Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Phytomedicine ; 82: 153449, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33387969

RESUMO

BACKGROUND/PURPOSE: Juglone, a natural compound widely found in Juglandaceae plants, has been suggested as a potential drug candidate for treating cancer, inflammation, and diabetic vascular complications. In the present study, the antiplatelet effect and underlying mechanisms of juglone were investigated for the first time. STUDY DESIGN/METHODS: Human platelet aggregation and activation were measured by turbidimetric aggregometry, flow cytometry, and Western blotting. In vitro antithrombotic activity of juglone was assessed using collagen-coated flow chambers under whole-blood flow conditions. The effect of juglone on protein disulfide isomerase (PDI) activity was determined by the dieosin glutathione disulfide assay. RESULTS: Juglone (1 - 5 µM) inhibited platelet aggregation and glycoprotein (GP) IIb/IIIa activation caused by various agonists. In a whole blood flow chamber system, juglone reduced thrombus formation on collagen-coated surfaces under arterial shear rates. Juglone abolished intracellular Ca2+ elevation and protein kinase C activation caused by collagen, but had no significant effect on that induced by G protein-coupled receptor agonists. In contrast, Akt activation caused by various agonists were inhibited in juglone-treated platelets. Additionally, juglone showed inhibitory effects on both recombinant human PDI and platelet surface PDI at concentrations similar to those needed to prevent platelet aggregation. CONCLUSION: Juglone exhibits potent in vitro antiplatelet and antithrombotic effects that are associated with inhibition of Akt activation and platelet surface PDI activity.


Assuntos
Naftoquinonas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Plaquetas/efeitos dos fármacos , Humanos , Ativação Plaquetária/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trombose/metabolismo
2.
Theranostics ; 9(8): 2282-2298, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31149044

RESUMO

Aberrant overexpression of endoplasmic reticulum (ER)-resident oxidoreductase protein disulfide isomerase (PDI) plays an important role in cancer progression. In this study, we demonstrate that PDI promotes glioblastoma (GBM) cell growth and describe a class of allosteric PDI inhibitors that are selective for PDI over other PDI family members. Methods: We performed a phenotypic screening triage campaign of over 20,000 diverse compounds to identify PDI inhibitors cytotoxic to cancer cells. From this screen, BAP2 emerged as a lead compound, and we assessed BAP2-PDI interactions with gel filtration, thiol-competition assays, and site-directed mutagenesis studies. To assess selectivity, we compared BAP2 activity across several PDI family members in the PDI reductase assay. Finally, we performed in vivo studies with a mouse xenograft model of GBM combining BAP2 and the standard of care (temozolomide and radiation), and identified affected gene pathways with nascent RNA sequencing (Bru-seq). Results: BAP2 and related analogs are novel PDI inhibitors that selectively inhibit PDIA1 and PDIp. Though BAP2 contains a weak Michael acceptor, interaction with PDI relies on Histidine 256 in the b' domain of PDI, suggesting allosteric binding. Furthermore, both in vitro and in vivo, BAP2 reduces cell and tumor growth. BAP2 alters the transcription of genes involved in the unfolded protein response, ER stress, apoptosis and DNA repair response. Conclusion: These results indicate that BAP2 has anti-tumor activity and the suppressive effect on DNA repair gene expression warrants combination with DNA damaging agents to treat GBM.


Assuntos
Antineoplásicos/farmacologia , Reparo do DNA/efeitos dos fármacos , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Glioblastoma/tratamento farmacológico , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/isolamento & purificação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/isolamento & purificação , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Transplante de Neoplasias , Ligação Proteica , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Transplante Heterólogo , Resultado do Tratamento
3.
Nutrients ; 11(1)2019 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-30658391

RESUMO

Pomegranate fruit is a functional food of high interest for human health due to its wide range of phytochemicals with antioxidant properties are implicated in the prevention of inflammation and cancer. Ellagitannins, such as punicalagin and ellagic acid, play a role as anti-atherogenic and neuroprotective molecules in the complex fighting against the degenerative diseases. The aim of this work was to evaluate the composition in punicalagins and ellagic acid of differently obtained extracts from whole fruit, peels and juices, prepared by squeezing or by centrifugation, of pomegranate belonging to different cultivars. Moreover, a wider phenolic fingerprint was also determined. The bioactivity of the extracts was tested on the redox activity of PDIA3 disulfide isomerase, an enzyme involved in the regulation of several cellular functions and associated with different diseases such as cancer, prion disorders, Alzheimer's and Parkinson's diseases. The results demonstrate that the different ratios between punicalagin and ellagic acid modulate the enzyme activity and other ellagitannins could interfere with this activity.


Assuntos
Inibidores Enzimáticos/farmacologia , Frutas/química , Taninos Hidrolisáveis/farmacologia , Lythraceae/química , Isomerases de Dissulfetos de Proteínas/metabolismo , Antioxidantes/farmacologia , Ácido Elágico/farmacologia , Sucos de Frutas e Vegetais/análise , Humanos , Extratos Vegetais/farmacologia , Polifenóis/farmacologia , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores
4.
Virus Res ; 247: 26-33, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29382552

RESUMO

Seasonal flu as well as potential pandemic flu outbreaks continuously underscores the importance of the preventive and therapeutic measures against influenza viruses. During screening of natural and synthetic small molecules against influenza A and B virus, we identified juniferdin as a highly effective inhibitor against both viruses in cells. Since juniferdin is known to inhibit protein disulfide isomerases (PDIs), multiple PDI inhibitors were tested against these viruses. Among PDI inhibitors, 16F16, PACMA31, isoquercetin, epigallocatechin-3-gallate or nitazoxanide significantly reduced the replication of influenza A and B viruses in MDCK and A549 cells. Furthermore, siRNAs specific to three PDI family members (PDI1, PDIA3 or PDIA4) also significantly reduced the replication of influenza A and B viruses in cells. These results suggest that PDIs may serve as excellent targets for the development of new anti-influenza drugs.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Parabenos/farmacologia , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , RNA Viral/antagonistas & inibidores , Sesquiterpenos/farmacologia , Células A549 , Animais , Catequina/análogos & derivados , Catequina/farmacologia , Cães , Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/metabolismo , Vírus da Influenza B/genética , Vírus da Influenza B/crescimento & desenvolvimento , Vírus da Influenza B/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Células Madin Darby de Rim Canino , Nitrocompostos , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Quercetina/análogos & derivados , Quercetina/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Viral/biossíntese , RNA Viral/genética , Tiazóis/farmacologia
5.
Zhongguo Zhong Yao Za Zhi ; 42(15): 3004-3010, 2017 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-29139271

RESUMO

Increasing evidence suggested that protein disulfide isomerase supported the survival and progression of several cancers. Nelumbinis Plumula is a Chinese traditional herb which showed antitumor activity. To find if the Nelumbinis Plumula affect protein disulfide isomerase activity, we studied its chemical constituents, and 12 monomeric compounds were isolated by means of solvent extraction, silica gel column chromatography, preparative HPLC and recrystallization. Among them, N-methylcoclaurine, kaempferol, chrysoeriol-7-O-neohesperidoside and mannitol were obtained for the first time. Following, we tested the compounds inhibitory activity on protein disulfide isomerase. The results showed that N-methylcoclaurine, neferine, liensinine and isoliensinine could inhibit the activity of protein disulfide isomerase in vitro, their IC50 values were 1.4, 2.9, 4.0 and 5.4 µmol•L⁻¹, respectively.


Assuntos
Medicamentos de Ervas Chinesas/química , Lotus/química , Compostos Fitoquímicos/análise , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Cromatografia Líquida de Alta Pressão , Plantas Medicinais/química
6.
Biochem Biophys Res Commun ; 489(2): 164-170, 2017 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-28549585

RESUMO

Orexins (orexin-A and orexin-B) are neuropeptides that are reduced in narcolepsy, a sleep disorder that is characterized by excessive daytime sleepiness, sudden sleep attacks and cataplexy. However, it remains unclear how orexins in the brain and orexin neurons are reduced in narcolepsy. Orexin-A has two closely located intramolecular disulfide bonds and is prone to misfolding due to the formation of incorrect disulfide bonds. Protein disulfide isomerase (PDI) possesses disulfide interchange activity. PDI can modify misfolded orexin-A to its native form by rearrangement of two disulfide bonds. We have previously demonstrated that sleep deprivation and a high fat diet increase nitric oxide in the brain. This increase triggers S-nitrosation and inactivation of PDI, leading to aggregation of orexin-A and reduction of orexin neurons. However, the relationship between PDI inactivation and loss of orexin neurons has not yet been fully elucidated. In the present study, we used a PDI inhibitor, cystamine, to elucidate the precise molecular mechanism by which PDI inhibition reduces the number of orexin neurons. In rat hypothalamic slice cultures, cystamine induced selective depletion of orexin-A, but not orexin-B and melanin-concentrating hormone. Moreover, cystamine triggered aggregation of orexin-A, but not orexin-B in the Golgi apparatus of hypothalamic slice cultures and in vivo mouse brains. However, cystamine did not induce endoplasmic reticulum (ER) stress, and an ER stress inducer did not trigger aggregation of orexin-A in slice cultures. Finally, we demonstrated that cystamine significantly decreased extracellular secretion of orexin-A in AD293 cells overexpressing prepro-orexin. These findings suggest that cystamine-induced PDI inhibition induces selective depletion, aggregation in the Golgi apparatus and impaired secretion of orexin-A. These effects may represent an initial step in the pathogenesis of narcolepsy.


Assuntos
Cistamina/farmacologia , Complexo de Golgi/efeitos dos fármacos , Orexinas/química , Orexinas/metabolismo , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Animais , Células Cultivadas , Cistamina/administração & dosagem , Complexo de Golgi/metabolismo , Hipotálamo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Ratos , Ratos Wistar
7.
Nat Commun ; 7: 12579, 2016 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-27573496

RESUMO

Protein disulfide isomerase (PDI) is an oxidoreductase essential for folding proteins in the endoplasmic reticulum. The domain structure of PDI is a-b-b'-x-a', wherein the thioredoxin-like a and a' domains mediate disulfide bond shuffling and b and b' domains are substrate binding. The b' and a' domains are connected via the x-linker, a 19-amino-acid flexible peptide. Here we identify a class of compounds, termed bepristats, that target the substrate-binding pocket of b'. Bepristats reversibly block substrate binding and inhibit platelet aggregation and thrombus formation in vivo. Ligation of the substrate-binding pocket by bepristats paradoxically enhances catalytic activity of a and a' by displacing the x-linker, which acts as an allosteric switch to augment reductase activity in the catalytic domains. This substrate-driven allosteric switch is also activated by peptides and proteins and is present in other thiol isomerases. Our results demonstrate a mechanism whereby binding of a substrate to thiol isomerases enhances catalytic activity of remote domains.


Assuntos
Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/metabolismo , Dobramento de Proteína , Regulação Alostérica/efeitos dos fármacos , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Domínio Catalítico/efeitos dos fármacos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/uso terapêutico , Voluntários Saudáveis , Humanos , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Isomerases de Dissulfetos de Proteínas/química , Estrutura Terciária de Proteína/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Trombose/sangue , Trombose/tratamento farmacológico , Trombose/patologia
8.
Oncotarget ; 7(8): 8797-808, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26848777

RESUMO

There is no effective drug to treat EV71 infection yet. Traditional Chinese herbs are great resources for novel antiviral compounds. Here we showed that Oblongifolin M (OM), an active compound isolated from Garcinia oblongifolia, potently inhibited EV71 infection in a dose dependent manner. To identify its potential effectors in the host cells, we successfully identified 18 proteins from 52 differentially expressed spots by comparative proteomics studies. Further studies showed that knockdown of ERp57 inhibited viral replication through downregulating viral IRES (internal ribosome entry site) activities, whereas ectopic expression of ERp57 increased IRES activity and partly rescued the inhibitory effects of OM on viral replication. We demonstrated that OM is an effective antiviral agent; and that ERp57 is one of its cellular effectors against EV71 infection.


Assuntos
Infecções por Enterovirus/prevenção & controle , Garcinia/química , Extratos Vegetais/farmacologia , Isomerases de Dissulfetos de Proteínas/metabolismo , Rabdomiossarcoma/prevenção & controle , Terpenos/farmacologia , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Western Blotting , Eletroforese em Gel Bidimensional , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/patologia , Infecções por Enterovirus/virologia , Genoma Viral , Humanos , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Isomerases de Dissulfetos de Proteínas/genética , Proteômica , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Rabdomiossarcoma/virologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Células Tumorais Cultivadas , Proteínas Virais/metabolismo
9.
PLoS One ; 9(11): e112986, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25419565

RESUMO

RATIONALE: The family of natriuretic peptides (NPs), including atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), exert important and diverse actions for cardiovascular and renal homeostasis. The autocrine and paracrine functions of the NPs are primarily mediated through the cellular membrane bound guanylyl cyclase-linked receptors GC-A (NPR-A) and GC-B (NPR-B). As the ligands and receptors each contain disulfide bonds, a regulatory role for the cell surface protein disulfide isomerase (PDI) was investigated. OBJECTIVE: We utilized complementary in vitro and in vivo models to determine the potential role of PDI in regulating the ability of the NPs to generate its second messenger, cyclic guanosine monophosphate. METHODS AND RESULTS: Inhibition of PDI attenuated the ability of ANP, BNP and CNP to generate cGMP in human mesangial cells (HMCs), human umbilical vein endothelial cells (HUVECs), and human aortic smooth muscle cells (HASMCs), each of which were shown to express PDI. In LLC-PK1 cells, where PDI expression was undetectable by immunoblotting, PDI inhibition had a minimal effect on cGMP generation. Addition of PDI to cultured LLC-PK1 cells increased intracellular cGMP generation mediated by ANP. Inhibition of PDI in vivo attenuated NP-mediated generation of cGMP by ANP. Surface Plasmon Resonance demonstrated modest and differential binding of the natriuretic peptides with immobilized PDI in a cell free system. However, PDI was shown to co-localize on the surface of cells with GC-A and GC-B by co-immunoprecpitation and immunohistochemistry. CONCLUSION: These data demonstrate for the first time that cell surface PDI expression and function regulate the capacity of natriuretic peptides to generate cGMP through interaction with their receptors.


Assuntos
Membrana Celular/metabolismo , GMP Cíclico/metabolismo , Peptídeos Natriuréticos/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Animais , Bacitracina/farmacologia , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Toxina Diftérica/farmacologia , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células LLC-PK1 , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/metabolismo , Camundongos Endogâmicos C57BL , Microscopia Confocal , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Ligação Proteica , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Isomerases de Dissulfetos de Proteínas/genética , Interferência de RNA , Receptores Acoplados a Guanilato Ciclase/metabolismo , Suínos
10.
Oncol Res ; 19(10-11): 445-53, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22715587

RESUMO

Protein disulfide isomerase (PDI) is a multifunctional protein of the thioredoxin superfamily. PDI mediates proper protein folding by oxidation or isomerization and disrupts disulfide bonds by reduction; it also has chaperone and antichaperone activities. Although PDI localizes primarily to the endoplasmic reticulum (ER), it is secreted and expressed on the cell surface. In the ER, PDI is primarily involved in protein folding, whereas on the cell surface, it reduces disulfide bonds. The functions of PDI depend on its localization and the redox state of its active site cysteines. The ER-based functions of PDI are linked to cancer invasion and migration. Surface-associated PDI facilitates the entry of viruses, such as HIV-1, and toxins, such as diphtheria and cholera. Thus, based on its involvement in pathological events, PDI is considered a potential drug target. However, a significant challenge in the therapeutic targeting of PDI is discovering function-specific inhibitors for it. To this end, a wide range of therapeutic agents, such as antibiotics, thiol blockers, estrogenic compounds, and arsenical compounds, have been used, although few are bona fide specific inhibitors. In this review, we will describe the potential of PDI as a therapeutic drug target.


Assuntos
Inibidores Enzimáticos/farmacologia , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Animais , Bacitracina/farmacologia , Estrogênios/farmacologia , Humanos , Parabenos/farmacologia , Isomerases de Dissulfetos de Proteínas/química , Isomerases de Dissulfetos de Proteínas/fisiologia , Estrutura Terciária de Proteína , Ribostamicina/farmacologia , Sesquiterpenos/farmacologia
11.
Cancer Sci ; 100(11): 2040-5, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19703194

RESUMO

Mitomycin C (MMC), a chemotherapeutic agent in breast cancer treatments, inhibits tumor growth through DNA cross-linking and breaking, but it has severe side effects. Here we examined whether and how curcumin reduced the side effects of MMC. We found that combination treatment with MMC and curcumin reduced tumor weight by 70% and 36% compared with saline and curcumin-treated groups, respectively. The combination treatment reduced weight loss and improved kidney function and bone marrow suppression compared with MMC treatment alone. Moreover, the combination treatment inhibited glucose regulatory protein (GRP58)-mediated DNA cross-linking. The combination treatment inhibited GRP58 through the ERK/p38 MAPK pathway. In conclusion, the current study provided evidence that MMC and curcumin combination treatment reduced MMC side effects by inhibiting GRP58-mediated DNA cross-linking through the ERK/p38 MAPK pathway.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Reagentes de Ligações Cruzadas/efeitos adversos , Curcumina/uso terapêutico , Dano ao DNA/efeitos dos fármacos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Mitomicina/efeitos adversos , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , DNA/química , DNA/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Feminino , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Transplante de Neoplasias , Transplante Heterólogo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
12.
Cancer Res ; 68(13): 5363-9, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18593938

RESUMO

Exploiting vulnerabilities in the intracellular signaling pathways of tumor cells is a key strategy for the development of new drugs. The activation of cellular stress responses mediated by the endoplasmic reticulum (ER) allows cancer cells to survive outside their normal environment. Many proteins that protect cells against ER stress are active as protein disulfide isomerases (PDI) and the aim of this study was to test the hypothesis that apoptosis in response to ER stress can be increased by inhibiting PDI activity. We show that the novel chemotherapeutic drugs fenretinide and velcade induce ER stress-mediated apoptosis in melanoma cells. Both stress response and apoptosis were enhanced by the PDI inhibitor bacitracin. Overexpression of the main cellular PDI, procollagen-proline, 2-oxoglutarate-4-dioxygenase beta subunit (P4HB), resulted in increased PDI activity and abrogated the apoptosis-enhancing effect of bacitracin. In contrast, overexpression of a mutant P4HB lacking PDI activity did not increase cellular PDI activity or block the effects of bacitracin. These results show that inhibition of PDI activity increases apoptosis in response to agents which induce ER stress and suggest that the development of potent, small-molecule PDI inhibitors has significant potential as a powerful tool for enhancing the efficacy of chemotherapy in melanoma.


Assuntos
Apoptose/efeitos dos fármacos , Bacitracina/farmacologia , Retículo Endoplasmático/efeitos dos fármacos , Melanoma/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bacitracina/administração & dosagem , Ácidos Borônicos/administração & dosagem , Ácidos Borônicos/farmacologia , Bortezomib , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Fenretinida/administração & dosagem , Fenretinida/farmacologia , Humanos , Melanoma/patologia , Estresse Oxidativo/fisiologia , Pirazinas/administração & dosagem , Pirazinas/farmacologia , Resultado do Tratamento , Células Tumorais Cultivadas
13.
J Pharm Pharmacol ; 57(2): 213-8, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15720785

RESUMO

The metalloantibiotic bacitracin is a known inhibitor of protein disulfide isomerase (PDI). The disulfide-linked interleukin-12 (IL-12) alphabeta-heterodimer and beta2-homodimer forms are crucial mediators of cell-mediated immune responses and inflammatory reactions. Bacitracin was found to potently block secretion of both the alphabeta- and beta2-dimer forms of IL-12, while it did not affect secretion of the beta-monomer. This inhibition coincided with a reduction in the intracellular amount of PDI found in complex with the beta-chain during intracellular transit. Bacitracin did not affect mRNA levels of the alphabeta- and beta-chain. Similar to bacitracin, N-acetylcysteine blocked alphabeta- and beta2-secretion as well as PDI-beta-chain complex formation. Thus, blocking PDI or shifting the endoplasmic reticulum towards a more reduced status disrupts the oxidative folding pathway or assembly of IL-12 dimer forms. The assembly stage of cytokines in the endoplasmic reticulum may represent a novel target for pharmacological intervention.


Assuntos
Bacitracina/farmacologia , Interleucina-12/antagonistas & inibidores , Interleucina-12/metabolismo , Acetilcisteína/farmacologia , Animais , Bacitracina/química , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/enzimologia , Humanos , Interleucina-12/genética , Metaloproteínas/química , Metaloproteínas/farmacologia , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Isomerases de Dissulfetos de Proteínas/fisiologia , RNA Mensageiro , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA