Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biotechnol Appl Biochem ; 68(2): 267-271, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32311159

RESUMO

The aims of this study were to compare mRNA levels of melanoma differentiation-associated protein 5 (MDA5) and retinoic acid-inducible gene 1 (RIG-1) in multiple sclerosis (MS) patients in comparison to the healthy controls as well as investigating the effects of IFN-ß 1a on the expression of these molecules. In this study, mRNA levels of MDA5 and RIG-1 in peripheral leukocytes of 30 new cases of MS patients and 35 healthy controls were evaluated using the real-time-PCR method. mRNA levels of MDA5 and RIG-1 were determined in the MS patients 6 months after treatment with standard doses of IFN-ß 1a. mRNA levels of MDA5 and RIG-1 were significantly decreased in the MS patients in comparison to the healthy controls. The analysis also revealed that IFN-ß 1a therapy leads to the upregulation of RIG-1, but not MDA5, in the total MS patients and the female group. MS patients suffer from insufficient expression of MDA5 and RIG-1, and IFN-ß 1a therapy results in the upregulation of RIG-1 in the patients, especially in the female patients. Thus, it seems that IFN-ß 1a not only decreased pathogenic inflammatory responses but also modulated the expression of RIG-1 to protect the patients from infectious diseases and upregulation of IFN-I in a positive feedback.


Assuntos
Proteína DEAD-box 58/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon beta-1a/farmacologia , Helicase IFIH1 Induzida por Interferon/biossíntese , Leucócitos/metabolismo , Esclerose Múltipla/metabolismo , Receptores Imunológicos/biossíntese , Feminino , Humanos , Leucócitos/patologia , Masculino , Esclerose Múltipla/patologia
2.
Neurochem Int ; 138: 104777, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32479984

RESUMO

Chemobrain is a well-established clinical syndrome that impairs patient's daily function, in particular attentiveness, coordination and multi-tasking. Thus, it interferes with patient's quality of life. The putative pharmacological intervention against chemobrain relies on understanding the molecular mechanisms underlying it. This study aimed to examine the potential neuroprotective effects of two immunomodulators: Interferon-ß-1a (IFN-ß-1a), as well as Tumor necrosis function-alpha (TNF-α) inhibitor; Infliximab in doxorubicin (DOX)-induced chemobrain in rats. Besides, the current study targets investigating the possible molecular mechanisms in terms of neuromodulation and interference with different death routes controlling neural homeostasis. Herein, the two immunomodulators IFN-ß-1a at a dose of 300,000 units; s.c.three times per week, or Infliximab at a dose of 5 mg/kg/week; i.p. once per week were examined against DOX (2 mg/kg/w, i.p.) once per week for 4 consecutive weeks in rats.The consequent behavioral tests and markers for cognitive impairment, oxidative stress, neuroinflammation, apoptosis and neurobiological abnormalities were further evaluated. Briefly, IFN-ß-1a or Infliximab significantly protected against DOX-induced chemobrain. IFN-ß-1a or Infliximab ameliorated DOX-induced hippocampal histopathological neurodegenerative changes, halted DOX-induced cognitive impairment, abrogated DOX-induced mitochondrial oxidative, inflammatory and apoptotic stress, mitigated DOX-induced autophagic dysfunction and finally upregulated the mitophagic machineries. In conclusion, these findings suggest that either IFN-ß-1a or Infliximab offers neuroprotection against DOX-induced chemobrain which could be explained by their antioxidant, anti-inflammatory, pro-autophagic, pro-mitophagic and antiapoptotic effects. Future clinical studies are recommended to personalize either use of IFN-ß-1a or infliximab to ameliorate DOX-induced chemobrain.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Comprometimento Cognitivo Relacionado à Quimioterapia/tratamento farmacológico , Doxorrubicina/toxicidade , Fatores Imunológicos/uso terapêutico , Infliximab/uso terapêutico , Interferon beta-1a/uso terapêutico , Animais , Comprometimento Cognitivo Relacionado à Quimioterapia/imunologia , Comprometimento Cognitivo Relacionado à Quimioterapia/psicologia , Fatores Imunológicos/farmacologia , Infliximab/farmacologia , Interferon beta-1a/farmacologia , Locomoção/efeitos dos fármacos , Locomoção/imunologia , Masculino , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/imunologia , Ratos
3.
AAPS J ; 21(2): 26, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30737590

RESUMO

Recombinant human interferon-ß (rhIFN-ß) therapy is the first-line treatment in relapsing-remitting forms of multiple sclerosis (MS). The mechanism of action underlying its therapeutic activity is only partially understood as IFN-ßs induce the expression of over 1000 genes modifying multiple immune pathways. Currently, assessment of potency for IFN-ß products is based on their antiviral effect, which is not linked to its therapeutic effect. Here, we explore the use of a multiplexed gene expression system to more broadly characterize IFN-ß bioactivity. We find that MM6 cells stimulated with US-licensed rhIFN-ßs induce a dose-dependent and reproducible pattern of gene expression. This pattern of gene expression was used to compare the bioactivity profile of biosimilar candidates with the corresponding US-licensed rhIFN-ß products, Rebif and Betaseron. While the biosimilar candidate for Rebif matched the pattern of gene expression, there were differences in the expression of a subset of interferon-inducible genes including CXCL-10, CXCL-11, and GBP1 induced by the biosimilar candidate for Betaseron. Assessment of product impurities in both products suggested that the difference was rooted in the presence of innate immune response modulating impurities (IIRMIs) in the licensed product. These studies indicate that determining the expression levels for an array of reporter genes that monitor different pathways can be informative as part of the demonstration of biosimilarity or comparability for complex immunomodulatory products such as IFN-ß, but the sensitivity of each gene to potential impurities in the product should be examined to fully understand the results.


Assuntos
Adjuvantes Imunológicos/farmacologia , Medicamentos Biossimilares/farmacologia , Contaminação de Medicamentos , Expressão Gênica/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Linhagem Celular Tumoral , Perfilação da Expressão Gênica/métodos , Humanos , Interferon beta-1a/farmacologia , Interferon beta-1b/farmacologia , Proteínas Recombinantes/farmacologia
4.
Sheng Wu Gong Cheng Xue Bao ; 34(6): 964-972, 2018 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-29943542

RESUMO

Recombinant human interferon beta (rhIFN-ß) is a glycoprotein produced by genetically engineered cells and has anti-virus, anti-tumor and immunoregulation functions. Although studies have shown that other subtypes of IFN such as IFN-γ affects cell proliferation and differentiation to some extent, the effect of rhIFN-ß on chondrogenic differentiation of human bone marrow mesenchymal stem cells (hMSCs) is less known. In this study we studied the effect of rhIFN-ß on the chondrogenic differentiation of hMSCs by inducing hMSCs into cartilage pellet via adding IFN-ß1a into regular TGF-ß3 chondrogenic differentiation medium. We collected the induced pellets and then detected GAG content, assessed pellets size, observed agreecan using alcian blue staining, and analyzed the expression of Sox and CollangenⅡusing real-time PCR and Western blotting. Addition of 100 ng/mL IFN-ß1a to regular TGF-ß3 chondrogenic differentiation medium could improve the concentration of GAG, increase the size of pellets, promote the formation of aggrecan and up-regulate the expression of CollangenII and Sox9. IFN-ß1a combined with TGF-ß3 could promote chondrogenic differentiation of hMSCs.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Interferon beta-1a/farmacologia , Células-Tronco Mesenquimais/citologia , Células Cultivadas , Colágeno Tipo II/metabolismo , Meios de Cultura , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Fatores de Transcrição SOX9/metabolismo , Fator de Crescimento Transformador beta3/farmacologia
5.
J Endocrinol Invest ; 40(7): 761-770, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28247216

RESUMO

PURPOSE: Type I interferons (IFN-α and IFN-ß) are a class of cytokines that exert several biological activities, such as modulation of cell proliferation and differentiation and of the immune system. Although these cytokines interact with a common receptor complex, IFN-ß showed a more potent antitumor activity than IFN-α in several tumor models. New recombinant human IFN-ß products, such as IFN-ß1a and IFN-ß1b, have been produced in order to improve the stability and bioavailability of natural IFN-ß. In this report, we analyzed the effects of recombinant IFN-ß1a on the cell proliferation of two human androgen-resistant prostate cancer cell lines with neuroendocrine differentiation (DU-145, PC-3) and related mechanisms of action. METHODS: The effects of IFN-ß1a on the cell growth proliferation, cell cycle, and apoptosis have been evaluated in DU-145 and PC-3 cells through MTT assay, DNA flow cytometry with propidium iodide, and Annexin V-FITC/propidium iodide staining, respectively. Moreover, the expression of neuron-specific enolase (NSE), cleaved caspase-3, caspase-8, and PARP was evaluated through Western blotting. RESULTS: IFN-ß1a showed a significant anti-proliferative activity in both androgen-resistant cell lines. This effect was related to cell cycle perturbation and induction in apoptosis, as shown by flow cytometric analysis, the activation of caspase-3 and caspase-8 and PARP cleavage during incubation with IFN-ß1a. Moreover, this cytokine reduced the expression of NSE in both cell lines. CONCLUSIONS: Recombinant IFN-ß1a (Rebif) showed a potent in vitro anti-proliferative activity in androgen-resistant prostate cancer cells, and it could represent a promising tool for the treatment of this tumor.


Assuntos
Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Interferon beta-1a/farmacologia , Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Tumores Neuroendócrinos/patologia , Neoplasias de Próstata Resistentes à Castração/patologia , Células Tumorais Cultivadas
6.
Oncotarget ; 8(8): 13957-13970, 2017 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-28086218

RESUMO

In this study, we examined the molecular mechanism underlying the resistance of cancer cells to R27T, a glycoengineered version of recombinant human interferon (IFN)-ß1a, and sought to overcome R27T resistance through combination therapy. R27T has been shown to induce anti-proliferation and apoptosis in human OVCAR-3 and MCF-7 cells, but not in HeLa cells. R27T treatment increased caspase-8 activity and the consequent cleavage of caspase-8 and -3 in R27T-sensitive OVCAR-3 cells, but not in R27T-resistant HeLa cells. Conversely, R27T increased the expression of cellular FLICE-like inhibitory protein (cFLIP) in HeLa cells, but not in OVCAR-3 cells. The sensitization of HeLa cells with cFLIP small interfering RNA or 4,5,6,7-tetrabromobenzotriazole (TBB, an inhibitor of casein kinase-2) facilitated R27T-induced caspase activation, and consequently apoptosis. In OVCAR-3-xenografted mice, intraperitoneal administration of R27T showed 2.1-fold higher anti-tumor efficacy than did the control vehicle. The combined administration of R27T and TBB showed the greatest anti-tumor effect in HeLa tumor-bearing mice, reducing the relative tumor volume by 35.7% compared to that in R27T-treated mice. Taken together, our results suggest that R27T has potential as an anti-cancer drug, and combination therapy with cFLIP inhibitors may be an effective strategy for overcoming R27T resistance.


Assuntos
Antineoplásicos/farmacologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Interferon beta-1a/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Feminino , Células HeLa , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Proteínas Recombinantes/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Interferon Cytokine Res ; 36(9): 534-41, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27348209

RESUMO

Human type I Interferons (IFN-ß, IFN-ɛ, IFN-κ, IFN-ω, and 12 subtypes of IFN-α) are a family of pleiotropic cytokines with antiviral, antiproliferative, and immunomodulatory activities. They signal through the same cell surface receptors, IFNAR1 and IFNAR2, yet evoking markedly differential potency. One differentiating factor of IFN-ß from other type I interferons is the presence of a consensus sequence (NG) for deamidation. Comparing almost completely deamidated IFN-ß-1a with untreated IFN-ß-1a, this present study reports the increased activities in 3 in-vitro bioassays testing the antiviral, antiproliferative, and immunomodulatory properties, respectively, of the molecule. Deamidated IFN-ß-1a has the potential to improve current therapies in multiple sclerosis, and its ability to potentiate the MHC-Class I expression suggests a clinical benefit in diseases where the downmodulation of the MHC-class I expression plays a role (eg, in immuno-oncology combination therapies or antiviral agents). The present study on IFN-ß deamidation adds a new prospective on deamidation as part of a posttranslational modification code that allows the modulation of the biological properties of proteins. Moreover, it underlines the unique IFN-ß-1a properties that differentiate this molecule from other members of the type I interferon family.


Assuntos
Interferon beta-1a/metabolismo , Interferon beta-1a/farmacologia , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/farmacologia , Antivirais/química , Antivirais/metabolismo , Antivirais/farmacologia , Células CHO , Dicroísmo Circular , Cricetulus , Humanos , Fatores Imunológicos/química , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Interferon beta-1a/química , Oxirredução , Fragmentos de Peptídeos , Desnaturação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA