimulated by counter-regulatory factors acting in concert, although how adipocytes integrate these lipolytic inputs is unknown. We tested the role of angiopoietin-like 4 (Angptl4), a secreted protein induced by fasting or glucocorticoid treatment, in modulating intracellular adipocyte lipolysis. Glucocorticoid receptor blockade prevented fasting-induced tissue Angptl4 expression and WAT TG hydrolysis in mice, and TG hydrolysis induced by fasts of 6 or 24 h was greatly reduced in mice lacking Angptl4 (Angptl4(-/-)). Glucocorticoid treatment mimicked the lipolytic effects of fasting, although with slower kinetics, and this too required Angptl4. Thus, fasting-induced WAT TG hydrolysis requires glucocorticoid action and Angptl4. Both fasting and glucocorticoid treatment also increased WAT cAMP levels and downstream phosphorylation of lipolytic enzymes. Angptl4 deficiency markedly reduced these effects, suggesting that Angptl4 may stimulate lipolysis by modulating cAMP-dependent signaling. In support of this, cAMP levels and TG hydrolysis were reduced in primary Angptl4(-/-) murine adipocytes treated with catecholamines, which stimulate cAMP-dependent signaling to promote lipolysis, and was restored by treatment with purified human ANGPTL4. Remarkably, human ANGPTL4 treatment alone increased cAMP levels and induced lipolysis in these cells. Pharmacologic agents revealed that Angptl4 modulation of cAMP-dependent signaling occurs upstream of adenylate cyclase and downstream of receptor activation. We show that Angptl4 is a glucocorticoid-responsive mediator of fasting-induced intracellular lipolysis and stimulates cAMP signaling in adipocytes. Such a role is relevant to diseases of aberrant lipolysis, such as insulin resistance.


Assuntos
Adipócitos Brancos/metabolismo , Tecido Adiposo Branco/metabolismo , Angiopoietinas/metabolismo , Jejum/fisiologia , Lipólise/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Angiopoietinas/genética , Angiopoietinas/farmacologia , Animais , AMP Cíclico/genética , AMP Cíclico/metabolismo , Glucocorticoides/metabolismo , Glucocorticoides/farmacologia , Humanos , Resistência à Insulina/genética , Lipólise/efeitos dos fármacos , Camundongos , Camundongos Knockout , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Triglicerídeos/genética , Triglicerídeos/metabolismo
10.
Biochim Biophys Acta ; 1821(10): 1370-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22732211

RESUMO

Lipoprotein lipase (LPL) is important for clearance of triacylglycerols (TG) from plasma both as an enzyme and as a bridging factor between lipoproteins and receptors for endocytosis. The amount of LPL at the luminal side of the capillary endothelium determines to what extent lipids are taken up. Mechanisms to control both the activity of LPL and its transport to the endothelial sites are regulated, but poorly understood. Angiopoietin-like proteins (ANGPTLs) 3 and 4 are potential control proteins for LPL, but plasma concentrations of ANGPTLs do not correlate with plasma TG levels. We investigated the effects of recombinant human N-terminal (NT) ANGPTLs3 and 4 on LPL-mediated bridging of TG-rich lipoproteins to primary mouse hepatocytes and found that the NT-ANGPTLs, in concentrations sufficient to cause inactivation of LPL in vitro, were unable to prevent LPL-mediated lipoprotein uptake. We therefore investigated the effects of lipoproteins (chylomicrons, VLDL and LDL) on the inactivation of LPL in vitro by NT-ANGPTLs3 and 4 and found that LPL activity was protected by TG-rich lipoproteins. In vivo, postprandial TG protected LPL from inactivation by recombinant NT-ANGPTL4 injected to mice. We conclude that lipoprotein-bound LPL is stabilized against inactivation by ANGPTLs. The levels of ANGPTLs found in blood may not be sufficient to overcome this stabilization. Therefore it is likely that the prime site of action of ANGPTLs on LPL is in subendothelial compartments where TG-rich lipoprotein concentration is lower than in blood. This could explain why the plasma levels of TG and ANGPTLs do not correlate.


Assuntos
Angiopoietinas/farmacologia , Lipase Lipoproteica/metabolismo , Lipoproteínas/fisiologia , Triglicerídeos/fisiologia , Proteína 3 Semelhante a Angiopoietina , Proteína 4 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Animais , Quilomícrons/fisiologia , Ativação Enzimática , Hepatócitos/metabolismo , Humanos , Lipoproteínas LDL/fisiologia , Lipoproteínas VLDL/fisiologia , Camundongos
11.
Nat Med ; 12(2): 240-5, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16429146

RESUMO

Successful ex vivo expansion of hematopoietic stem cells (HSCs) would greatly benefit the treatment of disease and the understanding of crucial questions of stem cell biology. Here we show, using microarray studies, that the HSC-supportive mouse fetal liver CD3(+) cells specifically express the proteins angiopoietin-like 2 (Angptl2) and angiopoietin-like 3 (Angptl3). We observed a 24- or 30-fold net expansion of long-term HSCs by reconstitution analysis when we cultured highly enriched HSCs for 10 days in the presence of Angptl2 or Angptl3 together with saturating levels of other growth factors. The coiled-coil domain of Angptl2 was capable of stimulating expansion of HSCs. Furthermore, angiopoietin-like 5, angiopoietin-like 7 and microfibril-associated glycoprotein 4 also supported expansion of HSCs in culture.


Assuntos
Angiopoietinas/farmacologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/genética , Angiopoietinas/metabolismo , Animais , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Proteínas Sanguíneas/farmacologia , Linhagem Celular , Hematopoese/efeitos dos fármacos , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transfecção
12.
Acta Histochem ; 125(8): 152100, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37837833

RESUMO

OBJECTIVE: Idiopathic pulmonary fibrosis (IPF) is considered as a chronic interstitial lung disease with underlying mechanism of IPF remaining unclear, while there are no definitive treatment options. In recent years, scientists have gradually paid attention to the influence of angiogenesis on IPF. Because IPF is a progressive with microvascular remodeling disorder, scientists have postulated that angiogenesis may also be one of the initiating and contributing factors of the disease. Bupleurum is a common natural Chinese herbal medicine with antibacterial, anti-inflammatory, anti-tumor and other pharmacological effects. As the most important active monomer of Bupleurum, Saikosaponin-d (SSd) is a new discovery with anti-pulmonary fibrosis (PF) activity. This study attempts to investigate the role of SSd in the interference of PF through regulation of angiogenesis in IPF through Angiopoietin (Angpt) /Tie receptor 2 (Tie2) pathway. METHODS: Randomly, we allocated C57BL/6 mice into four groups (n = 20 in each group). Afterwards, establishment of IPF model was accomplished via intratracheal administration of bleomycin (BLM, 5 mg/kg), while corresponding drug intervention was given accordingly. On 3rd, 7th, 14th and 28th days after modeling, we performed histopathological examination through staining. Meanwhile, immunohistochemistry (IHC) of PF and the expression of related factors were observed, while Ang/Tie2 pathway was assessed by ELISA with the effect of SSd on angiogenesis related proteins in IPF being explored with IHC and Western Blot technique. RESULTS: Our results showed that SSd could reduce inflammation and PF levels in lung tissue of experimental mice, while levels of angiogenesis-related factors, namely Tie-2, Ang-1 and ANGPT2 (Ang-2), fibrosis- associated factors like Alpha-smooth muscle actin (α-SMA), collagen-I and hydroxyproline in SSd and dexamethasone (DXM) mice were significantly reduced at each time point compared to BLM (p < 0.01). Additionally, we discovered substantial decreased expressions of Ang-1, Ang-2, Tie-2, α-SMA and collagen-I at protein level in SSd and DXM mice at each time point compared to BLM (p < 0.05). Besides, insignificant differences were observed between SSd and DXM groups (p > 0.05). CONCLUSION: This study has demonstrated that SSd could down-regulate the expression of ANG-1, Ang-2 and Tie2 in the Ang/Tie2 pathway, and may reduce lung inflammation and PF in BLM-induced mice via inhibition of angiogenesis.


Assuntos
Angiopoietinas , Fibrose Pulmonar Idiopática , Camundongos , Animais , Angiopoietinas/metabolismo , Angiopoietinas/farmacologia , Camundongos Endogâmicos C57BL , Pulmão/metabolismo , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Colágeno Tipo I/metabolismo , Bleomicina/farmacologia , Bleomicina/metabolismo
13.
J Exp Med ; 220(1)2023 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-36269299

RESUMO

Primary tumors and distant site metastases form a bidirectionally communicating system. Yet, the molecular mechanisms of this crosstalk are poorly understood. Here, we identified the proteolytically cleaved fragments of angiopoietin-like 4 (ANGPTL4) as contextually active protumorigenic and antitumorigenic contributors in this communication ecosystem. Preclinical studies in multiple tumor models revealed that the C-terminal fragment (cANGPTL4) promoted tumor growth and metastasis. In contrast, the N-terminal fragment of ANGPTL4 (nANGPTL4) inhibited metastasis and enhanced overall survival in a postsurgical metastasis model by inhibiting WNT signaling and reducing vascularity at the metastatic site. Tracing ANGPTL4 and its fragments in tumor patients detected full-length ANGPTL4 primarily in tumor tissues, whereas nANGPTL4 predominated in systemic circulation and correlated inversely with disease progression. The study highlights the spatial context of the proteolytic cleavage-dependent pro- and antitumorigenic functions of ANGPTL4 and identifies and validates nANGPTL4 as a novel biomarker of tumor progression and antimetastatic therapeutic agent.


Assuntos
Proteína 4 Semelhante a Angiopoietina , Neoplasias , Humanos , Proteína 4 Semelhante a Angiopoietina/farmacologia , Proteína 4 Semelhante a Angiopoietina/uso terapêutico , Angiopoietinas/farmacologia , Angiopoietinas/uso terapêutico , Biomarcadores Tumorais , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico
14.
Blood Cells Mol Dis ; 48(1): 25-9, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21983347

RESUMO

Several angiopoietin-like (ANGPTL) molecules have been implicated in enhancement of ex-vivo expansion of murine and human (hu) hematopoietic stem cells, but there are no reports on hematopoietic progenitor cells (HPCs). We assessed purified recombinant endotoxin-free hu ANGPTL-2 Coiled-Coil (CC), -3, -3CC, -3 fibrinogen-like domain (FLD), -4, -4CC, -5CC, -6 and -7 for effects on proliferation and survival of HPCs from hu cord blood (CB). None of the ANGPTL molecules stimulated CB HPC proliferation, or enhanced or inhibited colony formation of CB HPC stimulated by various growth factors. However, ANGPTL-2CC, -3, and -3CC significantly enhanced survival of HPC (CFU-GM, BFU-E, CFU-GEMM) subjected to delayed addition of growth factors. Survival enhancing effects of ANGPTL-3 were neutralized by purified anti-ANGPTL-3, but not by anti-ANGPTL-4, -6, or -7. ANGPTL-2CC, -3, and -3CC, but not -4, -6, or -7 also enhanced replating capacity of single CB CFU-GEMM colonies, an estimate of the self-renewal capabilities of HPCs, by greater than 2 fold. Effects of at least ANGPTL-3CC may in part be mediated through phosphorylation of ERK. The ANGPTL molecules did not influence ex-vivo expansion of hu CB CD34(+) cells, alone, or in combination with SCF, TPO, Flt3-ligand, with or without IL-3. Thus, among ANGPTL family members, ANGPTL-2 and -3 had enhancing activities on human HPC survival and replating activity, effects requiring the CC domain of the ANGPTL molecules. This information is of relevance to hu HPC regulation.


Assuntos
Angiopoietinas/genética , Sangue Fetal/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteínas Recombinantes/genética , Transdução de Sinais/fisiologia , Proteína 2 Semelhante a Angiopoietina , Proteína 3 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/antagonistas & inibidores , Angiopoietinas/química , Angiopoietinas/farmacologia , Anticorpos Neutralizantes/farmacologia , Antígenos CD34/biossíntese , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Eritropoetina/farmacologia , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia
15.
Nat Med ; 28(12): 2633-2645, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36456835

RESUMO

Osteoarthritis (OA) is a common, debilitating, chronic disease with no disease-modifying drug approved to date. We discovered LNA043-a derivative of angiopoietin-like 3 (ANGPTL3)-as a potent chondrogenesis inducer using a phenotypic screen with human mesenchymal stem cells. We show that LNA043 promotes chondrogenesis and cartilage matrix synthesis in vitro and regenerates hyaline articular cartilage in preclinical OA and cartilage injury models in vivo. LNA043 exerts at least part of these effects through binding to the fibronectin receptor, integrin α5ß1 on mesenchymal stem cells and chondrocytes. In a first-in-human (phase 1), randomized, double-blinded, placebo-controlled, single ascending dose, single-center trial ( NCT02491281 ; sponsored by Novartis Pharmaceuticals), 28 patients with knee OA were injected intra-articularly with LNA043 or placebo (3:1 ratio) either 2 h, 7 d or 21 d before total knee replacement. LNA043 met its primary safety endpoint and showed short serum pharmacokinetics, cartilage penetration and a lack of immunogenicity (secondary endpoints). Post-hoc transcriptomics profiling of cartilage revealed that a single LNA043 injection reverses the OA transcriptome signature over at least 21 d, inducing the expression of hyaline cartilage matrix components and anabolic signaling pathways, while suppressing mediators of OA progression. LNA043 is a novel disease-modifying OA drug candidate that is currently in a phase 2b trial ( NCT04864392 ) in patients with knee OA.


Assuntos
Cartilagem Articular , Osteoartrite do Joelho , Humanos , Osteoartrite do Joelho/tratamento farmacológico , Condrócitos , Transdução de Sinais , Angiopoietinas/metabolismo , Angiopoietinas/farmacologia , Angiopoietinas/uso terapêutico , Proteína 3 Semelhante a Angiopoietina
16.
Int J Biol Macromol ; 221: 1428-1438, 2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36122781

RESUMO

Angiogenesis forms new vessels from existing ones. Abnormal angiogenesis, which is what gives tumor microenvironments their distinctive features, is characterised by convoluted, permeable blood vessels with a variety of shapes and high perfusion efficiency. Tumor angiogenesis controls cancer growth by allowing invasion and metastasis and is highly controlled by signalling networks. Therapeutic techniques targeting VEGF, PDGF, FGF Notch, Angiopoietin, and HGF signalling restrict the tumor's vascular supply. Numerous pathways regulate angiogenesis, and when one of those processes is blocked, the other pathways may step in to help. VEGF signalling inhibition alone has limits as an antiangiogenic therapy, and additional angiogenic pathways such as FGF, PDGF, Notch, angiopoietin, and HGF are important. For the treatment of advanced solid tumors, there are also new, emerging medicines that target multiple angiogenic pathways. Recent therapies block numerous signalling channels concurrently. This study focuses on 'alternative' methods to standard antiangiogenic medicines, such as cyclooxygenase-2 blocking, oligonucleotide binding complementary sites to noncoding RNAs to regulate mRNA target, matrix metalloproteinase inhibition and CRISPR/Cas9 based gene edition and dissecting alternative angiogenesis mechanism in tumor microenvironment.


Assuntos
Neoplasias , Fator A de Crescimento do Endotélio Vascular , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Angiopoietinas/farmacologia , Angiopoietinas/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Transdução de Sinais , Inibidores da Angiogênese/farmacologia , Neoplasias/metabolismo , Microambiente Tumoral , Fator de Crescimento de Hepatócito/uso terapêutico
17.
J Biol Chem ; 285(43): 32999-33009, 2010 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-20729546

RESUMO

A dynamic cell-matrix interaction is crucial for a rapid cellular response to changes in the environment. Appropriate cell behavior in response to the changing wound environment is required for efficient wound closure. However, the way in which wound keratinocytes modify the wound environment to coordinate with such cellular responses remains less studied. We demonstrated that angiopoietin-like 4 (ANGPTL4) produced by wound keratinocytes coordinates cell-matrix communication. ANGPTL4 interacts with vitronectin and fibronectin in the wound bed, delaying their proteolytic degradation by metalloproteinases. This interaction does not interfere with integrin-matrix protein recognition and directly affects cell-matrix communication by altering the availability of intact matrix proteins. These interactions stimulate integrin- focal adhesion kinase, 14-3-3, and PKC-mediated signaling pathways essential for effective wound healing. The deficiency of ANGPTL4 in mice delays wound re-epithelialization. Further analysis revealed that cell migration was impaired in the ANGPTL4-deficient keratinocytes. Altogether, the findings provide molecular insight into a novel control of wound healing via ANGPTL4-dependent regulation of cell-matrix communication. Given the known role of ANGPTL4 in glucose and lipid homeostasis, it is a prime therapeutic candidate for the treatment of diabetic wounds. It also underscores the importance of cell-matrix communication during angiogenesis and cancer metastasis.


Assuntos
Angiopoietinas/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Queratinócitos/metabolismo , Cicatrização , Ferimentos e Lesões/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Angiopoietinas/farmacologia , Animais , Complicações do Diabetes/tratamento farmacológico , Complicações do Diabetes/genética , Complicações do Diabetes/metabolismo , Proteínas da Matriz Extracelular/genética , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ferimentos e Lesões/tratamento farmacológico , Ferimentos e Lesões/genética
18.
FASEB J ; 24(12): 4648-59, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20667978

RESUMO

Hypoxia and the hypoxia-inducible factor (HIF) transcription factor regulate angiogenic-osteogenic coupling and osteoclast-mediated bone resorption. To determine how HIF might coordinate osteoclast and osteoblast function, we studied angiopoietin-like 4 (ANGPTL4), the top HIF target gene in an Illumina HumanWG-6 v3.0 48k array of normoxic vs. hypoxic osteoclasts differentiated from human CD14(+) monocytes (14.3-fold induction, P<0.0004). ANGPTL4 mRNA and protein were induced by 24 h at 2% O(2) in human primary osteoclasts, monocytes, and osteoblasts. ANGPTL4 protein was observed by immunofluorescence in osteoclasts and osteoblasts in vivo. Normoxic inducers of HIF (CoCl(2), desferrioxamine, and l-mimosine) and 100 ng/ml ANGPTL4 stimulated osteoclastic resorption 2- to 3-fold in assays of lacunar dentine resorption, without affecting osteoclast viability. Isoform-specific HIF-1α small interfering RNA ablated hypoxic induction of ANGPTL4 and of resorption, which was rescued by addition of exogenous ANGPTL4 (P<0.001). In the osteoblastic Saos2 cell line, ANGPTL4 caused a dose-dependent increase in proliferation (P<0.01, 100 ng/ml) and, at lower doses (1-25 ng/ml), mineralization. These results demonstrate that HIF is sufficient to enhance osteoclast-mediated bone resorption and that ANGPTL4 can compensate for HIF-1α deficiency with respect to stimulation of osteoclast activity and also augments osteoblast proliferation and differentiation.


Assuntos
Angiopoietinas/metabolismo , Reabsorção Óssea/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Angiopoietinas/farmacologia , Western Blotting , Reabsorção Óssea/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Imuno-Histoquímica , Osteoclastos/efeitos dos fármacos , Reação em Cadeia da Polimerase , RNA Interferente Pequeno
19.
J Cell Biol ; 169(2): 239-43, 2005 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-15851516

RESUMO

The Tie1 receptor tyrosine kinase was isolated over a decade ago, but so far no ligand has been found to activate this receptor. Here, we have examined the potential of angiopoietins, ligands for the related Tie2 receptor, to mediate Tie1 activation. We show that a soluble Ang1 chimeric protein, COMP-Ang1, stimulates Tie1 phosphorylation in endothelial cells with similar kinetics and angiopoietin dose dependence when compared with Tie2. The phosphorylation of overexpressed Tie1 was weakly induced by COMP-Ang1 also in transfected cells that do not express Tie2. When cotransfected, Tie2 formed heteromeric complexes with Tie1, enhanced Tie1 activation, and induced phosphorylation of a kinase-inactive Tie1 in a ligand-dependent manner. Tie1 phosphorylation was also induced by native Ang1 and Ang4, although less efficiently than with COMP-Ang1. In conclusion, we show that Tie1 phosphorylation is induced by multiple angiopoietin proteins and that the activation is amplified via Tie2. These results should be important in dissecting the signal transduction pathways and biological functions of Tie1.


Assuntos
Angiopoietinas/farmacologia , Células Endoteliais/enzimologia , Ativação Enzimática/efeitos dos fármacos , Receptor de TIE-1/metabolismo , Receptor TIE-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Angiopoietinas/genética , Linhagem Celular , Expressão Gênica , Humanos , Fosforilação , Receptor de TIE-1/genética , Receptor TIE-2/genética , Proteínas Recombinantes de Fusão/farmacologia , Transfecção
20.
Circ Res ; 102(4): 497-504, 2008 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-18096817

RESUMO

Tyr83 in endothelial nitric oxide synthase (eNOS) has been identified previously as a site of Src kinase-mediated phosphorylation of eNOS in bovine aortic endothelial cells (BAECs) that is phosphorylated in response to oxidant stress. In the present study, we have used a phospho-specific antibody to show that Tyr83 in eNOS is also phosphorylated in both BAECs and intact blood vessel segments in response to treatment with a variety of different eNOS-activating agonists, including thapsigargin, vascular endothelial growth factor, bradykinin, ATP, sphingosine-1-phosphate, estrogen, angiopoietin, and acetylcholine. Agonist stimulation of eNOS Tyr83 phosphorylation as well as agonist stimulation of endothelial NO release in BAECs is blocked by Src kinase inhibition by either 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo [3,4-d] pyrimidine (PP2) or by dominant negative Src. Mutation of Tyr83 to a nonphosphorylatable Phe blocks agonist stimulation of NO release from eNOS-reconstituted eNOS knockdown endothelial cells. Mutation of Tyr83 also attenuates agonist-induced relaxation of eNOS-reconstituted aortic rings from eNOS knockout mice. Phosphorylation of eNOS at Tyr83 thus appears to be a common covalent modification that is induced, not only by oxidant stress but also by other physiologically relevant extracellular signals known to be important in regulation of eNOS activity in vivo. Moreover, our results demonstrate an important role for Src-mediated phosphorylation of eNOS at Tyr83 in agonist stimulation of eNOS activation and vascular relaxation.


Assuntos
Células Endoteliais/enzimologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Vasodilatação/fisiologia , Trifosfato de Adenosina/farmacologia , Angiopoietinas/farmacologia , Animais , Aorta/citologia , Bradicinina/farmacologia , Células COS , Bovinos , Chlorocebus aethiops , Células Endoteliais/citologia , Inibidores Enzimáticos/farmacologia , Estrogênios/farmacologia , Humanos , Rim/citologia , Lisofosfolipídeos/farmacologia , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tapsigargina/farmacologia , Tirosina/metabolismo , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA