Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Acta Physiol (Oxf) ; 240(4): e14116, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38400621

RESUMO

AIM: Protein disulfide isomerases (PDIs) are involved in platelet aggregation and intravascular thrombosis, but their role in regulating endothelial function is unclear. Here, we characterized the involvement of vascular PDIA1 in angiotensin II (Ang II)-induced endothelial dysfunction in mice. METHODS: Endothelial dysfunction was induced in C57BL/6JCmd male mice via Ang II subcutaneous infusion, and PDIA1 was inhibited with bepristat. Endothelial function was assessed in vivo with magnetic resonance imaging and ex vivo with a myography, while arterial stiffness was measured as pulse wave velocity. Nitric oxide (NO) bioavailability was measured in the aorta (spin-trapping electron paramagnetic resonance) and plasma (NO2 - and NO3 - levels). Oxidative stress, eNOS uncoupling (DHE-based aorta staining), and thrombin activity (thrombin-antithrombin complex; calibrated automated thrombography) were evaluated. RESULTS: The inhibition of PDIA1 by bepristat in Ang II-treated mice prevented the impairment of NO-dependent vasodilation in the aorta as evidenced by the response to acetylcholine in vivo, increased systemic NO bioavailability and the aortic NO production, and decreased vascular stiffness. Bepristat's effect on NO-dependent function was recapitulated ex vivo in Ang II-induced endothelial dysfunction in isolated aorta. Furthermore, bepristat diminished the Ang II-induced eNOS uncoupling and overproduction of ROS without affecting thrombin activity. CONCLUSION: In Ang II-treated mice, the inhibition of PDIA1 normalized the NO-ROS balance, prevented endothelial eNOS uncoupling, and, thereby, improved vascular function. These results indicate the importance of vascular PDIA1 in regulating endothelial function, but further studies are needed to elucidate the details of the mechanisms involved.


Assuntos
Angiotensina II , Doenças Vasculares , Camundongos , Masculino , Animais , Angiotensina II/farmacologia , Angiotensina II/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Isomerases de Dissulfetos de Proteínas/farmacologia , Análise de Onda de Pulso , Trombina/metabolismo , Trombina/farmacologia , Camundongos Endogâmicos C57BL , Doenças Vasculares/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Endotélio Vascular , Óxido Nítrico/metabolismo
2.
Breast Cancer Res ; 26(1): 1, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-38167446

RESUMO

BACKGROUND: Despite radiotherapy ability to significantly improve treatment outcomes and survival in triple-negative breast cancer (TNBC) patients, acquired resistance to radiotherapy poses a serious clinical challenge. Protein disulfide isomerase exists in endoplasmic reticulum and plays an important role in promoting protein folding and post-translational modification. However, little is known about the role of protein disulfide isomerase family member 4 (PDIA4) in TNBC, especially in the context of radiotherapy resistance. METHODS: We detected the presence of PDIA4 in TNBC tissues and paracancerous tissues, then examined the proliferation and apoptosis of TNBC cells with/without radiotherapy. As part of the validation process, xenograft tumor mouse model was used. Mass spectrometry and western blot analysis were used to identify PDIA4-mediated molecular signaling pathway. RESULTS: Based on paired clinical specimens of TNBC patients, we found that PDIA4 expression was significantly higher in tumor tissues compared to adjacent normal tissues. In vitro, PDIA4 knockdown not only increased apoptosis of tumor cells with/without radiotherapy, but also decreased the ability of proliferation. In contrast, overexpression of PDIA4 induced the opposite effects on apoptosis and proliferation. According to Co-IP/MS results, PDIA4 prevented Tax1 binding protein 1 (TAX1BP1) degradation by binding to TAX1BP1, which inhibited c-Jun N-terminal kinase (JNK) activation. Moreover, PDIA4 knockdown suppressed tumor growth xenograft model in vivo, which was accompanied by an increase in apoptosis and promoted tumor growth inhibition after radiotherapy. CONCLUSIONS: The results of this study indicate that PDIA4 is an oncoprotein that promotes TNBC progression, and targeted therapy may represent a new and effective anti-tumor strategy, especially for patients with radiotherapy resistance.


Assuntos
Sistema de Sinalização das MAP Quinases , Neoplasias de Mama Triplo Negativas , Humanos , Animais , Camundongos , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Isomerases de Dissulfetos de Proteínas/farmacologia , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/radioterapia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Carcinogênese , Transformação Celular Neoplásica , Família , Linhagem Celular Tumoral , Proliferação de Células
3.
Cell Mol Biol (Noisy-le-grand) ; 69(9): 207-212, 2023 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-37807312

RESUMO

The purpose of this study arose to investigate the mechanism of miR-204-5p targeting P4HB to regulate inflammation and apoptosis in HUVEC cells. Serum specimens were obtained from lower extremity DVT patients and healthy subjects. Targetscan predicted P4HB as a target gene for miR-204-5p. A dual luciferase reporter assay was conducted to determine the modulating effect of miR-204-5p on P4HB. qRT-PCR was used to detect miR-204-5p and P4HB expression. Established CoCl2-induced hypoxia/ischemia model of HUVEC, transfected with miR-204-5p mimics and pcDNA3. 1-P4HB. CCK-8 assay for cell viability. Apoptosis was assayed by flow cytometry, western blot and western blot. Immunofluorescence and ELISA were carried out to detect ROS, MDA, SOD, LDH, GSH-px, TNF-α, IL-1ß and IL-6 expression. miR-204-5p was reduced markedly in the sera of DVT patients. miR-204-5p negatively regulated P4HB. P4HB expression was raised in the sera of DVT patients. Exposure to CoCl2 decreased miR-204-5p expression and increased P4HB in HUVEC. Over-expressed miR-204-5p effectively increased cell viability and inhibited apoptosis; its effect was counteracted by continued overexpression of P4HB. In addition, miR- 204-5p mimics clearly reduced CoCl2-induced ROS and inflammation, and pcDNA3. 1-P4HB acted counteractively. miR-204-5p may inhibit HUVEC proliferation, ROS generation and cellular inflammation through negative regulation of P4HB. miR-204-5p promises to become a potential target for DVT therapy.


Assuntos
Apoptose , Inflamação , MicroRNAs , Humanos , Cobalto/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inflamação/metabolismo , MicroRNAs/metabolismo , Pró-Colágeno-Prolina Dioxigenase/farmacologia , Isomerases de Dissulfetos de Proteínas/farmacologia , Espécies Reativas de Oxigênio
4.
Antiviral Res ; 212: 105560, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36822370

RESUMO

Venezuelan equine encephalitis virus (VEEV) is an alphavirus transmitted by mosquitos that can cause a febrile illness and induce severe neurological complications in humans and equine populations. Currently there are no FDA approved vaccines or antiviral treatments to combat VEEV. Proteomic techniques were utilized to create an interactome of the E1 fusion glycoprotein of VEEV. VEEV E1 interacted with a number of cellular chaperone proteins including protein disulfide isomerase family A member 6 (PDIA6). PDI inhibition through LOC14 and/or nitazoxanide treatment effectively decreased production of VEEV and other alphaviruses in vitro, including eastern equine encephalitis virus, Sindbis virus, and chikungunya virus. Decreased oxidoreductive capabilities of PDIs through LOC14 or nitazoxanide treatment impacted both early and late events in viral replication, including the production of non-infectious virions and decreased VEEV E1 disulfide bond formation. Results from this study identified PDIs as critical regulators of alphavirus replication and potential therapeutic targets.


Assuntos
Alphavirus , Vírus Chikungunya , Vírus da Encefalite Equina Venezuelana , Encefalomielite Equina Venezuelana , Humanos , Animais , Cavalos , Proteômica , Linhagem Celular , Replicação Viral , Encefalomielite Equina Venezuelana/tratamento farmacológico , Isomerases de Dissulfetos de Proteínas/farmacologia , Isomerases de Dissulfetos de Proteínas/uso terapêutico
5.
Am J Physiol Cell Physiol ; 324(1): C113-C132, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36374169

RESUMO

Metastasis and recurrence of breast cancer remain major causes of patient mortality, and there is an ongoing need to identify new therapeutic targets relevant to tumor invasion. Protein disulfide isomerase A3 (PDIA3) is a disulfide oxidoreductase and isomerase of the endoplasmic reticulum that has known extracellular substrates and has been correlated with aggressive breast cancers. We show that either prior PDIA3 inhibition by the disulfide isomerase inhibitor 16F16 or depletion of heparin-binding proteins strongly reduces the activity of conditioned medium (CM) of MDA-MB-231 human breast cancer cells to support promigratory cell spreading and F-actin organization by newly adherent MDA-MB-231 cells. Quantitative proteomics to investigate effects of 16F16 inhibition on heparin-binding proteins in the CM of MDA-MB-231 cells identified 80 proteins reproducibly decreased at least twofold (at q ≤ 0.05) after 16F16 treatment. By Gene Ontology analysis, many of these have roles in extracellular matrix (ECM) structure and function and cell adhesion; ribosomal proteins that also correlate with extracellular vesicles were also identified. Protein-protein interaction analysis showed that many of the extracellular proteins have known network interactions with each other. The predominant types of disulfide-bonded domains in the extracellular proteins contained ß-hairpin folds, with the knottin fold the most common. From human breast cancer data sets, the extracellular proteins were found to correlate specifically with the basal subtype of breast cancer and their high expression in tumors correlated with reduced distant metastasis-free survival. These data provide new evidence that PDIA3 may be a relevant therapeutic target to alter properties of the ECM-associated microenvironment in basal breast cancer.


Assuntos
Neoplasias da Mama , Isomerases de Dissulfetos de Proteínas , Humanos , Feminino , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Isomerases de Dissulfetos de Proteínas/farmacologia , Neoplasias da Mama/patologia , Adesão Celular , Comunicação Celular , Heparina/farmacologia , Linhagem Celular Tumoral , Microambiente Tumoral
6.
Shock ; 58(6): 556-564, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36374735

RESUMO

ABSTRACT: Background: Dexmedetomidine (DEX) attenuates intestinal I/R injury, but its mechanism of action remains to be further elucidated. Protein disulfide isomerase A3 (PDIA3) has been reported as a therapeutic protein for the prevention and treatment of intestinal I/R injury. This study was to investigate whether PDIA3 is involved in intestinal protection of DEX and explore the underlying mechanisms. Methods: The potential involvement of PDIA3 in DEX attenuation of intestinal I/R injury was tested in PDIA3 Flox/Flox mice and PDIA3 conditional knockout (cKO) in intestinal epithelium mice subjected to 45 min of superior mesenteric artery occlusion followed by 4 h of reperfusion. Furthermore, the α2-adrenergic receptor (α2-AR) antagonist, yohimbine, was administered in wild-type C57BL/6N mice intestinal I/R model to investigate the role of α2-AR in the intestinal protection conferred by DEX. Results: In the present study, we identified intestinal I/R-induced obvious inflammation, endoplasmic reticulum (ER) stress-dependent apoptosis, and oxidative stress, and all the aforementioned changes were improved by the administration of DEX. PDIA3 cKO in the intestinal epithelium have reversed the protective effects of DEX. Moreover, yohimbine also reversed the intestinal protection of DEX and downregulated the messenger RNA and protein levels of PDIA3. Conclusion: DEX prevents PDIA3 decrease by activating α2-AR to inhibit intestinal I/R-induced inflammation, ER stress-dependent apoptosis, and oxidative stress in mice.


Assuntos
Dexmedetomidina , Animais , Camundongos , Dexmedetomidina/farmacologia , Dexmedetomidina/uso terapêutico , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/farmacologia , Camundongos Endogâmicos C57BL , Apoptose , Ioimbina/farmacologia , Inflamação/tratamento farmacológico
7.
Sci Rep ; 11(1): 17557, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34475430

RESUMO

Mutations in Fused in Sarcoma (FUS) are present in familial and sporadic cases of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). FUS is localised in the nucleus where it has important functions in DNA repair. However, in ALS/FTD, mutant FUS mislocalises from the nucleus to the cytoplasm where it forms inclusions, a key pathological hallmark of neurodegeneration. Mutant FUS also inhibits protein import into the nucleus, resulting in defects in nucleocytoplasmic transport. Fragmentation of the neuronal Golgi apparatus, induction of endoplasmic reticulum (ER) stress, and inhibition of ER-Golgi trafficking are also associated with mutant FUS misfolding in ALS. Protein disulphide isomerase (PDI) is an ER chaperone previously shown to be protective against misfolding associated with mutant superoxide dismutase 1 (SOD1) and TAR DNA-binding protein-43 (TDP-43) in cellular and zebrafish models. However, a protective role against mutant FUS in ALS has not been previously described. In this study, we demonstrate that PDI is protective against mutant FUS. In neuronal cell line and primary cultures, PDI restores defects in nuclear import, prevents the formation of mutant FUS inclusions, inhibits Golgi fragmentation, ER stress, ER-Golgi transport defects, and apoptosis. These findings imply that PDI is a new therapeutic target in FUS-associated ALS.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Estresse do Retículo Endoplasmático , Demência Frontotemporal/tratamento farmacológico , Mutação , Pró-Colágeno-Prolina Dioxigenase/farmacologia , Isomerases de Dissulfetos de Proteínas/farmacologia , Proteína FUS de Ligação a RNA/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Linhagem Celular , Demência Frontotemporal/metabolismo , Demência Frontotemporal/patologia , Humanos , Técnicas In Vitro , Modelos Teóricos , Dobramento de Proteína
8.
Neuroreport ; 31(5): 399-405, 2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32101952

RESUMO

Visual imprinting is a learning process whereby young animals come to prefer a visual stimulus after exposure to it (training). The intermediate medial mesopallium in the domestic chick forebrain is critical for visual imprinting and contributes to molecular regulation of memory formation. Criteria used to infer that a change following training is learning-related have been formulated and published. Cognin (protein disulphide isomerase) is one of several identified plasma membrane and mitochondrial proteins that are upregulated in a learning-related way 24 hours after training. Since virtually nothing is known about the cognin interactome, we have used immunoaffinity chromatography and mass spectrometry to identify proteins that interact with cognin in the cytoplasmic and plasma membrane-mitochondrial fractions. As the learning-related upregulation of cognin has been shown to occur in the plasma membrane-mitochondrial fraction and not in the cytoplasmic fraction, we studied the effect of training on three cognin-interacting partners in the plasma membrane-mitochondrial fraction: the b5 subunit of mitochondrial ATP synthase and the alpha-2 and alpha-3 subunits of sodium-potassium ATPase. Learning-related upregulation was found in the left intermediate medial mesopallium 24 hours after training for the b5 subunit of mitochondrial ATP synthase and the alpha-2 subunit of sodium-potassium ATPase. The hemispheric asymmetry revealed here is consistent with the predominance of many other learning-related effects in the left intermediate medial mesopallium. The alpha-2 subunit of sodium-potassium ATPase is mainly expressed in astrocytes, supporting a role for these glial cells in memory.


Assuntos
Fixação Psicológica Instintiva/fisiologia , Aprendizagem/fisiologia , Proteínas de Membrana/metabolismo , Memória/fisiologia , Membranas Mitocondriais/metabolismo , Animais , Proteínas Mitocondriais/metabolismo , Isomerases de Dissulfetos de Proteínas/farmacologia
9.
Brain Behav ; 10(3): e01534, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31957985

RESUMO

INTRODUCTION: We examined the effects of exogenous protein disulfide isomerase A3 (PDIA3) on hippocampal neurogenesis in gerbils under control and ischemic damage. METHODS: To facilitate the delivery of PDIA3 to the brain, we constructed Tat-PDIA3 protein and administered vehicle (10% glycerol) or Tat-PDIA3 protein once a day for 28 days. On day 24 of vehicle or Tat-PDIA3 treatment, ischemia was transiently induced by occlusion of both common carotid arteries for 5 min. RESULTS: Administration of Tat-PDIA3 significantly reduced ischemia-induced spontaneous motor activity, and the number of NeuN-positive nuclei in the Tat-PDIA3-treated ischemic group was significantly increased in the CA1 region compared to that in the vehicle-treated ischemic group. Ki67- and DCX-immunoreactive cells were significantly higher in the Tat-PDIA3-treated group compared to the vehicle-treated control group. In vehicle- and Tat-PDIA3-treated ischemic groups, the number of Ki67- and DCX-immunoreactive cells was significantly higher as compared to those in the vehicle- and Tat-PDIA3-treated control groups, respectively. In the dentate gyrus, the numbers of Ki67-immunoreactive cells were comparable between vehicle- and Tat-PDIA3-treated ischemic groups, while more DCX-immunoreactive cells were observed in the Tat-PDIA3-treated group. Transient forebrain ischemia increased the expression of phosphorylated cAMP-response element-binding protein (pCREB) in the dentate gyrus, but the administration of Tat-PDIA3 robustly increased pCREB-positive nuclei in the normal gerbils, but not in the ischemic gerbils. Brain-derived neurotrophic factor (BDNF) mRNA expression was significantly increased in the Tat-PDIA3-treated group compared to that in the vehicle-treated group. Transient forebrain ischemic increased BDNF mRNA levels in both vehicle- and Tat-PDIA3-treated groups, and there were no significant differences between groups. CONCLUSIONS: These results suggest that Tat-PDIA3 enhances cell proliferation and neuroblast numbers in the dentate gyrus in normal, but not in ischemic gerbils, by increasing BDNF mRNA and phosphorylation of pCREB.


Assuntos
Isquemia Encefálica/patologia , Proliferação de Células/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/farmacologia , Animais , Contagem de Células , Gerbillinae , Masculino , Fosforilação
10.
Antiviral Res ; 143: 246-251, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28461071

RESUMO

Alphaviruses require conserved cysteine residues for proper folding and assembly of the E1 and E2 envelope glycoproteins, and likely depend on host protein disulfide isomerase-family enzymes (PDI) to aid in facilitating disulfide bond formation and isomerization in these proteins. Here, we show that in human HEK293 cells, commercially available inhibitors of PDI or modulators thereof (thioredoxin reductase, TRX-R; endoplasmic reticulum oxidoreductin-1, ERO-1) inhibit the replication of CHIKV chikungunya virus (CHIKV) in vitro in a dose-dependent manner. Further, the TRX-R inhibitor auranofin inhibited Venezuelan equine encephalitis virus and the flavivirus Zika virus replication in vitro, while PDI inhibitor 16F16 reduced replication but demonstrated notable toxicity. 16F16 significantly altered the viral genome: plaque-forming unit (PFU) ratio of CHIKV in vitro without affecting relative intracellular viral RNA quantities and inhibited CHIKV E1-induced cell-cell fusion, suggesting that PDI inhibitors alter progeny virion infectivity through altered envelope function. Auranofin also increased the extracellular genome:PFU ratio but decreased the amount of intracellular CHIKV RNA, suggesting an alternative mechanism of action. Finally, auranofin reduced footpad swelling and viremia in the C57BL/6 murine model of CHIKV infection. Our results suggest that targeting oxidative folding pathways represents a potential new anti-alphavirus therapeutic strategy.


Assuntos
Antivirais/farmacologia , Febre de Chikungunya/virologia , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Infecções por Alphavirus/virologia , Animais , Auranofina/antagonistas & inibidores , Febre de Chikungunya/mortalidade , Vírus Chikungunya/patogenicidade , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/efeitos dos fármacos , Flavivirus/efeitos dos fármacos , Células HEK293 , Humanos , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Isomerases de Dissulfetos de Proteínas/farmacologia , Dobramento de Proteína , Tiorredoxina Dissulfeto Redutase/farmacologia , Proteínas do Envelope Viral/metabolismo , Replicação Viral/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Infecção por Zika virus/virologia
11.
J Clin Invest ; 122(6): 2104-13, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22565308

RESUMO

Thrombosis, or blood clot formation, and its sequelae remain a leading cause of morbidity and mortality, and recurrent thrombosis is common despite current optimal therapy. Protein disulfide isomerase (PDI) is an oxidoreductase that has recently been shown to participate in thrombus formation. While currently available antithrombotic agents inhibit either platelet aggregation or fibrin generation, inhibition of secreted PDI blocks the earliest stages of thrombus formation, suppressing both pathways. Here, we explored extracellular PDI as an alternative target of antithrombotic therapy. A high-throughput screen identified quercetin-3-rutinoside as an inhibitor of PDI reductase activity in vitro. Inhibition of PDI was selective, as quercetin-3-rutinoside failed to inhibit the reductase activity of several other thiol isomerases found in the vasculature. Cellular assays showed that quercetin-3-rutinoside inhibited aggregation of human and mouse platelets and endothelial cell-mediated fibrin generation in human endothelial cells. Using intravital microscopy in mice, we demonstrated that quercetin-3-rutinoside blocks thrombus formation in vivo by inhibiting PDI. Infusion of recombinant PDI reversed the antithrombotic effect of quercetin-3-rutinoside. Thus, PDI is a viable target for small molecule inhibition of thrombus formation, and its inhibition may prove to be a useful adjunct in refractory thrombotic diseases that are not controlled with conventional antithrombotic agents.


Assuntos
Plaquetas/metabolismo , Fibrinolíticos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Rutina/farmacologia , Trombose/tratamento farmacológico , Animais , Inibidores Enzimáticos/farmacologia , Fibrina/genética , Fibrina/metabolismo , Humanos , Camundongos , Isomerases de Dissulfetos de Proteínas/efeitos adversos , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/farmacologia , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Trombose/induzido quimicamente , Trombose/enzimologia
12.
Blood ; 119(7): 1737-46, 2012 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-22207737

RESUMO

A close homologue to protein disulfide isomerase (PDI) called ERp57 forms disulfide bonds in glycoproteins in the endoplasmic reticulum and is expressed on the platelet surface. We generated 2 rabbit Abs to ERp57. One Ab strongly inhibited ERp57 in a functional assay and strongly inhibited platelet aggregation. There was minimal cross-reactivity of this Ab with PDI by Western blot or in the functional assay. This Ab substantially inhibited activation of the αIIbß3 fibrinogen receptor and P-selectin expression. Furthermore, adding ERp57 to platelets potentiated aggregation. In contrast, adding a catalytically inactive ERp57 inhibited platelet aggregation. When infused into mice the inactive ERp57 prolonged the tail bleeding times. We generated 2 IgG2a mAbs that reacted with ERp57 by immunoblot. One of these Abs inhibited both ERp57 activity and platelet aggregation. The other Ab did not inhibit ERp57 activity or platelet aggregation. The inhibitory Ab inhibited activation of αIIbß3 and P-selectin expression, prolonged tail bleeding times, and inhibited FeCl(3)-induced thrombosis in mice. Finally, we found that a commonly used mAb to PDI also inhibited ERp57 activity. We conclude that a glycoprotein-specific member of the PDI family, ERp57, is required for platelet aggregation, hemostasis, and thrombosis.


Assuntos
Hemostasia/genética , Agregação Plaquetária/genética , Isomerases de Dissulfetos de Proteínas/fisiologia , Trombose/genética , Animais , Cisteína/genética , Hemostasia/efeitos dos fármacos , Hemostasia/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Proteínas Mutantes/farmacologia , Proteínas Mutantes/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Isomerases de Dissulfetos de Proteínas/farmacologia , Coelhos , Serina/genética , Trombose/enzimologia , Trombose/metabolismo
13.
Cancer Lett ; 210(2): 219-26, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15183538

RESUMO

Tuberous sclerosis (TSC) is an autosomal dominant tumour suppressor gene syndrome affecting about 1 in 6000 individuals. It is characterized by mental retardation and epilepsy. A variety of tumours characteristically occur in different organs of TSC patients. Typically, highly epileptogenic dysplastic lesions (tubers) composed of abnormal shaped neurones can be detected in the cerebral cortex. Two tumour suppressor genes have been shown to be responsible for this disease: TSC1, encoding hamartin, and TSC2, encoding tuberin. In this study we performed a proteomic approach of two-dimensional gel electrophoresis with subsequent mass spectrometrical identification of protein spots after ectopic overexpression of human TSC1 or TSC2. We found the protein levels of alpha1-tubulin, protein disulfide isomerase, tropomyosin 3 and 5 and vimentin to be regulated by the two tuberous sclerosis gene products. The here presented findings suggest that deregulation of the control of these target proteins might contribute to the development of tumours in tuberous sclerosis patients. These data provide important new insights into the molecular development of this disease especially since alpha1-tubulin, protein disulfide isomerase and certain tropomyosins have also been implicated in the regulation of neuronal differentiation.


Assuntos
Isomerases de Dissulfetos de Proteínas/análise , Tropomiosina/análise , Esclerose Tuberosa/complicações , Esclerose Tuberosa/genética , Tubulina (Proteína)/análise , Vimentina/análise , Western Blotting , Diferenciação Celular , Transformação Celular Neoplásica , Eletroforese em Gel Bidimensional , Feminino , Regulação da Expressão Gênica , Células HeLa , Humanos , Espectrometria de Massas , Isomerases de Dissulfetos de Proteínas/biossíntese , Isomerases de Dissulfetos de Proteínas/farmacologia , Proteômica , Tropomiosina/biossíntese , Tropomiosina/farmacologia , Tubulina (Proteína)/biossíntese , Tubulina (Proteína)/farmacologia , Vimentina/biossíntese , Vimentina/farmacologia
14.
FEBS Lett ; 514(2-3): 290-4, 2002 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-11943168

RESUMO

The heavy chain (Hc) and light chain (Lc) genes of the Fab fragment of a catalytic antibody 6D9 were simultaneously expressed in an Escherichia coli in vitro transcription/translation system without a reducing agent. The intermolecular disulfide bond between the Hc and Lc was found formed, suggesting a correct formation of the Fab fragment in the in vitro system. In enzyme-linked immunosorbent assay, the Fab fragment synthesized in vitro exhibited an antigen-binding activity. Addition of reduced glutathione, oxidized glutathione, protein disulfide-isomerase and molecular chaperones, GroEL and GroES, increased the solubility and the antigen-binding activity of the Fab fragment greatly. The in vitro synthesized Fab was purified by means of a hexa-histidine tag attached to the C-terminus of the Hc. Catalytic assay of the purified Fab fragment showed that the His-tagged Fab fragment synthesized in vitro had a catalytic activity comparable to that produced in vivo.


Assuntos
Anticorpos Catalíticos/genética , Fragmentos Fab das Imunoglobulinas/biossíntese , Fragmentos Fab das Imunoglobulinas/química , Antígenos/metabolismo , Catálise/efeitos dos fármacos , Sistema Livre de Células/química , Sistema Livre de Células/metabolismo , Chaperonina 10/farmacologia , Chaperonina 60/farmacologia , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Escherichia coli , Glutationa/farmacologia , Dissulfeto de Glutationa/farmacologia , Haptenos/metabolismo , Fragmentos Fab das Imunoglobulinas/genética , Chaperonas Moleculares/farmacologia , Oxirredução , Ligação Proteica/efeitos dos fármacos , Biossíntese de Proteínas/fisiologia , Isomerases de Dissulfetos de Proteínas/farmacologia , Solubilidade/efeitos dos fármacos , Transcrição Gênica/fisiologia
15.
Free Radic Biol Med ; 32(7): 584-95, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11909693

RESUMO

Alanyl aminopeptidase (APN) is a surface-bound metallopeptidase that processes the N-terminals of biologically active peptides such as enkephalins, angiotensins, neurokinins, and cytokines. It exerts profound activity on vital processes such as immune response, cellular growth, and blood pressure control. Inhibition of either APN gene expression or its enzymatic activity severely affects leukocyte growth and function. We show here that oxidoreductase-mediated modulations of the cell surface thiol status affect the enzymatic activity of APN. Additional evidence for the pivotal role of extracellular cysteines in the APN molecule was obtained when substitution of any of these six cysteines caused complete loss of surface expression and enzymatic activity. In contrast, the transmembrane Cys24 appears to have no similar function. Enzymatically inactive cysteine mutants were retained in the endoplasmic reticulum as shown by high-resolution imaging and Endoglycosidase H digestion. In the absence of any crystal-structure data, the demonstration that individual extracellular cysteines contribute to APN expression and function appears to be of particular importance. The data are the first to show thiol-dependent modulation of the activity of a typical surface-bound peptidase at the cell surface, probably reflecting a general regulating mechanism. This may relate to various disease processes such as inflammation or malignant transformation.


Assuntos
Antígenos CD13/metabolismo , Cisteína/metabolismo , Amidoidrolases/metabolismo , Antígenos de Superfície/metabolismo , Primers do DNA/química , Etilmaleimida/farmacologia , Proteínas de Fluorescência Verde , Hexosaminidases/metabolismo , Humanos , Immunoblotting , Iodoacetamida/farmacologia , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase , Reação em Cadeia da Polimerase , Isomerases de Dissulfetos de Proteínas/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Frações Subcelulares , Células U937/metabolismo
16.
J Biochem ; 129(5): 821-6, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11328607

RESUMO

Reduced apo-alpha-lactalbumin (r-LA) in the pre-molten globule state is soluble in neutral and reduced buffer at 25 degrees C but becomes aggregated when aggregates of various proteins are added. However, protein aggregates do not induce the aggregation of apo-alpha-lactalbumin in the molten globule state. The presence of the molecular chaperone protein disulfide isomerase or the "chemical chaperone" polyethyleneglycol inhibits the induced aggregation. Native proteins, aggregation-free folding intermediates, and soluble aggregates do not induce the aggregation. The interaction between r-LA and protein aggregates is hydrophobic in nature. These findings suggest that pre-molten globule state of LA is the target not only for chaperones but also for protein aggregates.


Assuntos
Apoproteínas/química , Lactalbumina/química , Chaperonas Moleculares/química , Isomerases de Dissulfetos de Proteínas/química , Animais , Soluções Tampão , Bovinos , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Isomerases de Dissulfetos de Proteínas/farmacologia , Dobramento de Proteína
17.
J Protein Chem ; 19(7): 569-74, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11233170

RESUMO

Simultaneous presence of two chaperones, GroEL and protein disulfide isomerase (PDI), assists the reactivation of denatured D-glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in an additive way. Delayed addition of chaperones to the refolding solution after dilution of denatured GAPDH indicates an interaction with intermediates formed mainly in the first 5 min for PDI and formed within a longer time period for GroEL-ATP. The above indicate that the two chaperones interact with different folding intermediates of GAPDH. After delayed addition of one chaperone to the refolding mixture containing the other at 4 degrees C, GroEL binds with all GAPDH intermediates dissociated from PDI, and PDI interacts with the intermediates released from GroEL during the first 10-20 min. It is suggested that the GAPDH folding intermediates released from the chaperone-bound complex are still partially folded so as to be rebound by the other chaperone. The above results clearly support the network model of GroEL and PDI.


Assuntos
Chaperonina 60/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Bovinos , Sinergismo Farmacológico , Gliceraldeído 3-Fosfato/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/química , Gliceraldeído-3-Fosfato Desidrogenases/efeitos dos fármacos , Modelos Químicos , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/farmacologia , Ligação Proteica , Isomerases de Dissulfetos de Proteínas/farmacologia , Dobramento de Proteína , Renaturação Proteica/efeitos dos fármacos , Coelhos , Fatores de Tempo
18.
FEBS Lett ; 456(1): 143-5, 1999 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-10452546

RESUMO

The role of protein disulfide isomerase (PDI) in the regeneration of ribonuclease A with dithiothreitol (DTT) was investigated at three different temperatures. The rates of formation of the native protein were markedly increased in the presence of PDI, 9-fold at 15 degrees C, 6-fold at 25 degrees C and 62-fold at 37 degrees C, respectively. In the presence of PDI, major changes were found in the distribution of intermediates in the three-disulfide region at 25 and 15 degrees C and also in the one-disulfide region at 15 degrees C, with the fast accumulation of the two native-like species des-[65-72] and des-[40-95]. The present results indicate that PDI does not alter the two major parallel pathways involving des-[65-72] and des-[40-95] in the regeneration of ribonuclease A with DTT.


Assuntos
Ditiotreitol/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Ribonuclease Pancreático/química , Ribonuclease Pancreático/metabolismo , Animais , Bovinos , Isomerases de Dissulfetos de Proteínas/farmacologia , Dobramento de Proteína , Ribonuclease Pancreático/efeitos dos fármacos , Temperatura
19.
Protein Eng ; 11(12): 1293-8, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9930680

RESUMO

The refolding characteristics of Taka-amylase A (TAA) from Aspergillus oryzae in the presence of the chaperonin GroE were studied in terms of activity and fluorescence. Disulfide-bonded (intact) TAA and non-disulfide-bonded (reduced) TAA were unfolded in guanidine hydrochloride and refolded by dilution into buffer containing GroE. The intermediates of both intact and reduced enzymes were trapped by GroEL in the absence of nucleotide. Upon addition of nucleotides such as ATP, ADP, CTP or UTP, the intermediates were released from GroEL and recovery of activity was detected. In both cases, the refolding yields in the presence of GroEL and ATP were higher than spontaneous recoveries. Fluorescence studies of intrinsic tryptophan and a hydrophobic probe, 8-anilinonaphthalene-1-sulfonate, suggested that the intermediates trapped by GroEL assumed conformations with different hydrophobic properties. The presence of protein disulfide isomerase or reduced and oxidized forms of glutathione in addition to GroE greatly enhanced the refolding reaction of reduced TAA. These findings suggest that GroE has an ability to recognize folding intermediates of TAA protein and facilitate refolding, regardless of the existence or absence of disulfide bonds in the protein.


Assuntos
Aspergillus oryzae/enzimologia , Proteínas de Bactérias/farmacologia , Dissulfetos/metabolismo , Proteínas de Choque Térmico/farmacologia , Dobramento de Proteína , alfa-Amilases/química , Trifosfato de Adenosina/farmacologia , Proteínas de Bactérias/administração & dosagem , Chaperonina 10/farmacologia , Chaperonina 60/metabolismo , Chaperonina 60/farmacologia , Chaperoninas , Fenômenos Químicos , Físico-Química , Proteínas de Escherichia coli , Glutationa/farmacologia , Proteínas de Choque Térmico/administração & dosagem , Nucleotídeos/farmacologia , Oxirredução , Isomerases de Dissulfetos de Proteínas/farmacologia , Espectrometria de Fluorescência , alfa-Amilases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA