Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Braz. J. Pharm. Sci. (Online) ; 58: e18984, 2022. graf
Artículo en Inglés | LILACS | ID: biblio-1364429

RESUMEN

Interferon-ß-1a (INF-ß-1a) has gained significant attention due to its emerging applications in the treatment of different human diseases. Therefore, many researchers have attempted to produce it in large quantities and also in a biologically active form using different expression systems. In the present study, we aimed to improve the expression level of INF-ß-1a by Pichia pastoris using optimization of culture conditions. The codon-optimized INF-ß- 1a gene was cloned into pPICZαA plasmid under the control of alcohol oxidase I (AOX1) promoter. The protein expression was induced using different concentrations of methanol at different pHs and temperatures. The biological activity of produced protein was evaluated by anti-proliferative assay. The ideal culture conditions for the expression of INF-ß-1a by P. pastoris were found to be induction with 2% methanol at pH 7.0 culture medium at 30 C which yielded a concentration of 15.5 mg/L INF-ß-1a in a shake flask. Our results indicate that differences in glycosylation pattern could result in different biological activities as INF- ß-1a produced by P. pastoris could significantly more reduce the cell viability of HepG-2 cells, a hepatocellular carcinoma cell line, than a commercially available form of this protein produced by CHO


Asunto(s)
Pichia/clasificación , Interferón beta/agonistas , Carcinoma Hepatocelular/patología , Optimización de Procesos , Codón , Células , Carcinoma Hepatocelular , Concentración de Iones de Hidrógeno
2.
Rev. chil. enferm. respir ; 37(2): 139-148, jun. 2021.
Artículo en Español | LILACS | ID: biblio-1388143

RESUMEN

Resumen Desde la notificación de la pandemia por SARS-CoV-2, agente patógeno responsable del COVID-19, muchos de los tratamientos dirigidos a su manejo han estado sometidos a estudios de manera constante, con el fin de comprobar su eficacia y seguridad. El conocimiento de su virología y etiopatogenia posibilitaría objetivar los pasos moleculares específicos que puedan ser blancos terapéuticos de variados fármacos actualmente disponibles. Esta experiencia proviene principalmente de las infecciones por SARS-CoV y MERS-CoV, con resultados variados 'in vitro' en el SARS-CoV-2, sin evidencia clínica que demuestre efectividad y seguridad de dichos tratamientos. A la fecha, no se ha podido concretar con claridad un esquema de tratamiento específico, debido a que la evidencia surgida ha puesto en jaque cada uno de los fármacos propuestos. Esto ha motivado a continuar en la búsqueda de una estrategia efectiva que permita manejar esta pandemia con la seguridad y eficacia necesaria para que el beneficio terapéutico esté por sobre los posibles efectos adversos que estos esquemas farmacológicos pudiesen presentar. La siguiente revisión pretende mostrar la evidencia disponible a la fecha, definiendo la actividad de cada fármaco en función de su mecanismo de acción.


Since the beginning of the pandemic by SARS-CoV-2, the pathogen responsible for COVID-19, many of the therapeutic options for its management have been under constant revision, in order to verify their safety and efficiency. Knowledge of the viral structure and pathogenesis make it possible to determine the molecular pathways that may be targeted with current available drugs. The experience with these drugs comes mainly from infections caused by SARS-CoV and MERS-CoV, in vitro studies with SARS-CoV-2 that yield variable results, and clinical experience that does not ensure effectiveness and safety of such drugs. To date, it has not been possible to elucidate a specific treatment scheme, because of the constant release of evidence that challenges the usefulness of the proposed drugs. This has motived us to continue seeking for an effective strategy that allows to manage this pandemic in a safe and efficient manner, so that therapeutic benefit surpasses the related adverse drug reactions that can occur. The following review aims to showcase the evidence available to date by defining the activity of each drug based on its mechanism of action.


Asunto(s)
Humanos , Antivirales/administración & dosificación , SARS-CoV-2/efectos de los fármacos , COVID-19/tratamiento farmacológico , Plasma , Ivermectina/administración & dosificación , Adenosina Monofosfato/análogos & derivados , Cloroquina/administración & dosificación , Interleucina-6/antagonistas & inhibidores , Interleucina-1/antagonistas & inhibidores , Interferón beta/administración & dosificación , Corticoesteroides/administración & dosificación , Ritonavir/administración & dosificación , Alanina/análogos & derivados , Lopinavir/administración & dosificación , Anticoagulantes/administración & dosificación
3.
Rev. méd. Maule ; 36(2): 15-23, dic. 2020. tab
Artículo en Español | LILACS | ID: biblio-1344586

RESUMEN

INTRODUCTION: Multiple sclerosis (MS) is a demyelinating disease of the central nervous system, which occurs in up to 85% of cases as relapsing remitting (RR), with episodes of neurological dysfunction partially forwarded. Its treatment in Chile is financially protected by the Explicit Health Guarantees (GES) and Law 20,850 on high-cost diseases. The Regional Hospital of Talca (HRT) has 25 patients benefiting from Law 20,850 in treatment with second-line biologic therapy. Adverse reactions (RAM) to the use of these drugs have been described and to date there are no case reports or studies of significant adverse events in Chile. Objectives: To present the experience of the use of biologic therapy in EMRR in HRT, in relation to adverse events. METHODS: A review of the current guidelines in Chile for the treatment of relapsing-remitting multiple sclerosis and the protocol of law 20,850 was carried out, the clinical records of 25 patients benefiting from the law in the HRT were reviewed, with emphasis on the adverse events presented before First and second line therapies and the con sequences of these events on the continuity of therapy. RESULTS: Half of the patients who started their treatment with first-line drugs had adverse effects, of which 28% involved a change in therapy, the remaining changed from therapy due to failure to treatment. Of the 26 patients included in the sample, 24 are currently using second-line drugs. The profile of adverse effects should be a variable to consider when indicating a therapy for MS.


Asunto(s)
Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Anciano , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/tratamiento farmacológico , Chile , Interferón beta/administración & dosificación , Interferón beta/efectos adversos , Guías de Práctica Clínica como Asunto , Acetato de Glatiramer/efectos adversos , Inmunosupresores , Esclerosis Múltiple/complicaciones
4.
Buenos Aires; IECS; 13 oct. 2020.
No convencional en Español | LILACS, BRISA | ID: biblio-1140945

RESUMEN

CONTEXTO CLÍNICO: La Enfermedad por el Coronavirus 2019 (COVID­19, por su sigla en inglés Coronavirus Disease 2019) es una enfermedad respiratoria de humanos producida por un nuevo coronavirus identificado con la sigla SARS-CoV-2. El 11 de marzo de 2020 la Organización Mundial de la Salud (OMS) declaro la COVID-19 como uma pandemia. Desde ese momento hasta este octubre 2020 su circulación se ha reportado en 205 países reportándose más de 36 millones de casos y más de un millón de muertes. El período de incubación de la infección por 2019­nCoV es de 2 a 14 días. La mayor parte de los contagios se producen persona a persona, siendo altamente transmisible. La clínica varía desde casos asintomáticos a cuadros febriles con tos y dificultad respiratoria, neumonía y distrés respiratorio. También puede acompañarse de alteraciones gastrointestinales.2 En los casos con mal pronóstico, el paciente presenta un importante deterioro respiratorio en 4-8 días. Las imágenes radiológicas muestran generalmente neumonía focal o generalizada semejante al síndrome de distress respiratório agudo. La mayoría de los casos graves requieren ingreso hospitalario, siendo mayoritariamente casos primarios en pacientes de edad avanzada y con comorbilidades (diabetes, enfermedad crónica renal, hipertensión, enfermedad cardiaca y enfermedad pulmonar crónica). La tasa media de letalidad de los pacientes ingresados a UTI es cercana al 49%, siendo los valores más elevados en pacientes masculinos de más de 50 años con comorbilidades múltiples. Actualmente el tratamiento de la COVID­19 es sintomático y de sostén no existiendo hasta el momento tratamiento farmacológico específico curativo. Los tratamientos que se han propuestos son: inhibidores de la ARN polimerasa dependiente de ARN (remdesivir, favipiramir), inhibidores de la neuraminidasa (oseltamivir), inhibidores de la protease (lopinavir/ritonavir, desulfura, inhibidores de la enzima convertidora de angiotensina 2, inhibidores de quinasa (imatinib, baricitinib, ribavirin), inmunomoduladores (plasma de convaleciente, anticuerpos ante receptores IL-6 como tocilizumab y otros, dentro de los cuales se incluye el interferón, los glucorticoides y el umifenovir. En otras pandemias con virus SARS y MERS se ha usado el interferón, dado que múltiples estudios in vitro demuestran acción antiviral. Estudios in vitro específicos en el estudio del virus SARS-CoV-2 definen que la cinética del interferón podría ser variable en los distintos grados de enfermedad y sobre todo en la definición que cuando comenzar su uso, dado el desconocimiento de las curvas virales del SARS-CoV-2 y ventanas terapéuticas apropriadas. El uso asociado de interferón con otros antivirales surge de los experiencia de algún beneficio en estúdios realizados en el tratamiento de los virus MERS-CoV. Dada la falta de vacunas o tratamentos específicos para el nuevo coronavirus SARS-CoV-2, se postula el uso de interferón para el tratamento de pacientes COVID-19 positivos. TECNOLOGÍA: Los distintos tipos de Interferones (IFN) tienen acciones inmunomoduladoras y antivirales uniéndose a receptores celulares, realizando procesos transcripcionales que activan elsistema de citoquinas ante infecciones virales. Los IFN se clasifican en tipo I, II y III. Los IFN tipo I (IFN-α y IFN-ß) son los que tienen mayor actividad antiviral. Los IFN producen expresión de genes que estimulan el estado antiviral celular. Este mecanismo es el que los propuso para los ensayos clinicos en los virus MERS-CoV y SARS-CoV. OBJETIVO: El objetivo del presente informe es evaluar la evidencia disponible acerca de la eficacia, seguridad y aspectos relacionados a las políticas de cobertura del uso de interferón beta en infección por COVID-19. MÉTODOS: Se realizó una búsqueda en las principales bases de datos bibliográficas, en buscadores genéricos de internet, y financiadores de salud. Se priorizó la inclusión de revisiones sistemáticas (RS), ensayos clínicos controlados aleatorizados (ECAs), evaluaciones de tecnologías sanitarias (ETS), evaluaciones económicas, guías de práctica clínica (GPC) y recomendaciones de diferentes organizaciones de salud. CONCLUSIONES: No se ha encontrado evidencia del uso de interferón como mono droga para el tratamiento de pacientes con infección por COVID-19. Evidencia de muy baja calidad proveniente de un ensayo clínico de pocos pacientes y varios sesgos que evaluó el uso de interferón asociado a otros antivirales (lopinavir-ritonavir o atazanavir-ritonavir) en comparación con cualquiera de estas asociaciones, no ha demostrado una reducción de la mortalidad ni mejora clínica a los 28 días en pacientes con neumopatía severa sintomática por COVID-19. Ninguna de las guías o protocolos gubernamentales relevados y/o de sociedades científicas americanas, latinoamericanas y europeas recomiendan el uso interferón como mono droga o uso combinado con otros antivirales. Solo está autorizado su uso en el contexto de investigación clínica. Algunos ensayos clínicos aleatorizados que evalúan la eficacia y seguridad de esta droga em pacientes con cuadros respiratorios por COVID-19 se encuentran en curso, por lo que podrían cambiar la evidencia actualmente disponible.


Asunto(s)
Humanos , Neumonía Viral/tratamiento farmacológico , Interferón beta/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Betacoronavirus/efectos de los fármacos , Evaluación de la Tecnología Biomédica , Análisis Costo-Beneficio
5.
Brasília; s.n; 6 jul. 2020. 31 p.
No convencional en Portugués | BRISA, LILACS, PIE | ID: biblio-1117629

RESUMEN

O Informe Diário de Evidências é uma produção do Ministério da Saúde que tem como objetivo acompanhar diariamente as publicações científicas sobre tratamento farmacológico e vacinas para a COVID-19. Dessa forma, são realizadas buscas estruturadas em bases de dados biomédicas, referente ao dia anterior desse informe. Não são incluídos estudos pré-clínicos (in vitro, in vivo, in silico). A frequência dos estudos é demonstrada de acordo com a sua classificação metodológica (revisões sistemáticas, ensaios clínicos randomizados, coortes, entre outros). Para cada estudo é apresentado um resumo com avaliação da qualidade metodológica. Essa avaliação tem por finalidade identificar o grau de certeza/confiança ou o risco de viés de cada estudo. Para tal, são utilizadas ferramentas já validadas e consagradas na literatura científica, na área de saúde baseada em evidências. Cabe ressaltar que o documento tem caráter informativo e não representa uma recomendação oficial do Ministério da Saúde sobre a temática. Foram encontrados 17 artigos.


Asunto(s)
Humanos , Neumonía Viral/tratamiento farmacológico , Infecciones por Coronavirus/tratamiento farmacológico , Betacoronavirus/efectos de los fármacos , Evaluación de la Tecnología Biomédica , gammaglobulinas/uso terapéutico , Inmunoglobulinas/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Vacunas/uso terapéutico , Cloroquina/uso terapéutico , Interferón beta/uso terapéutico , Aldehído Reductasa/antagonistas & inhibidores , Corticoesteroides/uso terapéutico , Azitromicina/uso terapéutico , Sulfato de Zinc/uso terapéutico , Ritonavir/uso terapéutico , Oseltamivir/uso terapéutico , Lopinavir/uso terapéutico , Hidroxicloroquina/uso terapéutico
6.
Braz. j. med. biol. res ; 53(3): e8876, 2020. graf
Artículo en Inglés | LILACS | ID: biblio-1089338

RESUMEN

The immune stimulatory and anti-neoplastic functions of type I interferon have long been applied for the treatment of melanoma. However, the systemic application of high levels of this recombinant protein is often met with toxicity. An approach that provides localized, yet transient, production of type I interferon may overcome this limitation. We propose that the use of mesenchymal stem cells (MSCs) as delivery vehicles for the production of interferon-β (IFNβ) may be beneficial when applied together with our cancer gene therapy approach. In our previous studies, we have shown that adenovirus-mediated gene therapy with IFNβ was especially effective in combination with p19Arf gene transfer, resulting in immunogenic cell death. Here we showed that MSCs derived from mouse adipose tissue were susceptible to transduction with adenovirus, expressed the transgene reliably, and yet were not especially sensitive to IFNβ production. MSCs used to produce IFNβ inhibited B16 mouse melanoma cells in a co-culture assay. Moreover, the presence of p19Arf in the B16 cells sensitizes them to the IFNβ produced by the MSCs. These data represent a critical demonstration of the use of MSCs as carriers of adenovirus encoding IFNβ and applied as an anti-cancer strategy in combination with p19Arf gene therapy.


Asunto(s)
Animales , Masculino , Conejos , Melanoma Experimental/terapia , Interferón beta/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/administración & dosificación , Células Madre Mesenquimatosas/metabolismo , Transducción Genética , Melanoma Experimental/metabolismo , Terapia Genética , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
7.
Arq. neuropsiquiatr ; 77(3): 166-173, Mar. 2019. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1001345

RESUMEN

ABSTRACT It is currently unknown how genetic factors may influence the clinical course of multiple sclerosis (MS). Objective: We examined the impact of CIITA polymorphisms −168A/G (rs3087456) and +1614G/C (rs4774) on the risk of disability progression, severity and on responses to first-line immunomodulator treatments. Methods: Genomic DNA was extracted from blood samples. We used ABI3730xl and GeneMapper v.4.0 software to identify genotype variations. All patients were followed up and clinically reassessed at three-month intervals. Disability progression was measured by the Expanded Disability Status Scale and disease severity by the Multiple Sclerosis Spasticity Scale (MSSS). Results: We included 37 men and 80 women. We found no evidence regarding the influence of the single nucleotide polymorphisms studied in the Expanded Disability Status Scale or therapeutic response of the evaluated drugs. We performed a logistic regression analysis with the MSSS and found that a less severe MS course was associated with wild type CIITA −168AA and CIITA +1614GG, as the chance of the patient progressing to MSSS2 and MSSS3 decreased in 61% and 75% with CIITA −168AA and 66% and 75% with CIITA +1614GG, respectively (p < 0.0001). Although less significant, the CIITA +1614 GC also pointed to a less severe MS course and the chance of the patient progressing to MSSS3 decreased 79% (p = 0.015). We also observed that the CIITA −168GG genotype was more frequent in MSSS2 and MSSS3 and had 40% lower odds ratio to becoming more severe MS. Conclusion: These data suggest that CIITA −168AA, CIITA +1614GG and CIITA +1614 GC polymorphisms may be associated with a better MS clinical course. This knowledge may be useful for a better understanding of MS and its therapeutic management.


RESUMO Atualmente não se sabe como os fatores genéticos podem influenciar o curso clínico da esclerose múltipla (EM). Objetivo: Examinamos o impacto dos polimorfismos CIITA −168A/G (rs3087456) e CIITA +1614G/C (rs4774) no risco de progressão da incapacidade, gravidade e resposta aos tratamentos imunomoduladores de primeira linha. Métodos: O DNA genômico foi extraído de amostras de sangue. Utilizamos o software ABI3730xl e GeneMapper v.4.0 (Applied Biosystems) para identificar variações genotípicas. Todos os pacientes foram acompanhados e reavaliados clinicamente em intervalos de três meses. A progressão da incapacidade foi medida pela EDSS e a gravidade da doença pelo MSSS. Resultados: Incluímos 37 homens e 80 mulheres. Não encontramos evidências sobre a influência dos SNPs estudados no EDSS e na resposta terapêutica aos fármacos avaliados. Realizamos uma análise de regressão logística com o MSSS e observamos uma evolução menos grave da EM associada aos tipos selvagens CIITA −168AA e CIITA +1614GG, pois a chance do paciente atingir MSSS2 e MSSS3 diminuiu em 61%/75%, e 66/75% respectivamente (p < 0,0001). Embora menos significativo, o CIITA +1614GC também foi relacionado com evolução menos grave da EM e a chance do paciente atingir o MSSS3 diminuiu 79% (p = 0,015). Nós também observamos que o genótipo CIITA −168GG foi mais frequente no MSSS2 e MSSS3 e teve uma razão de chance 40% menor para atingir forma mais grave da EM. Conclusão: Estes dados sugerem que os polimorfismos CIITA −168AA, CIITA +1614GG e CIITA +1614GC podem estar associados a um melhor curso clínico da EM. Este conhecimento pode ser útil para uma melhor compreensão da EM e o seu manejo terapêutico.


Asunto(s)
Humanos , Masculino , Femenino , Adolescente , Adulto , Persona de Mediana Edad , Anciano , Adulto Joven , Proteínas Nucleares/genética , Transactivadores/genética , Progresión de la Enfermedad , Polimorfismo de Nucleótido Simple/genética , Esclerosis Múltiple/genética , Factores de Tiempo , Índice de Severidad de la Enfermedad , Modelos Logísticos , Estudios Retrospectivos , Interferón beta/uso terapéutico , Evaluación de la Discapacidad , Estimación de Kaplan-Meier , Estudios de Asociación Genética , Acetato de Glatiramer/uso terapéutico , Frecuencia de los Genes , Genotipo , Factores Inmunológicos/uso terapéutico , Esclerosis Múltiple/mortalidad , Esclerosis Múltiple/tratamiento farmacológico
8.
Journal of Clinical Neurology ; : 186-190, 2018.
Artículo en Inglés | WPRIM | ID: wpr-714337

RESUMEN

BACKGROUND AND PURPOSE: Patients treated with interferon-beta (IFN-β) can develop neutralizing antibodies (NAbs) against IFN-β that can negatively affect the therapeutic response. This study assessed the prevalence of NAbs and the impact of NAb positivity on the therapeutic response to IFN-β in Korean patients with multiple sclerosis (MS). METHODS: This was a multicenter study involving 150 MS patients from 9 Korean medical centers who were treated with IFN-β for at least 6 months. Sera that had not been influenced by acute treatment were assessed for NAbs using a luciferase reporter gene assay. To evaluate the association between persistent positivity for NAbs and disease activity, NAbs were tested at 2 different time points in 75 of the 150 patients. Disease activity was defined as the presence of clinical exacerbations and/or active MRI lesions during a 1-year follow-up after NAb positivity was confirmed. RESULTS: NAbs were found in 39 of the 150 (26%) MS patients: 30 of the 85 (35%) who were treated with subcutaneous IFN-β-1b, 9 of the 60 (15%) who were treated with subcutaneous IFN-β-1a, and 0 of the 5 (0%) who were treated with intramuscular IFN-β-1a. Thirty of the 39 patients exhibiting NAb positivity were tested at different time points, and 20 of them exhibited persistent NAb positivity. Disease activity was observed more frequently in patients with persistent NAb positivity than in those with transient positivity or persistent negativity [16/20 (80%) vs. 4/55 (7%), respectively; p < 0.001]. When disease activity was compared between patients with persistent and transient NAb positivity, the difference was unchanged and remained statistically significant [16/20 (80%) vs. 2/10 (20%), p=0.004]. CONCLUSIONS: These results further support that persistent NAb positivity is associated with disease activity in MS patients treated with IFN-β.


Asunto(s)
Humanos , Anticuerpos Neutralizantes , Estudios de Seguimiento , Genes Reporteros , Interferón beta , Luciferasas , Imagen por Resonancia Magnética , Esclerosis Múltiple , Prevalencia
10.
Protein & Cell ; (12): 799-807, 2018.
Artículo en Inglés | WPRIM | ID: wpr-757994

RESUMEN

Virus infection induces the production of type I interferons (IFNs). IFNs bind to their heterodimeric receptors to initiate downstream cascade of signaling, leading to the up-regulation of interferon-stimulated genes (ISGs). ISGs play very important roles in innate immunity through a variety of mechanisms. Although hundreds of ISGs have been identified, it is commonly recognized that more ISGs await to be discovered. The aim of this study was to identify new ISGs and to probe their roles in regulating virus-induced type I IFN production. We used consensus interferon (Con-IFN), an artificial alpha IFN that was shown to be more potent than naturally existing type I IFN, to treat three human immune cell lines, CEM, U937 and Daudi cells. Microarray analysis was employed to identify those genes whose expressions were up-regulated. Six hundred and seventeen genes were up-regulated more than 3-fold. Out of these 617 genes, 138 were not previously reported as ISGs and thus were further pursued. Validation of these 138 genes using quantitative reverse transcription PCR (qRT-PCR) confirmed 91 genes. We screened 89 genes for those involved in Sendai virus (SeV)-induced IFN-β promoter activation, and PIM1 was identified as one whose expression inhibited SeV-mediated IFN-β activation. We provide evidence indicating that PIM1 specifically inhibits RIG-I- and MDA5-mediated IFN-β signaling. Our results expand the ISG library and identify PIM1 as an ISG that participates in the regulation of virus-induced type I interferon production.


Asunto(s)
Humanos , Células Cultivadas , Biblioteca de Genes , Interferón Tipo I , Metabolismo , Interferón beta , Genética , Metabolismo , Proteínas Proto-Oncogénicas c-pim-1 , Genética , Regulación hacia Arriba
11.
Clinics ; 73(supl.1): e479s, 2018. graf
Artículo en Inglés | LILACS | ID: biblio-952830

RESUMEN

While cancer immunotherapy has gained much deserved attention in recent years, many areas regarding the optimization of such modalities remain unexplored, including the development of novel approaches and the strategic combination of therapies that target multiple aspects of the cancer-immunity cycle. Our own work involves the use of gene transfer technology to promote cell death and immune stimulation. Such immunogenic cell death, mediated by the combined transfer of the alternate reading frame (p14ARF in humans and p19Arf in mice) and the interferon-β cDNA in our case, was shown to promote an antitumor immune response in mouse models of melanoma and lung carcinoma. With these encouraging results, we are now setting out on the road toward translational and preclinical development of our novel immunotherapeutic approach. Here, we outline the perspectives and challenges that we face, including the use of human tumor and immune cells to verify the response seen in mouse models and the incorporation of clinically relevant models, such as patient-derived xenografts and spontaneous tumors in animals. In addition, we seek to combine our immunotherapeutic approach with other treatments, such as chemotherapy or checkpoint blockade, with the goal of reducing dosage and increasing efficacy. The success of any translational research requires the cooperation of a multidisciplinary team of professionals involved in laboratory and clinical research, a relationship that is fostered at the Cancer Institute of Sao Paulo.


Asunto(s)
Humanos , Terapia Genética/métodos , Sistemas de Lectura/genética , Interferón beta/uso terapéutico , Técnicas de Transferencia de Gen , Inmunoterapia/métodos , Neoplasias/terapia , Muerte Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteína p14ARF Supresora de Tumor/genética , Neoplasias/inmunología
12.
Cancer Research and Treatment ; : 79-91, 2017.
Artículo en Inglés | WPRIM | ID: wpr-127967

RESUMEN

PURPOSE: Genetically engineered stem cells may be advantageous for gene therapy against various human cancers due to their inherent tumor-tropic properties. In this study, genetically engineered human neural stem cells (HB1.F3) expressing Escherichia coli cytosine deaminase (CD) (HB1.F3.CD) and human interferon-β (IFN-β) (HB1.F3.CD.IFN-β) were employed against lymph node–derived metastatic colorectal adenocarcinoma. MATERIALS AND METHODS: CD can convert a prodrug, 5-fluorocytosine (5-FC), to active 5-fluorouracil, which inhibits tumor growth through the inhibition of DNA synthesis,while IFN-β also strongly inhibits tumor growth by inducing the apoptotic process. In reverse transcription polymerase chain reaction analysis, we confirmed that HB1.F3.CD cells expressed the CD gene and HB1.F3.CD.IFN-β cells expressed both CD and IFN-β genes. RESULTS: In results of a modified trans-well migration assay, HB1.F3.CD and HB1.F3.CD.IFN-β cells selectively migrated toward SW-620, human lymph node–derived metastatic colorectal adenocarcinoma cells. The viability of SW-620 cells was significantly reduced when co-cultured with HB1.F3.CD or HB1.F3.CD.IFN-β cells in the presence of 5-FC. In addition, it was found that the tumor-tropic properties of these engineered human neural stem cells (hNSCs) were attributed to chemoattractant molecules including stromal cell-derived factor 1, c-Kit, urokinase receptor, urokinase-type plasminogen activator, and C-C chemokine receptor type 2 secreted by SW-620 cells. In a xenograft mouse model, treatment with hNSC resulted in significantly inhibited growth of the tumor mass without virulent effects on the animals. CONCLUSION: The current results indicate that engineered hNSCs and a prodrug treatment inhibited the growth of SW-620 cells. Therefore, hNSC therapy may be a clinically effective tool for the treatment of lymph node metastatic colorectal cancer.


Asunto(s)
Animales , Humanos , Ratones , Adenocarcinoma , Quimiocina CXCL12 , Neoplasias Colorrectales , Citosina Desaminasa , Citosina , ADN , Escherichia coli , Flucitosina , Fluorouracilo , Terapia Genética , Xenoinjertos , Interferón beta , Ganglios Linfáticos , Metástasis Linfática , Células-Madre Neurales , Reacción en Cadena de la Polimerasa , Transcripción Reversa , Células Madre , Activador de Plasminógeno de Tipo Uroquinasa
13.
Mem. Inst. Oswaldo Cruz ; 111(6): 378-384, June 2016. tab, graf
Artículo en Inglés | LILACS | ID: lil-784249

RESUMEN

Dengue is an acute febrile disease caused by the mosquito-borne dengue virus (DENV) that according to clinical manifestations can be classified as asymptomatic, mild or severe dengue. Severe dengue cases have been associated with an unbalanced immune response characterised by an over secretion of inflammatory cytokines. In the present study we measured type I interferon (IFN-I) transcript and circulating levels in primary and secondary DENV infected patients. We observed that dengue fever (DF) and dengue haemorrhagic fever (DHF) patients express IFN-I differently. While DF and DHF patients express interferon-α similarly (52,71 ± 7,40 and 49,05 ± 7,70, respectively), IFN- β were associated with primary DHF patients. On the other hand, secondary DHF patients were not able to secrete large amounts of IFN- β which in turn may have influenced the high-level of viraemia. Our results suggest that, in patients from our cohort, infection by DENV serotype 3 elicits an innate response characterised by higher levels of IFN- β in the DHF patients with primary infection, which could contribute to control infection evidenced by the low-level of viraemia in these patients. The present findings may contribute to shed light in the role of innate immune response in dengue pathogenesis.


Asunto(s)
Humanos , Masculino , Femenino , Adolescente , Adulto , Adulto Joven , Interferón beta/sangre , Dengue Grave/sangre , Enfermedad Aguda , Brasil , Dengue Grave/inmunología
16.
Journal of Clinical Neurology ; : 188-193, 2016.
Artículo en Inglés | WPRIM | ID: wpr-88931

RESUMEN

BACKGROUND AND PURPOSE: Brain lesions involving the cerebral cortex are rarely described in patients with neuromyelitis optica spectrum disorder (NMOSD), in contrast to multiple sclerosis. We investigated cerebral cortex involvement using conventional brain magnetic resonance imaging (MRI) in anti-aquaporin-4 (AQP4)-antibody-positive NMOSD patients. METHODS: The study enrolled 215 NMOSD patients who were seropositive for the anti-AQP4 antibody from 5 referral hospitals, and retrospectively analyzed their demographic, clinical, and MRI findings. Abnormal cerebral cortex lesions on brain MRI were identified by a neuroradiologist and two neurologists using consensus. RESULTS: Most of the 215 enrolled patients (87%) were female. The median age at onset was 22.5 years (range: 15-36 years) and the mean follow-up duration was 123 months. Brain lesions were found in 143 of 194 patients (74%) in whom MRI was performed during follow-up. Brain lesions involving the cerebral cortex were identified in 6 of these 194 patients (3.1%). Five of the patients were female, and the six patients together had a median age of 29 years (range: 15-36 years) at the time of lesion presentation. Three of them showed leptomeningeal enhancement in the lesions. At presentation of the cortex-involving lesions, five of these patients were not being treated at the time of presentation, while the sixth was being treated with interferon-beta. CONCLUSIONS: Although rare, cortical involvement occurs in NMOSD and is commonly combined with leptomeningeal enhancement. We speculate that this occurs only in patients who are not treated appropriately with immunosuppressant drugs.


Asunto(s)
Femenino , Humanos , Encéfalo , Corteza Cerebral , Consenso , Estudios de Seguimiento , Interferón beta , Imagen por Resonancia Magnética , Esclerosis Múltiple , Neuromielitis Óptica , Derivación y Consulta , Estudios Retrospectivos
17.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 335-343, 2016.
Artículo en Inglés | WPRIM | ID: wpr-285266

RESUMEN

Cytosolic retinoic acid-inducible gene I (RIG-I) is an important innate immune RNA sensor and can induce antiviral cytokines, e.g., interferon-β (IFN-β). Innate immune response to hepatitis B virus (HBV) plays a pivotal role in viral clearance and persistence. However, knowledge of the role that RIG-I plays in HBV infection is limited. The woodchuck is a valuable model for studying HBV infection. To characterize the molecular basis of woodchuck RIG-I (wRIG-I), we analyzed the complete coding sequences (CDSs) of wRIG-I, containing 2778 base pairs that encode 925 amino acids. The deduced wRIG-I protein was 106.847 kD with a theoretical isoelectric point (pI) of 6.07, and contained three important functional structures [caspase activation and recruitment domains (CARDs), DExD/H-box helicases, and a repressor domain (RD)]. In woodchuck fibroblastoma cell line (WH12/6), wRIG-I-targeted small interfering RNA (siRNA) down-regulated RIG-I and its downstrean effector-IFN-β transcripts under RIG-I' ligand, 5'-ppp double stranded RNA (dsRNA) stimulation. We also measured mRNA levels of wRIG-I in different tissues from healthy woodchucks and in the livers from woodchuck hepatitis virus (WHV)-infected woodchucks. The basal expression levels of wRIG-I were abundant in the kidney and liver. Importantly, wRIG-I was significantly up-regulated in acutely infected woodchuck livers, suggesting that RIG-I might be involved in WHV infection. These results may characterize RIG-I in the woodchuck model, providing a strong basis for further study on RIG-I-mediated innate immunity in HBV infection.


Asunto(s)
Animales , Línea Celular Tumoral , Clonación Molecular , Proteína 58 DEAD Box , Genética , Alergia e Inmunología , Fibroblastos , Alergia e Inmunología , Patología , Expresión Génica , Hepatitis B , Genética , Alergia e Inmunología , Patología , Virus de la Hepatitis B de la Marmota , Inmunidad Innata , Interferón beta , Genética , Alergia e Inmunología , Punto Isoeléctrico , Riñón , Alergia e Inmunología , Patología , Virología , Hígado , Alergia e Inmunología , Patología , Virología , Marmota , Genética , Alergia e Inmunología , Virología , Sistemas de Lectura Abierta , Dominios Proteicos , ARN Bicatenario , ARN Interferente Pequeño , Genética , Metabolismo , Enfermedades de los Roedores , Genética , Alergia e Inmunología , Patología , Virología
18.
Allergy, Asthma & Immunology Research ; : 489-496, 2015.
Artículo en Inglés | WPRIM | ID: wpr-114296

RESUMEN

PURPOSE: Asthma exacerbation from human rhinovirus (HRV) infection is associated with deficient antiviral interferon (IFN) secretion. Although chronic rhinosinusitis (CRS), an inflammatory upper airway disease, is closely linked to asthma, IFN-beta responses to HRV infections in human nasal epithelial cells (HNECs) from CRS patients remain to be studied. We evaluated inflammatory and antiviral responses to HRV infection in HNECs from CRS patients. METHODS: HNECs, isolated from turbinate tissue of 13 patients with CRS and 14 non-CRS controls, were infected with HRV16 for 4 hours. The HRV titer, LDH activity, production of proinflammatory cytokines and IFN-beta proteins, and expression levels of RIG-I and MDA5 mRNA were assessed at 8, 24, and 48 hours after HRV16 infection. RESULTS: The reduction in viral titer was slightly delayed in the CRS group compared to the non-CRS control group. IL-6 and IL-8 were significantly increased to a similar extent in both groups after HRV infection. In the control group, IFN-beta production and MDA5 mRNA expression were significantly increased at 8 and 24 hours after HRV16 infection, respectively. By contrast, in the CRS group, IFN-beta was not induced by HRV infection; however, HRV-induced MDA5 mRNA expression was increased, but the increase was slightly delayed compared to the non-CRS control group. The RIG-I mRNA level was not significantly increased by HRV16 infection in either group. CONCLUSIONS: HRV-induced secretion of proinflammatory cytokines in CRS patients was not different from that in the non-CRS controls. However, reductions in viral titer, IFN-beta secretion, and MDA5 mRNA expression in response to HRV infection in CRS patients were slightly impaired compared to those in the controls, suggesting that HRV clearance in CRS patients might be slightly deficient.


Asunto(s)
Humanos , Asma , Citocinas , Células Epiteliales , Interferón beta , Interferones , Interleucina-6 , Interleucina-8 , Rhinovirus , ARN Helicasas , ARN Mensajero , Cornetes Nasales
19.
Obstetrics & Gynecology Science ; : 210-216, 2015.
Artículo en Inglés | WPRIM | ID: wpr-125648

RESUMEN

OBJECTIVE: This study was to identify small inhibitory RNAs (siRNAs) that are effective in inhibiting growth of cervical cancer cell lines harboring human papilloma virus (HPV) and to examine how siRNAs interact with interferon beta (IFN-beta) and thimerosal. METHODS: The HPV18-positive HeLa and C-4I cell lines were used. Four types of siRNAs were designed according to their target (both E6 and E7 vs. E6 only) and sizes (21- vs. 27-nucleotides); Ex-18E6/21, Ex-18E6/27, Sp-18E6/21, and Sp-18E6/27. Each siRNA-transfected cells were cultured with or without IFN-b and thimerosal and their viability was measured. RESULTS: The viabilities of HPV18-positive tumor cells were reduced by 21- and 27-nucleotide siRNAs in proportion to the siRNA concentrations. Of the two types of siRNAs, the 27-nucleotide siRNA constructs showed greater inhibitory efficacy. Sp-18E6 siRNAs, which selectively downregulates E6 protein only, were more effective than the E6- and E7-targeting Ex-18E6 siRNAs. siRNAs and IFN-beta showed the synergistic effect to inhibit HeLa cell survival and the effect was proportional to both siRNA and IFN-beta concentrations. Thimerosal in the presence of siRNA exerted a dose-dependent inhibition of C-4I cell survival. Finally, co-treatment with siRNA, IFN-beta, and thimerosal induced the most profound decrease in the viability of both cell lines. CONCLUSION: Long (27-nucleotides) siRNAs targeting E6-E7 mRNAs effectively reduce the viability of HPV18-positive cervical cancer cells and show the synergistic effect in combination with IFN-b and thimerosal. It is necessary to find the rational design of siRNAs and effective co-factors to eradicate particular cervical cancer.


Asunto(s)
Humanos , Línea Celular , Supervivencia Celular , Células HeLa , Interferón beta , Papiloma , ARN , ARN Mensajero , ARN Interferente Pequeño , Timerosal , Neoplasias del Cuello Uterino
20.
Journal of Korean Neurosurgical Society ; : 323-328, 2015.
Artículo en Inglés | WPRIM | ID: wpr-83801

RESUMEN

OBJECTIVE: Malignant gliomas are the most common primary tumors of the central nervous system and the prognosis of patients with gliomas is poor. The combination of interferon-bata (IFN-beta) and temozolomide (TMZ) has shown significant additive antitumor effects in human glioma xenograft models. Considering that the poor survival of patients with human malignant gliomas relates partly to the inability to deliver therapeutic agents to the tumor, the tropism of human bone marrow-derived mesenchymal stem cells (MSC) for malignant gliomas can be exploited to therapeutic advantages. We investigated the combination effects of TMZ and MSCs that secrete IFN-beta on gliomas. METHODS: We engineered human MSCs to secret mouse IFN-beta (MSC-IFN-beta) via adenoviral transduction and confirmed their secretory capacity using enzyme-linked immunosorbent assays. In vitro and in vivo experiments were performed to determine the effects of the combined TMZ and MSC-IFN-beta treatment. RESULTS: In vitro, the combination of MSC-IFN-beta and TMZ showed significantly enhanced antitumor effects in GL26 mouse glioma cells. In vivo, the combined MSC-IFN-beta and TMZ therapy significantly reduced the tumor size and improved the survival rates compared to each treatment alone. CONCLUSION: These results suggest that MSCs can be used as an effective delivery vehicle so that the combination of MSC-IFN-beta and TMZ could be considered as a new option for the treatment of malignant gliomas.


Asunto(s)
Animales , Humanos , Ratones , Médula Ósea , Sistema Nervioso Central , Ensayo de Inmunoadsorción Enzimática , Glioma , Xenoinjertos , Interferón beta , Células Madre Mesenquimatosas , Pronóstico , Tasa de Supervivencia , Tropismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA