Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Circ Res ; 125(3): 328-340, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31159652

RESUMO

RATIONALE: Noncoding RNAs (ncRNAs), including microRNAs (miRNAs), circular RNAs (circRNAs), and long noncoding RNAs (lncRNAs), are proposed novel biomarkers of myocardial injury. Their release kinetics have not been explored without confounding by heparin nor has their relationship to myocardial protein biomarkers. OBJECTIVE: To compare ncRNA types in heparinase-treated samples with established and emerging protein biomarkers for myocardial injury. METHODS AND RESULTS: Screening of 158 circRNAs and 21 lncRNAs in human cardiac tissue identified 12 circRNAs and 11 lncRNAs as potential biomarkers with cardiac origin. Eleven miRNAs were included. At low spike-in concentrations of myocardial tissue, significantly higher regression coefficients were observed across ncRNA types compared with cardiac troponins and cMyBP-C (cardiac myosin-binding protein C). Heparinase treatment of serial plasma and serum samples of patients undergoing transcoronary ablation of septal hypertrophy removed spurious correlations between miRNAs in non-heparinase-treated samples. After transcoronary ablation of septal hypertrophy, muscle-enriched miRNAs (miR-1 and miR-133a) showed a steeper and earlier increase than cardiac-enriched miRNAs (miR-499 and miR-208b). Putative cardiac lncRNAs, including LIPCAR (long intergenic noncoding RNA predicting cardiac remodeling and survival), did not rise, refuting a predominant cardiac origin. Cardiac circRNAs remained largely undetectable. In a validation cohort of acute myocardial infarction, receiver operating characteristic curve analysis revealed noninferiority of cardiac-enriched miRNAs, but miRNAs failed to identify cases presenting with low troponin values. cMyBP-C was validated as a biomarker with highly sensitive properties, and the combination of muscle-enriched miRNAs with high-sensitive cardiac troponin T and cMyBP-C returned the highest area under the curve values. CONCLUSIONS: In a comparative assessment of ncRNAs and protein biomarkers for myocardial injury, cMyBP-C showed properties as the most sensitive cardiac biomarker while miRNAs emerged as promising candidates to integrate ncRNAs with protein biomarkers. Sensitivity of current miRNA detection is inferior to cardiac proteins but a multibiomarker combination of muscle-enriched miRNAs with cMyBP-C and cardiac troponins could open a new path of integrating complementary characteristics of different biomarker types.


Assuntos
Biomarcadores/sangue , Cardiomiopatias/sangue , Proteínas de Transporte/sangue , RNA não Traduzido/sangue , Troponina T/sangue , Artefatos , Heparina , Heparina Liase/farmacologia , Humanos , MicroRNAs/sangue , Miocárdio/química , Plasma/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real
2.
Virol J ; 16(1): 134, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31718685

RESUMO

BACKGROUND: Duck tembusu virus (DTMUV, genus Flaviviruses, family Flaviviridae) is an emerging flavivirus that can infect a wide range of cells and cell lines in vitro, though the initial step of virus invasion remains obscure. METHODS: In this study, drug treatments that including heparin, chondroitin sulfate, heparinase I, chondroitinase ABC and trypsin were applied to detect the influence of DTMUV absorption, subsequently, the copy number of viral genome RNA was analyzed by quantitative real-time PCR. The inhibition process of viral absorption or entry by heparin was determined by western blotting, and the cytotoxicity of drug treated cells was detected by cell counting kit-8. RESULTS: We found that the desulfation of glycosaminoglycans (GAGs) with sodium chlorate had a significant effect on the adsorption of DTMUV in both BHK21 and DEF cells. Based on this result, we incubated cells with a mixture of DTMUV and GAGs competition inhibitors or pre-treated cells with inhibitors, after incubation with the virus, the NS5 expression of DTMUV and viral titers were detected. The data suggested that heparin can significantly inhibit the absorption of DTMUV in a dose dependent manner but not at the step of viral entry in BHK21 and DEF cells. Meanwhile, heparinase I can significantly inhibit DTMUV attachment step. CONCLUSIONS: Our results clearly proved that heparin sulfate plays an important role in the first step of DTMUV entry, viral attachment, in both BHK21 and DEF cells, which sheds light on the entry mechanism of DTMUV.


Assuntos
Flavivirus/fisiologia , Heparina/farmacologia , Ligação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Cloratos/farmacologia , Cricetinae , Patos , Fibrinolíticos/farmacologia , Flavivirus/efeitos dos fármacos , Glicosaminoglicanos/metabolismo , Heparina Liase/farmacologia , Tripsina/farmacologia
3.
Cell Microbiol ; 19(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28008697

RESUMO

Flagella are nanofibers that drive bacterial movement. The filaments are generally composed of thousands of tightly packed flagellin subunits with a terminal cap protein, named FliD. Here, we report that the FliD protein of the bacterial pathogen Campylobacter jejuni binds to host cells. Live-cell imaging and confocal microscopy showed initial contact of the bacteria with epithelial cells via the flagella tip. Recombinant FliD protein bound to the surface of intestinal epithelial cells in a dose-dependent fashion. Search for the FliD binding site on the host cell using cells with defined glycosylation defects indicated glycosaminoglycans as a putative target. Heparinase treatment of wild type cells and an excess of soluble heparin abolished FliD binding. Binding assays showed direct and specific binding of FliD to heparin. Addition of an excess of purified FliD or heparin reduced the attachment of viable C. jejuni to the host cells. The host cell binding domain of FliD was mapped to the central region of the protein. Overall, our results indicate that the C. jejuni flagellar tip protein FliD acts as an attachment factor that interacts with cell surface heparan sulfate glycosaminoglycan receptors.


Assuntos
Aderência Bacteriana/fisiologia , Proteínas de Bactérias/metabolismo , Campylobacter jejuni/metabolismo , Flagelos/metabolismo , Glicosaminoglicanos/metabolismo , Mucosa Intestinal/parasitologia , Animais , Aderência Bacteriana/efeitos dos fármacos , Proteínas de Bactérias/genética , Sítios de Ligação/fisiologia , Células CHO , Infecções por Campylobacter/microbiologia , Infecções por Campylobacter/patologia , Linhagem Celular Tumoral , Cricetulus , Células Epiteliais/citologia , Células Epiteliais/parasitologia , Flagelina/metabolismo , Células HT29 , Heparina Liase/farmacologia , Humanos , Mucosa Intestinal/citologia
4.
Cereb Cortex ; 27(2): 903-918, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28119345

RESUMO

Heparan sulfate (HS) proteoglycans represent a major component of the extracellular matrix and are critical for brain development. However, their function in the mature brain remains to be characterized. Here, acute enzymatic digestion of HS side chains was used to uncover how HSs support hippocampal function in vitro and in vivo. We found that long-term potentiation (LTP) of synaptic transmission at CA3-CA1 Schaffer collateral synapses was impaired after removal of highly sulfated HSs with heparinase 1. This reduction was associated with decreased Ca2+ influx during LTP induction, which was the consequence of a reduced excitability of CA1 pyramidal neurons. At the subcellular level, heparinase treatment resulted in reorganization of the distal axon initial segment, as detected by a reduction in ankyrin G expression. In vivo, digestion of HSs impaired context discrimination in a fear conditioning paradigm and oscillatory network activity in the low theta band after fear conditioning. Thus, HSs maintain neuronal excitability and, as a consequence, support synaptic plasticity and learning.


Assuntos
Discriminação Psicológica/fisiologia , Heparitina Sulfato/fisiologia , Plasticidade Neuronal/fisiologia , Células Piramidais/fisiologia , Sinapses/fisiologia , Animais , Anquirinas/biossíntese , Anquirinas/genética , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/fisiologia , Região CA3 Hipocampal/citologia , Região CA3 Hipocampal/fisiologia , Sinalização do Cálcio/fisiologia , Condicionamento Psicológico , Medo/fisiologia , Heparina Liase/farmacologia , Técnicas In Vitro , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/fisiologia , Ritmo Teta
5.
Biochim Biophys Acta ; 1858(8): 1860-7, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27117133

RESUMO

Cell-penetrating peptides (CPPs) are considered as one of the most promising tools to mediate the cellular delivery of various biologically active compounds that are otherwise cell impermeable. CPPs can internalize into cells via two different pathways - endocytosis and direct translocation across the plasma membrane. In both cases, the initial step of internalization requires interactions between CPPs and different plasma membrane components. Despite the extensive research, it is not yet fully understood, which of these cell surface molecules mediate the direct translocation of CPPs across the plasma- and endosomal membrane. In the present study we used giant plasma membrane vesicles (GPMVs) as a model membrane system to elucidate the specific molecular mechanisms behind the internalization and the role of cell surface glycosaminoglycans (GAGs) in the translocation of four well-known CPPs, classified as cationic (nona-arginine, Tat peptide) and amphipathic (transportan and TP10). We demonstrate here that GAGs facilitate the translocation of amphipathic CPPs, but not the internalization of cationic CPPs; and that the uptake is not mediated by a specific GAG class, but rather the overall amount of these polysaccharides is crucial for the internalization of amphipathic peptides.


Assuntos
Peptídeos Penetradores de Células/metabolismo , Glicosaminoglicanos/fisiologia , Vesículas Transportadoras/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Galanina/metabolismo , Heparina Liase/farmacologia , Humanos , Oligopeptídeos/metabolismo , Fragmentos de Peptídeos/metabolismo , Transporte Proteico , Receptores Adrenérgicos beta 1/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Vesículas Transportadoras/química , Venenos de Vespas/metabolismo , Aglutininas do Germe de Trigo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
6.
Am J Transplant ; 15(6): 1519-30, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25833002

RESUMO

Islet transplantation is a promising therapy for patients with diabetes, but its long-term success is limited by many factors, including the formation of islet amyloid deposits. Heparin is employed in clinical islet transplantation to reduce clotting but also promotes fibrillization of amyloidogenic proteins. We hypothesized that heparin treatment of islets during pre-transplant culture may enhance amyloid formation leading to beta cell loss and graft dysfunction. Heparin promoted the fibrillization of human islet amyloid polypeptide (IAPP) and enhanced its toxicity to INS-1 beta cells. Heparin increased amyloid deposition in cultured human islets, but surprisingly decreased islet cell apoptosis. Treatment of human islets with heparin prior to transplantation increased the likelihood of graft failure. Removal of islet heparan sulfate glycosaminoglycans, which localize with islet amyloid deposits in type 2 diabetes, by heparinase treatment decreased amyloid deposition and protected against islet cell death. These findings raise the possibility that pretransplant treatment of human islets with heparin could potentiate IAPP aggregation and amyloid formation and may be detrimental to subsequent graft function.


Assuntos
Amiloide/antagonistas & inibidores , Amiloide/metabolismo , Heparina Liase/farmacologia , Heparina/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Amiloide/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/cirurgia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Rejeição de Enxerto/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/métodos , Camundongos Endogâmicos NOD , Camundongos SCID , Estreptozocina/efeitos adversos
7.
FASEB J ; 28(8): 3589-99, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24784579

RESUMO

The pathogenesis of cancer is often driven by local invasion and metastasis. Recently, mechanical properties of the tumor microenvironment have been identified as potent regulators of invasion and metastasis, while matrix metalloproteinases (MMPs) are classically known as significant enhancers of cancer cell migration and invasion. Here we have been able to sensitively measure MMP activity changes in response to specific extracellular matrix (ECM) environments and cell contractility states. Cells of a pancreatic cancer cell line, Panc-1, up-regulate MMP activities between 3- and 10-fold with increased cell contractility. Conversely, they down-regulate MMP activities when contractility is blocked to levels seen with pan-MMP activity inhibitors. Similar, albeit attenuated, responses are seen in other pancreatic cancer cell lines, BxPC-3 and AsPC-1. In addition, MMP activity was modulated by substrate stiffness, collagen gel concentration, and the degree of collagen cross-linking, when cells were plated on collagen gels ranging from 0.5 to 5 mg/ml that span the physiological range of substrate stiffness (50-2000 Pa). Panc-1 cells showed enhanced MMP activity on stiffer substrates, whereas BxPC-3 and AsPC-1 cells showed diminished MMP activity. In addition, eliminating heparan sulfate proteoglycans using heparinase completely abrogated the mechanical induction of MMP activity. These results demonstrate the first functional link between MMP activity, contractility, and ECM stiffness and provide an explanation as to why stiffer environments result in enhanced cell migration and invasion.


Assuntos
Adenocarcinoma/enzimologia , Matriz Extracelular/fisiologia , Metaloproteinases da Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/enzimologia , Adenocarcinoma/patologia , Neoplasias da Mama/patologia , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Forma Celular , Colágeno , Elasticidade , Géis , Proteoglicanas de Heparan Sulfato/fisiologia , Heparina Liase/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Invasividade Neoplásica , Neoplasias Pancreáticas/patologia , Proteínas Recombinantes/metabolismo
8.
Eur J Pharmacol ; 960: 176138, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-37923158

RESUMO

Effective treatment strategies for skin wound repair are the focus of numerous studies. New pharmacological approaches appear necessary to guarantee a correct and healthy tissue regeneration. For these reasons, we purposed to investigate the effects of the combination between heparan sulfate and growth factors further adding the heparinase enzyme. Interestingly, for the first time, we have found that this whole association retains a marked pro-healing activity when topically administered to the wound. In detail, this combination significantly enhances the motility and activation of the main cell populations involved in tissue regeneration (keratinocytes, fibroblasts and endothelial cells), compared with single agents administered without heparinase. Notably, using an experimental C57BL/6 mouse model of skin wounding, we observed that the topical treatment of skin lesions with heparan sulfate + growth factors + heparinase promotes the highest closure of wounds compared to each substance mixed with the other ones in all the possible combinations. Eosin/hematoxylin staining of skin biopsies revealed that treatment with the whole combination allows the formation of a well-structured matrix with numerous new vessels. Confocal analyses for vimentin, FAP1α, CK10 and CD31 have highlighted the presence of activated fibroblasts, differentiated keratinocytes and endothelial cells at the closed region of wounds. Our results encourage defining this combined treatment as a new and appealing therapy expedient in skin wound healing, as it is able to activate cell components and promote a dynamic lesions closure.


Assuntos
Células Endoteliais , Pele , Camundongos , Animais , Heparina Liase/farmacologia , Camundongos Endogâmicos C57BL , Cicatrização , Heparitina Sulfato/farmacologia
9.
Cells ; 12(5)2023 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-36899880

RESUMO

Our previous studies demonstrated that enzymatic removal of highly sulfated heparan sulfates with heparinase 1 impaired axonal excitability and reduced expression of ankyrin G at the axon initial segments in the CA1 region of the hippocampus ex vivo, impaired context discrimination in vivo, and increased Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity in vitro. Here, we show that in vivo delivery of heparinase 1 in the CA1 region of the hippocampus elevated autophosphorylation of CaMKII 24 h after injection in mice. Patch clamp recording in CA1 neurons revealed no significant heparinase effects on the amplitude or frequency of miniature excitatory and inhibitory postsynaptic currents, while the threshold for action potential generation was increased and fewer spikes were generated in response to current injection. Delivery of heparinase on the next day after contextual fear conditioning induced context overgeneralization 24 h after injection. Co-administration of heparinase with the CaMKII inhibitor (autocamtide-2-related inhibitory peptide) rescued neuronal excitability and expression of ankyrin G at the axon initial segment. It also restored context discrimination, suggesting the key role of CaMKII in neuronal signaling downstream of heparan sulfate proteoglycans and highlighting a link between impaired CA1 pyramidal cell excitability and context generalization during recall of contextual memories.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Heparitina Sulfato , Animais , Camundongos , Anquirinas/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Heparina Liase/metabolismo , Heparina Liase/farmacologia , Heparitina Sulfato/metabolismo , Hipocampo/metabolismo
10.
J Biol Chem ; 286(7): 5359-67, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21135110

RESUMO

Properdin binds to proximal tubular epithelial cells (PTEC) and activates the complement system via the alternative pathway in vitro. Cellular ligands for properdin in the kidney have not yet been identified. Because properdin interacts with solid-phase heparin, we investigated whether heparan sulfate proteoglycans (HSPG) could be the physiological ligands of properdin. Kidneys from proteinuric rats showed colocalization of syndecan-1, a major epithelial HSPG, and properdin in the apical membranes of PTEC, which was not seen in control renal tissue. In vitro, PTEC did not constitutively express properdin. However, exogenous properdin binds to these cells in a dose-dependent fashion. Properdin binding was prevented by heparitinase pretreatment of the cells and was dose-dependently inhibited by exogenous heparin. ELISA and surface plasmon resonance spectroscopy (BIAcore) showed a strong dose-dependent interaction between heparan sulfate (HS) and properdin (K(d) = 128 nm). Pretreatment of HSPG with heparitinase abolished this interaction in ELISA. Competition assays, using a library of HS-like polysaccharides, showed that sulfation pattern, chain length, and backbone composition determine the interaction of properdin with glycosaminoglycans. Interestingly, two nonanticoagulant heparin derivatives inhibited properdin-HS interaction in ELISA and BIAcore. Incubation of PTEC with human serum as complement source led to complement activation and deposition of C3 on the cells. This C3 deposition is dependent on the binding of properdin to HS as shown by heparitinase pretreatment of the cells. Our data identify tubular HS as a novel docking platform for alternative pathway activation via properdin, which might play a role in proteinuric renal damage. Our study also suggests nonanticoagulant heparinoids may provide renoprotection in complement-dependent renal diseases.


Assuntos
Células Epiteliais/metabolismo , Heparitina Sulfato/metabolismo , Nefropatias/metabolismo , Túbulos Renais Proximais/metabolismo , Properdina/metabolismo , Proteinúria/metabolismo , Animais , Complemento C3/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Células Epiteliais/patologia , Heparina Liase/farmacologia , Humanos , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Nefropatias/patologia , Túbulos Renais Proximais/patologia , Masculino , Properdina/farmacologia , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Wistar
11.
Pflugers Arch ; 463(2): 269-78, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22057584

RESUMO

Plasma sodium, slightly above normal and in presence of aldosterone, stiffens vascular endothelium and reduces nitric oxide release with the consequence of endothelial dysfunction. This process is mediated by epithelial sodium channels (ENaC) and, most likely, the endothelial Na(+)/K(+)-ATPase. Both, ENaC and Na(+)/K(+)-ATPase, are located in the plasma membrane of endothelial cells and embedded in the endothelial glycocalyx (eGC). This negatively charged biopolymer is directly exposed to the blood stream and selectively buffers sodium ions. We hypothesize that the glycocalyx could interfere with endothelial sodium transport when extracellular sodium varies in the physiological range. Therefore, we modeled the endothelial cell as a pump-leak system measuring changes of intracellular sodium in cultured human endothelial cells. Experiments were performed under low/high extracellular sodium conditions before and after enzymatic eGC removal, and with inhibition of Na(+)/K(+)-ATPase and ENaC, respectively. Three major observations were made: (1) eGC removal by heparinase treatment facilitates sodium to enter/exit the endothelial cells. (2) The direction of net sodium movement across the endothelial plasma membrane depends on the concentration of extracellular sodium which regulates both the Na(+)/K(+)-ATPase and ENaC activity. (3) Removal of eGC and inhibition of sodium transport modify the electrical resistance of endothelial cells. We conclude that the eGC serves as a potential "firewall" preventing uncontrolled access of sodium to the pump-leak system of the endothelial cell. After eGC removal, sodium access to the system is facilitated. Thus the pump-leak system could be regulated by ambient sodium and control vascular permeability in pathophysiological conditions.


Assuntos
Endotélio Vascular/fisiologia , Glicocálix/fisiologia , Homeostase/fisiologia , Sódio/metabolismo , Transporte Biológico/fisiologia , Linhagem Celular , Permeabilidade da Membrana Celular/fisiologia , Endotélio Vascular/citologia , Canais Epiteliais de Sódio/fisiologia , Glicocálix/efeitos dos fármacos , Heparina Liase/farmacologia , Humanos , ATPase Trocadora de Sódio-Potássio/fisiologia
12.
Amino Acids ; 43(1): 365-78, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21952731

RESUMO

Several sporozoite proteins have been associated with Plasmodium falciparum cell traversal and hepatocyte invasion, including the cell-traversal protein for ookinetes and sporozoites (CelTOS), and thrombospondin-related sporozoite protein (TRSP). CelTOS and TRSP amino acid sequences have been finely mapped to identify regions specifically binding to HeLa and HepG2 cells, respectively. Three high-activity binding peptides (HABPs) were found in CelTOS and one HABP was found in TRSP, all of them having high α-helical structure content. These HABPs' specific binding was sensitive to HeLa and HepG2 cells' pre-treatment with heparinase I and chondroitinase ABC. Despite their similarity at three-dimensional (3D) structural level, TRSP and TRAP HABPs located in the TSR domain did not compete for the same binding sites. CelTOS and TRSP HABPs were used as a template for designing modified sequences to then be assessed in the Aotus monkey experimental model. Antibodies directed against these modified HABPs were able to recognize both the native parasite protein by immunofluorescence assay and the recombinant protein (expressed in Escherichia coli) by Western blot and ELISA assays. The results suggested that these modified HABPs could be promising targets in designing a fully effective, antimalarial vaccine.


Assuntos
Plasmodium falciparum/imunologia , Proteínas de Protozoários , Trombospondinas , Sequência de Aminoácidos , Animais , Aotus trivirgatus , Sítios de Ligação , Linhagem Celular Tumoral , Condroitina ABC Liase/farmacologia , Células HeLa , Células Hep G2 , Heparina Liase/farmacologia , Hepatócitos/imunologia , Hepatócitos/metabolismo , Hepatócitos/parasitologia , Humanos , Vacinas Antimaláricas/imunologia , Peptídeos/análise , Peptídeos/imunologia , Peptídeos/isolamento & purificação , Plasmodium falciparum/citologia , Plasmodium falciparum/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/isolamento & purificação , Proteínas Recombinantes/síntese química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Esporozoítos/citologia , Esporozoítos/imunologia , Esporozoítos/metabolismo , Trombospondinas/química , Trombospondinas/imunologia , Trombospondinas/isolamento & purificação
13.
Arterioscler Thromb Vasc Biol ; 31(12): 2972-4, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21979436

RESUMO

OBJECTIVE: Soluble fms-like tyrosine kinase 1 (sFlt1) is involved in the pathophysiology of preeclampsia and coronary artery disease. Because sFlt1 has a heparin-binding site, we investigated whether or not heparin releases sFlt1 from the extracellular matrix. METHODS AND RESULTS: We measured sFlt1 before and after heparin administration in 135 patients undergoing coronary angiography, percutanous coronary intervention, or both. sFlt1 was increased directly after heparin administration (from 254 to 13,440 pg/mL) and returned to baseline within 10 hours. Umbilical veins and endothelial cells treated with heparin released sFlt1. Heparinase I and III also increased sFlt1. Mice treated with heparin had elevated sFlt1 serum levels. Their serum inhibited endothelial tube formation. CONCLUSIONS: Heparin releases sFlt1 by displacing the sFlt1 heparin-binding site from heparan sulfate proteoglycans. Heparin could induce an antiangiogenic state.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Fibrinolíticos/farmacologia , Heparina/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue , Angioplastia Coronária com Balão , Animais , Células Cultivadas , Angiografia Coronária , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/terapia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Matriz Extracelular/metabolismo , Fibrinolíticos/administração & dosagem , Heparina/administração & dosagem , Heparina Liase/farmacologia , Humanos , Técnicas In Vitro , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Polissacarídeo-Liases/farmacologia
14.
Inflammation ; 45(2): 712-724, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34657233

RESUMO

Endothelial glycocalyx degradation is thought to facilitate the development of sepsis. Histone is a significant mediator in sepsis. Unfractionated heparin (UFH) possessed beneficial effects on sepsis. Thereby, this study aims to figure out whether histone can disrupt glycocalyx and to investigate the protective effect and mechanism of UFH. Male mice (C57BL/6, 8-10 weeks old, weighing 20-25 g) were randomly divided into five groups including control group, histone group, histone + UFH group, histone + heparinase (HPA) inhibitor group, and histone + UFH + HPA inhibitor group. The mice were treated with histone (50 mg/kg) via tail vein immediately after HPA (20 mg/kg) injection. UFH (400 U/kg) was injected 1h after histone administration. The other groups were injected with equal volume of sterile saline accordingly. UFH alleviated histone-induced lung injury and pulmonary edema. UFH inhibited histone-induced lung coagulation activation and inflammatory response. UFH treatment markedly inhibited pulmonary glycocalyx degradation by reducing the histone-induced decrease in the levels of lung syndecan-1 mRNA and protein. UFH downregulated histone-induced expression of HPA mRNA and protein, and thus alleviated glycocalyx degradation. UFH protects against histone-induced pulmonary glycocalyx injury partly by heparinase pathway.


Assuntos
Heparina , Histonas , Animais , Heparina/farmacologia , Heparina/uso terapêutico , Heparina Liase/farmacologia , Histonas/farmacologia , Pulmão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sindecana-1
15.
Viruses ; 13(9)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34578329

RESUMO

Heparan sulfate (HS) and heparan sulfate proteoglycans (HSPGs) are considered important for the entry of many different viruses. Previously, we demonstrated that heparanase (HPSE), the host enzyme responsible for cleaving HS chains, is upregulated by herpes simplex virus-1 (HSV-1) infection. Higher levels of HPSE accelerate HS removal from the cell surface, facilitating viral release from infected cells. Here, we study the effects of overexpressing HPSE on viral entry, cell-to-cell fusion, plaque formation, and viral egress. We provide new information that higher levels of HPSE reduce syncytial plaque formation while promoting egress and extracellular release of the virions. We also found that transiently enhanced expression of HPSE did not affect HSV-1 entry into host cells or HSV-1-induced cell-to-cell fusion, suggesting that HPSE activation is tightly regulated and facilitates extracellular release of the maturing virions. We demonstrate that an HSPG-shedding agonist, PMA; a protease, thrombin; and a growth factor, EGF as well as bacterially produced recombinant heparinases resulted in enhanced HSV-1 release from HeLa and human corneal epithelial (HCE) cells. Our findings here underscore the significance of syndecan-1 functions in the HSV-1 lifecycle, provide evidence that the shedding of syndecan-1 ectodomain is another way HPSE works to facilitate HSV-1 release, and add new evidence on the significance of various HSPG shedding agonists in HSV-1 release from infected cells.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Heparina Liase/farmacologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/fisiologia , Sindecana-1/genética , Trombina/farmacologia , Liberação de Vírus/efeitos dos fármacos , Córnea/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Células HeLa , Humanos , Sindecana-1/metabolismo , Regulação para Cima , Vírion/efeitos dos fármacos , Vírion/metabolismo , Internalização do Vírus
16.
Immunology ; 131(3): 357-70, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20518822

RESUMO

Stromal fibroblasts modify the initial recruitment of leucocytes by endothelial cells (EC), but their effects on subsequent transendothelial migration remain unclear. Here, EC and dermal or synovial fibroblasts were cultured on opposite surfaces of 3-µm pore filters and incorporated in static or flow-based migration assays. Fibroblasts had little effect on tumour necrosis factor-α-induced transendothelial migration of neutrophils, but tended to increase the efficiency of migration away from the endothelium. Surprisingly, similar close contact between EC and fibroblasts strongly reduced lymphocyte migration in static assays, and nearly abolished stable lymphocyte adhesion from flow. Fibroblasts did not alter endothelial surface expression of adhesion molecules or messenger RNA for chemokines. Inhibition of attachment did not occur when EC-fibroblast contact was restricted by using 0.4-µm pore filters, but under these conditions pre-treatment with heparinase partially inhibited adhesion. In the 3-µm pore co-cultures, inhibition of metalloproteinase activity partially recovered lymphocyte adhesion, but addition of CXCL12 (SDF-1α) to the endothelial surface did not. Hence, the ability of EC to present activating chemokines for lymphocytes may have been enzymatically inhibited by direct contact with fibroblasts. To avoid contact, we cultured EC and fibroblasts on separate 3-µm pore filters one above the other. Here, fibroblasts promoted the transendothelial migration of lymphocytes. Fibroblasts generate CXCL12, but blockade of CXCL12 receptor had no effect on lymphocyte migration. While stromal cells can provide signal(s) promoting leucocyte migration away from the sub-endothelial space, direct cell contact (which might occur in damaged tissue) may cause disruption of chemokine signalling, specifically inhibiting lymphocyte rather than neutrophil recruitment.


Assuntos
Adesão Celular , Movimento Celular , Endotélio/metabolismo , Fibroblastos/metabolismo , Linfócitos/metabolismo , Adesão Celular/efeitos dos fármacos , Comunicação Celular , Ensaios de Migração Celular , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CXCL12/biossíntese , Quimiocina CXCL12/genética , Quimiocina CXCL12/farmacologia , Técnicas de Cocultura , Endotélio/efeitos dos fármacos , Endotélio/imunologia , Endotélio/patologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/patologia , Heparina Liase/farmacologia , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/patologia , Pele/patologia , Membrana Sinovial/patologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
17.
J Cell Biochem ; 110(1): 70-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20213742

RESUMO

Accumulating evidences point to a significant role for the chromogranin A (CgA)-derived peptide vasostatin 1 (VS-1) in the protective modulation of the cardiovascular activity, because of its ability to counteract the adrenergic signal. We have recently shown that VS-1 induces a PI3K-dependent-nitric oxide (NO) release by endothelial cells, contributing to explain the mechanism of its cardio-suppressive and vasodilator properties. However, the cellular processes upstream the eNOS activation exerted by this peptide are still unknown, as typical high-affinity receptors have not been identified. Here we hypothesize that in endothelial cells VS-1 acts, on the basis of its cationic and amphipathic properties, as a cell penetrating peptide, binding to heparan sulfate proteoglycans (HSPGs) and activating eNOS phosphorylation (Ser1179) through a PI3K-dependent, endocytosis-coupled mechanism. In bovine aortic endothelial cells (BAE-1 cells) endocytotic vesicles trafficking was quantified by confocal microscopy with a water-soluble membrane dye; caveolin 1 (Cav1) shift from plasma membrane was studied by immunofluorescence staining; VS-1-dependent eNOS phosphorylation was assessed by immunofluorescence and immunoblot analysis. Our experiments demonstrate that VS-1 induces a marked increase in the caveolae-dependent endocytosis, (115 +/- 23% endocytotic spots/cell/field in VS-1-treated cells with respect to control cells), that is significantly reduced by both heparinase III (HEP, 17 +/- 15% above control) and Wortmannin (Wm, 7 +/- 22% above control). Heparinase, Wortmannin, and methyl-beta-cyclodextrin (MbetaCD) abolish the VS-1-dependent eNOS phosphorylation (P(Ser1179)eNOS). These results suggest a novel signal transduction pathway for endogenous cationic and amphipathic peptides in endothelial cells: HSPGs interaction and caveolae endocytosis, coupled with a PI3K-dependent eNOS phosphorylation.


Assuntos
Cromogranina A/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteoglicanas/metabolismo , Androstadienos/farmacologia , Animais , Bovinos , Cavéolas/efeitos dos fármacos , Cavéolas/metabolismo , Caveolina 1/metabolismo , Endocitose/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Heparina Liase/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Vesículas Transportadoras/efeitos dos fármacos , Vesículas Transportadoras/metabolismo , Wortmanina
18.
Thromb Haemost ; 120(8): 1173-1181, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32604425

RESUMO

BACKGROUND: Portal vein thrombosis (PVT) is the most common thrombotic complication in cirrhosis; however, local risk factors involved in its pathogenesis are still not fully investigated. The aim of the study was to evaluate hemostasis and endothelial damage in the portal vein in patients with cirrhosis and portal hypertension. METHODS: Adult cirrhotics undergoing transjugular intrahepatic portosystemic shunt were consecutively enrolled. Rotational thromboelastometry (ROTEM), dosage of total circulating glycosaminoglycans (GAGs), and endotoxemia levels (lipopolysaccharide [LPS]), along with evaluation of endothelial dysfunction by quantification of circulating endothelial microparticles (MPs), were performed on citrated peripheric and portal venous blood samples from each enrolled patient. RESULTS: Forty-five cirrhotics were enrolled. ROTEM analysis revealed the presence of a significant heparin-like effect in portal blood (median ɑ angle NATEM 50° vs. HEPTEM 55°, p = 0.027; median coagulation time NATEM 665 s vs. HEPTEM 585 s, p = 0.006), which was not detected in peripheral blood, and was associated with a higher concentration of circulating GAGs. Even though total annexin V-MP circulating MPs were less concentrated in the splanchnic district, the proportion of MPs of endothelial origin, with respect to annexin V-MP, was significantly increased in the portal district (p = 0.036). LPS concentration was higher in portal (197 pg/mL) compared with peripheral blood (165 pg/mL) (p < 0.001). CONCLUSION: Evidences of a damage of glycocalyx along with increased concentration of endothelial MPs suggest the presence of a significant endothelial alteration in the portal vein with respect to peripheral veins. Portal site-specific endothelial damage could hamper its antithrombotic properties and may represent an important local risk factor in the pathogenesis of PVT.


Assuntos
Endotélio Vascular/patologia , Cirrose Hepática/patologia , Veia Porta/patologia , Adulto , Coagulação Sanguínea , Micropartículas Derivadas de Células , Endotélio Vascular/fisiopatologia , Endotoxemia/sangue , Feminino , Glicocálix/patologia , Glicosaminoglicanos/sangue , Heparina Liase/farmacologia , Humanos , Hipertensão Portal/etiologia , Hipertensão Portal/cirurgia , Masculino , Pessoa de Meia-Idade , Derivação Portossistêmica Cirúrgica , Tromboelastografia , Trombose Venosa/prevenção & controle
19.
Cancer Lett ; 492: 71-83, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32860852

RESUMO

The blood vessel growth inhibitor bevacizumab targets vascular endothelial growth factor (VEGF), a crucial regulator of angiogenesis. Recently, small extracellular vesicles (sEVs) have been demonstrated to be important vehicles in the transport of growth factors to target cells. In this study, we isolated primary carcinoma-associated fibroblasts (CAFs) from four human oral squamous cell carcinoma (OSCC) specimens. Compared with other non-extracellular vesicle components, CAF-derived sEVs were found to be the main regulators of angiogenesis. The ability of CAF sEVs to activate VEGF receptor 2 (VEGFR2) signaling in human umbilical vein endothelial cells (HUVEC) was dependent on the association between sEVs and VEGF. In addition, sEV-bound VEGF secreted by CAFs further activated VEGFR2 signaling in HUVEC in a bevacizumab-resistant manner. VEGF was found to interact with heparan sulfate proteoglycans on the CAF sEV surface and could be released by heparinase I/III. The bioactivity of the dissociated VEGF was retained in vitro and in vivo and could be neutralized by bevacizumab. These findings suggest that the combined use of heparinase and bevacizumab might inhibit angiogenesis in patients with high levels of sEV-bound VEGF.


Assuntos
Bevacizumab/uso terapêutico , Fibroblastos Associados a Câncer/fisiologia , Vesículas Extracelulares/fisiologia , Neoplasias Bucais/irrigação sanguínea , Neovascularização Patológica/etiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/fisiologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Heparina Liase/farmacologia , Humanos , Neoplasias Bucais/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia
20.
Stem Cells ; 25(11): 2845-54, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17702986

RESUMO

Cell surface heparan sulfate (HS) and chondroitin sulfate (CS) proteoglycans have been implicated in a multitude of biological processes, including embryonic implantation, tissue morphogenesis, wound repair, and neovascularization through their ability to regulate growth factor activity and morphogenic gradients. However, the direct role of the glycosaminoglycan (GAG) sugar-side chains in the control of human mesenchymal stem cell (hMSC) differentiation into the osteoblast lineage is poorly understood. Here, we show that the abundant cell surface GAGs, HS and CS, are secreted in proteoglycan complexes that directly regulate the bone morphogenetic protein (BMP)-mediated differentiation of hMSCs into osteoblasts. Enzymatic depletion of the HS and CS chains by heparinase and chondroitinase treatment decreased HS and CS expression but did not alter the expression of the HS core proteins perlecan and syndecan. When digested separately, depletion of HS and CS chains did not effect hMSC proliferation but rather increased BMP bioactivity through SMAD1/5/8 intracellular signaling at the same time as increasing canonical Wnt signaling through LEF1 activation. Long-term culturing of cells in HS- and CS-degrading enzymes also increased bone nodule formation, calcium accumulation, and the expression of such osteoblast markers as alkaline phosphatase, RUNX2, and osteocalcin. Thus, the enzymatic disruption of HS and CS chains on cell surface proteoglycans alters BMP and Wnt activity so as to enhance the lineage commitment and osteogenic differentiation of hMSCs.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/fisiologia , Sulfatos de Condroitina/fisiologia , Heparitina Sulfato/fisiologia , Células-Tronco Mesenquimais/fisiologia , Osteogênese/fisiologia , Diferenciação Celular/efeitos dos fármacos , Sulfatos de Condroitina/antagonistas & inibidores , Condroitinases e Condroitina Liases/farmacologia , Heparina Liase/farmacologia , Heparitina Sulfato/antagonistas & inibidores , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA