Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Clin Cancer Res ; 29(2): 349-363, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36099324

RESUMEN

PURPOSE: Ovarian cancer is the most lethal gynecologic cancer and intrinsically resistant to checkpoint immunotherapies. We sought to augment innate immunity, building on previous work with IFNs and monocytes. PATIENTS AND METHODS: Preclinical experiments were designed to define the mechanisms of cancer cell death mediated by the combination of IFNs α and γ with monocytes. We translated these preclinical findings into a phase I trial of autologous IFN-activated monocytes administered intraperitoneally to platinum-resistant or -refractory ovarian cancer patients. RESULTS: IFN-treated monocytes induced caspase 8-dependent apoptosis by the proapoptotic TRAIL and mediated by the death receptors 4 and 5 (DR4 and DR5, respectively) on cancer cells. Therapy was well tolerated with evidence of clinical activity, as 2 of 9 evaluable patients had a partial response by RECIST criteria, and 1 additional patient had a CA-125 response. Upregulation of monocyte-produced TRAIL and cytokines was confirmed in peripheral blood. Long-term responders had alterations in innate and adaptive immune compartments. CONCLUSIONS: Given the mechanism of cancer cell death, and the acceptable tolerability of the clinical regimen, this platform presents a possibility for future combination therapies to augment anticancer immunity. See related commentary by Chow and Dorigo, p. 299.


Asunto(s)
Monocitos , Neoplasias Ováricas , Humanos , Femenino , Monocitos/metabolismo , Apoptosis , Interferón-alfa/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Inmunoterapia , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
4.
J Transl Med ; 17(1): 82, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30871636

RESUMEN

BACKGROUND: Monocytes are myeloid cells that reside in the blood and bone marrow and respond to inflammation. At the site of inflammation, monocytes express cytokines and chemokines. Monocytes have been shown to be cytotoxic to tumor cells in the presence of pro-inflammatory cytokines such as Interferon Alpha, Interferon Gamma, and IL-6. We have previously shown that monocytes stimulated with both interferons (IFNs) results in synergistic killing of ovarian cancer cells. We translated these observations to an ongoing clinical trial using adoptive cell transfer of autologous monocytes stimulated ex vivo with IFNs and infused into the peritoneal cavity of patients with advanced, chemotherapy resistant, ovarian cancer. Here we describe the optimization of the monocyte elutriation protocol and a cryopreservation protocol of the monocytes isolated from peripheral blood. METHODS: Counter flow elutriation was performed on healthy donors or women with ovarian cancer. The monocyte-containing, RO-fraction was assessed for total monocyte number, purity, viability, and cytotoxicity with and without a cryopreservation step. All five fractions obtained from the elutriation procedure were also assessed by flow cytometry to measure the percent of immune cell subsets in each fraction. RESULTS: Both iterative monocyte isolation using counter flow elutriation or cryopreservation following counter flow elutriation can yield over 2 billion monocytes for each donor with high purity. We also show that the monocytes are stable, viable, and retain cytotoxic functions when cultured with IFNs. CONCLUSION: Large scale isolation of monocytes from both healthy donors and patients with advanced, chemotherapy resistant ovarian cancer, can be achieved with high total number of monocytes. These monocytes can be cryopreserved and maintain viability and cytotoxic function. All of the elutriated cell fractions contain ample immune cells which could be used for other cell therapy-based applications.


Asunto(s)
Interferón alfa-2/farmacología , Interferón-alfa/farmacología , Interferón gamma/farmacología , Monocitos/metabolismo , Polietilenglicoles/farmacología , Animales , Recuento de Células , Muerte Celular/efectos de los fármacos , Separación Celular , Supervivencia Celular/efectos de los fármacos , Criopreservación , Femenino , Humanos , Interferón alfa-2/toxicidad , Interferón-alfa/toxicidad , Interferón gamma/toxicidad , Ratones , Monocitos/efectos de los fármacos , Polietilenglicoles/toxicidad , Estabilidad Proteica/efectos de los fármacos , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/toxicidad
5.
Immunotherapy ; 11(6): 483-496, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30860437

RESUMEN

AIM: We have shown that IL-4 fused to Pseudomonas exotoxin (IL-4-PE) is cytotoxic to ovarian cancer cell lines. The antineoplastic properties of IFN-α, IFN-γ and IL-4-PE have been studied and showed some promise in the clinical trials. Here, we investigated whether the combination of IL-4-PE, IFN-α and IFN-γ will result in increased ovarian cancer cell death in vitro and in vivo. MATERIALS & METHODS: Ovarian cancer cells were tested in vitro to analyze the cytotoxic effects of IL-4-PE, IFN-α and IFN-γ, and the combination of all three. Tumor-bearing xenograft mice were treated with the combination of IL-4-PE, IFN-α and IFN-γ to monitor their overall survival. The JAK/STAT phosphorylation signaling pathways were studied to delineate the mechanism of synergistic antitumor activity. RESULTS: The combination of IL-4-PE with IFN-α and IFN-γ resulted in increased ovarian cancer cell death in vitro and in vivo. Mechanistically, the synergistic antitumor effect was dependent on interferon signaling, but not IL-4-PE signaling as determined by signaling specific chemical inhibitors. The combination therapy induced the activation of critical mediators of apoptosis. CONCLUSION: The combination of IL-4-PE with interferons increased overall survival of mice with human ovarian cancer xenograft. These data suggest that this novel combination could provide a unique approach to treating ovarian cancer.


Asunto(s)
Inmunoterapia/métodos , Interferón-alfa/uso terapéutico , Interferón gamma/uso terapéutico , Neoplasias Ováricas/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , ADP Ribosa Transferasas/genética , Animales , Apoptosis , Toxinas Bacterianas/genética , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Exotoxinas/genética , Femenino , Humanos , Interleucina-4/genética , Ratones , Ratones Desnudos , Proteínas Recombinantes de Fusión/genética , Transducción de Señal , Factores de Virulencia/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
6.
Biologicals ; 56: 67-83, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30150108

RESUMEN

Sessions included an overview of past cell therapy (CT) conferences sponsored by the International Alliance for Biological Standardization (IABS). The sessions highlighted challenges in the field of human pluripotent stem cells (hPSCs) and also addressed specific points on manufacturing, bioanalytics and comparability, tumorigenicity testing, storage, and shipping. Panel discussions complemented the presentations. The conference concluded that a range of new standardization groups is emerging that could help the field, but ways must be found to ensure that these efforts are coordinated. In addition, there are opportunities for regulatory convergence starting with a gap analysis of existing guidelines to determine what might be missing and what issues might be creating divergence. More specific global regulatory guidance, preferably from WHO, would be welcome. IABS and the California Institute for Regenerative Medicine (CIRM) will explore with stakeholders the development of a practical and innovative road map to support early CT product (CTP) developers.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Células Madre Pluripotentes , Pruebas de Carcinogenicidad , Guías como Asunto , Humanos , Control de Calidad , Medicina Regenerativa
7.
J Transl Med ; 16(1): 196, 2018 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-30012146

RESUMEN

BACKGROUND: Ovarian cancer has no definitive second line therapeutic options, and largely recurs in the peritoneal cavity. Locoregional immune therapy using both interferons and monocytes can be used as a novel approach. Interferons have both cytostatic and cytotoxic properties, while monocytes stimulated with interferons have potent cytotoxic properties. Due to the highly immune suppressive properties of ovarian cancer, ex vivo stimulation of autologous patient monocytes with interferons and infusion of all three agents intraperitoneally (IP) can provide a strong pro-inflammatory environment at the site of disease to kill malignant cells. METHODS: Patient monocytes are isolated through counterflow elutriation and stimulated ex vivo with interferons and infused IP through a semi-permanent catheter. We have designed a standard 3 + 3 dose escalation study to explore the highest tolerated dose of interferons and monocytes infused IP in patients with chemotherapy resistant ovarian cancer. Secondary outcome measurements of changes in the peripheral blood immune compartment and plasma cytokines will be studied for correlations of response. DISCUSSION: We have developed a novel immunotherapy focused on the innate immune system for the treatment of ovarian cancer. We have combined the use of autologous monocytes and interferons alpha and gamma for local-regional administration directly into the peritoneal cavity. This therapy is highly unique in that it is the first study of its type using only components of the innate immune system for the locoregional delivery consisting of autologous monocytes and dual interferons alpha and gamma. Trial Registration ClinicalTrials.gov Identifier: NCT02948426, registered on October 28, 2016. https://clinicaltrials.gov/ct2/show/NCT02948426.


Asunto(s)
Interferón alfa-2/administración & dosificación , Interferón alfa-2/uso terapéutico , Interferón-alfa/administración & dosificación , Interferón-alfa/uso terapéutico , Interferón gamma/administración & dosificación , Interferón gamma/uso terapéutico , Monocitos/metabolismo , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Polietilenglicoles/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inyecciones Intraperitoneales , Interferón alfa-2/farmacología , Interferón-alfa/farmacología , Interferón gamma/farmacología , Monocitos/efectos de los fármacos , Polietilenglicoles/farmacología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Resultado del Tratamiento
8.
J Virol ; 91(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27974568

RESUMEN

Dengue virus (DENV) is a member of the genus Flavivirus and can cause severe febrile illness. Here, we show that FLJ11286, which we refer to as IRAV, is induced by DENV in an interferon-dependent manner, displays antiviral activity against DENV, and localizes to the DENV replication complex. IRAV is an RNA binding protein and localizes to cytoplasmic processing bodies (P bodies) in uninfected cells, where it interacts with the MOV10 RISC complex RNA helicase, suggesting a role for IRAV in the processing of viral RNA. After DENV infection, IRAV, along with MOV10 and Xrn1, localizes to the DENV replication complex and associates with DENV proteins. Depletion of IRAV or MOV10 results in an increase in viral RNA. These data serve to characterize an interferon-stimulated gene with antiviral activity against DENV, as well as to propose a mechanism of activity involving the processing of viral RNA. IMPORTANCE Dengue virus, a member of the family Flaviviridae, can result in a life-threatening illness and has a significant impact on global health. Dengue virus has been shown to be particularly sensitive to the effects of type I interferon; however, little is known about the mechanisms by which interferon-stimulated genes function to inhibit viral replication. A better understanding of the interferon-mediated antiviral response to dengue virus may aid in the development of novel therapeutics. Here, we examine the influence of the interferon-stimulated gene IRAV (FLJ11286) on dengue virus replication. We show that IRAV associates with P bodies in uninfected cells and with the dengue virus replication complex after infection. IRAV also interacts with MOV10, depletion of which is associated with increased viral replication. Our results provide insight into a newly identified antiviral gene, as well as broadening our understanding of the innate immune response to dengue virus infection.


Asunto(s)
Virus del Dengue/fisiología , Inmunidad Innata , ARN Helicasas/metabolismo , Proteínas de Unión al ARN/fisiología , Activación Transcripcional/inmunología , Células A549 , Aedes , Animales , Chlorocebus aethiops , Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Transporte de Proteínas , Regulación hacia Arriba , Células Vero , Replicación Viral
9.
J Infect Dis ; 214(suppl 3): S164-S168, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27707892

RESUMEN

Aware of the rapid spread of Ebola virus (EBOV) during the current West African epidemic, Mali took several proactive steps to rapidly identify cases within its borders. Under the Mali International Center for Excellence in Research program, a collaboration between the National Institute of Allergy and Infectious Diseases and the Malian Ministry of Higher Education and Scientific Research established a national EBOV diagnostic site at the University of Sciences, Techniques and Technologies of Bamako in the SEREFO Laboratory. Two separate introductions of EBOV occurred in Mali from neighboring Guinea, but both chains of transmission were quickly halted, and Mali was declared "Ebola free" on 18 January 2015 and has remained so since. The SEREFO Laboratory was instrumental in the success of Mali's Ebola response by providing timely and accurate diagnostics. As of today, the SEREFO Laboratory has tested 103 samples from 88 suspected cases, 10 of which were EBOV positive, since the Ebola diagnostics unit started in April 2014. The establishment of Ebola diagnostics in the SEREFO Laboratory, safety precautions, and diagnostics are described.


Asunto(s)
Servicios de Laboratorio Clínico/organización & administración , Brotes de Enfermedades , Ebolavirus/aislamiento & purificación , Fiebre Hemorrágica Ebola/diagnóstico , Ebolavirus/genética , Guinea , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/virología , Humanos , Malí/epidemiología , Manejo de Especímenes
10.
Clin Infect Dis ; 63(8): 1026-33, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27531847

RESUMEN

BACKGROUND: The ongoing Ebola outbreak in West Africa has resulted in 28 646 suspected, probable, and confirmed Ebola virus infections. Nevertheless, malaria remains a large public health burden in the region affected by the outbreak. A joint Centers for Disease Control and Prevention/National Institutes of Health diagnostic laboratory was established in Monrovia, Liberia, in August 2014, to provide laboratory diagnostics for Ebola virus. METHODS: All blood samples from suspected Ebola virus-infected patients admitted to the Médecins Sans Frontières ELWA3 Ebola treatment unit in Monrovia were tested by quantitative real-time polymerase chain reaction for the presence of Ebola virus and Plasmodium species RNA. Clinical outcome in laboratory-confirmed Ebola virus-infected patients was analyzed as a function of age, sex, Ebola viremia, and Plasmodium species parasitemia. RESULTS: The case fatality rate of 1182 patients with laboratory-confirmed Ebola virus infections was 52%. The probability of surviving decreased with increasing age and decreased with increasing Ebola viral load. Ebola virus-infected patients were 20% more likely to survive when Plasmodium species parasitemia was detected, even after controlling for Ebola viral load and age; those with the highest levels of parasitemia had a survival rate of 83%. This effect was independent of treatment with antimalarials, as this was provided to all patients. Moreover, treatment with antimalarials did not affect survival in the Ebola virus mouse model. CONCLUSIONS: Plasmodium species parasitemia is associated with an increase in the probability of surviving Ebola virus infection. More research is needed to understand the molecular mechanism underlying this remarkable phenomenon and translate it into treatment options for Ebola virus infection.


Asunto(s)
Coinfección , Ebolavirus , Fiebre Hemorrágica Ebola/complicaciones , Fiebre Hemorrágica Ebola/mortalidad , Malaria/complicaciones , Malaria/parasitología , Parasitemia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Ebolavirus/genética , Femenino , Fiebre Hemorrágica Ebola/diagnóstico , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Lactante , Recién Nacido , Malaria/diagnóstico , Malaria/epidemiología , Masculino , Ratones , Persona de Mediana Edad , Carga de Parásitos , Plasmodium/genética , Tasa de Supervivencia , Carga Viral , Adulto Joven
11.
J Interferon Cytokine Res ; 36(11): 652-665, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27508859

RESUMEN

Sendai virus (SeV), a murine paramyxovirus, has been used to study the induction of type I interferon (IFN) subtypes in robust quantities. Few studies have measured whether the IFN that SeV induces actually fulfills its intended purpose of interfering with virus-mediated effects in the cells in which it is produced. We determined the effects of IFN on SeV-mediated cytopathic effects (CPE) and the ability of IFN to protect against virus infection. SeV-induced biologically active IFN resulted in Jak/STAT activation and the production of a number of interferon-stimulated genes (ISGs). However, these responses did not inhibit SeV replication or CPE. This observation was not due to SeV effects on canonical IFN signaling. Furthermore, pretreating cells with type I IFN and establishing an antiviral state before infection did not mediate SeV effects. Therefore, the induction of canonical IFN signaling pathways and ISGs does not always confer protection against the IFN-inducing virus. Because type I IFNs are approved to treat various infections, our findings suggest that typical markers of IFN activity may not be indicative of a protective antiviral response and should not be used alone to determine whether an antiviral state against a particular virus is achieved.


Asunto(s)
Interferón Tipo I/inmunología , Quinasas Janus/genética , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/virología , Factores de Transcripción STAT/genética , Virus Sendai/patogenicidad , Humanos , Quinasas Janus/metabolismo , Infecciones por Respirovirus/inmunología , Factores de Transcripción STAT/metabolismo , Virus Sendai/inmunología , Células Tumorales Cultivadas , Replicación Viral
12.
J Infect Dis ; 214(suppl 3): S303-S307, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27471319

RESUMEN

The development of point-of-care clinical chemistry analyzers has enabled the implementation of these ancillary tests in field laboratories in resource-limited outbreak areas. The Eternal Love Winning Africa (ELWA) outbreak diagnostic laboratory, established in Monrovia, Liberia, to provide Ebola virus and Plasmodium spp. diagnostics during the Ebola epidemic, implemented clinical chemistry analyzers in December 2014. Clinical chemistry testing was performed for 68 patients in triage, including 12 patients infected with Ebola virus and 18 infected with Plasmodium spp. The main distinguishing feature in clinical chemistry of Ebola virus-infected patients was the elevation in alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and γ-glutamyltransferase levels and the decrease in calcium. The implementation of clinical chemistry is probably most helpful when the medical supportive care implemented at the Ebola treatment unit allows for correction of biochemistry derangements and on-site clinical chemistry analyzers can be used to monitor electrolyte balance.


Asunto(s)
Brotes de Enfermedades , Epidemias , Fiebre Hemorrágica Ebola/diagnóstico , Fiebre Hemorrágica Ebola/epidemiología , Malaria/diagnóstico , Adolescente , Alanina Transaminasa/análisis , Fosfatasa Alcalina/análisis , Aspartato Aminotransferasas/análisis , Química Clínica , Servicios de Laboratorio Clínico , Ebolavirus/inmunología , Ebolavirus/aislamiento & purificación , Fiebre Hemorrágica Ebola/virología , Humanos , Liberia/epidemiología , Pruebas de Función Hepática , Malaria/epidemiología , Malaria/parasitología , Masculino , Plasmodium/aislamiento & purificación , Plasmodium/metabolismo , Sistemas de Atención de Punto , gamma-Glutamiltransferasa/análisis
13.
J Infect Dis ; 214(suppl 3): S169-S176, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27333914

RESUMEN

West Africa experienced the first epidemic of Ebola virus infection, with by far the greatest number of cases in Guinea, Sierra Leone, and Liberia. The unprecedented epidemic triggered an unparalleled response, including the deployment of multiple Ebola treatment units and mobile/field diagnostic laboratories. The National Institute of Allergy and Infectious Diseases and the Centers for Disease Control and Prevention deployed a joint laboratory to Monrovia, Liberia, in August 2014 to support the newly founded Ebola treatment unit at the Eternal Love Winning Africa (ELWA) campus. The laboratory operated initially out of a tent structure but quickly moved into a fixed-wall building owing to severe weather conditions, the need for increased security, and the high sample volume. Until May 2015, when the laboratory closed, the site handled close to 6000 clinical specimens for Ebola virus diagnosis and supported the medical staff in case patient management. Laboratory operation and safety, as well as Ebola virus diagnostic assays, are described and discussed; in addition, lessons learned for future deployments are reviewed.


Asunto(s)
Servicios de Laboratorio Clínico/organización & administración , Ebolavirus/aislamiento & purificación , Epidemias/prevención & control , Fiebre Hemorrágica Ebola/epidemiología , África Occidental/epidemiología , Centers for Disease Control and Prevention, U.S. , Femenino , Guinea/epidemiología , Fiebre Hemorrágica Ebola/diagnóstico , Fiebre Hemorrágica Ebola/transmisión , Fiebre Hemorrágica Ebola/virología , Humanos , Cooperación Internacional , Liberia/epidemiología , Masculino , National Institute of Allergy and Infectious Diseases (U.S.) , Seguridad , Sierra Leona/epidemiología , Estados Unidos
14.
Cytokine Growth Factor Rev ; 29: 109-15, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27026228

RESUMEN

Cytokines and cells of the innate immune system have been shown to be critical regulators in the elimination, equilibrium and escape of malignant cells. Despite in vitro and in vivo evidence, components of the innate immune system have shown limited efficacy in the treatment of ovarian cancer. Intraperitoneal immunotherapies are a promising field that has not yet been fully explored in ovarian cancer. Cytokine immunotherapy using interferon alpha (IFN-α) and interferon gamma (IFN-γ) has predominantly been used intraperitoneally in ovarian cancer, with promising results. Early studies also showed that autologous monocytes infused into the peritoneum have anti-tumor properties. Combination therapies have been shown to be more effective in treating cancer than mono-therapies. Based on these observations the combination of cell therapy with cytokine therapy may provide a unique strategy for the treatment of chemotherapy resistant solid cancers.


Asunto(s)
Interferón-alfa/uso terapéutico , Interferón gamma/uso terapéutico , Transfusión de Leucocitos , Monocitos , Neoplasias Ováricas/terapia , Animales , Femenino , Humanos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología
15.
Emerg Infect Dis ; 22(2): 323-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26814608

RESUMEN

Malaria is a major public health concern in the countries affected by the Ebola virus disease epidemic in West Africa. We determined the feasibility of using molecular malaria diagnostics during an Ebola virus disease outbreak and report the incidence of Plasmodium spp. parasitemia in persons with suspected Ebola virus infection.


Asunto(s)
Coinfección , Brotes de Enfermedades , Ebolavirus , Fiebre Hemorrágica Ebola/epidemiología , Malaria/diagnóstico , Malaria/parasitología , Humanos , Malaria Falciparum/diagnóstico , Malaria Falciparum/parasitología , Carga de Parásitos , Plasmodium falciparum/clasificación , Plasmodium falciparum/genética , Prevalencia
16.
Emerg Infect Dis ; 22(2): 331-4, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26812583

RESUMEN

Rapid sequencing of RNA/DNA from pathogen samples obtained during disease outbreaks provides critical scientific and public health information. However, challenges exist for exporting samples to laboratories or establishing conventional sequencers in remote outbreak regions. We successfully used a novel, pocket-sized nanopore sequencer at a field diagnostic laboratory in Liberia during the current Ebola virus outbreak.


Asunto(s)
Ebolavirus/genética , Fiebre Hemorrágica Ebola/microbiología , Nanoporos , Análisis de Secuencia de ADN/métodos , Brotes de Enfermedades , Genoma Viral , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Mutación
17.
J Virol ; 89(22): 11534-48, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26355085

RESUMEN

UNLABELLED: Type I interferons (IFNs) are induced upon viral infection and important mediators of innate immunity. While there is 1 beta interferon (IFN-ß) protein, there are 12 different IFN-α subtypes. It has been reported extensively that different viruses induce distinct patterns of IFN subtypes, but it has not been previously shown how the viral multiplicity of infection (MOI) can affect IFN induction. In this study, we discovered the novel finding that human U937 cells infected with 2 different concentrations of Sendai virus (SeV) induce 2 distinct type I IFN subtype profiles. Cells infected at the lower MOI induced more subtypes than cells infected at the higher MOI. We found that this was due to the extent of signaling through the IFN receptor (IFNAR). The cells infected at the lower viral MOI induced the IFNAR2-dependent IFN-α subtypes 4, 6, 7, 10, and 17, which were not induced in cells infected at higher virus concentrations. IFN-ß and IFN-α1, -2, and -8 were induced in an IFNAR-independent manner in cells infected at both virus concentrations. IFN-α5, -14, -16, and -21 were induced in an IFNAR-dependent manner in cells infected at lower virus concentrations and in an IFNAR-independent manner in cells infected at higher virus concentrations. These differences in IFN subtype profiles in the 2 virus concentrations also resulted in distinct interferon-stimulated gene induction. These results present the novel finding that different viral MOIs differentially activate JAK/STAT signaling through the IFNAR, which greatly affects the profile of IFN subtypes that are induced. IMPORTANCE: Type I IFNs are pleiotropic cytokines that are instrumental in combating viral diseases. Understanding how the individual subtypes are induced is important in developing strategies to block viral replication. Many studies have reported that different viruses induce distinct type I IFN subtype profiles due to differences in the way viruses are sensed in different cell types. However, we report in our study the novel finding that the amount of virus used to infect a system can also affect which type I IFN subtypes are induced due to the extent of activation of certain signaling pathways. These distinct IFN subtype profiles in cells infected at different MOIs are correlated with differences in interferon-stimulated gene induction, indicating that the same virus can induce distinct antiviral responses depending on the MOI. Because type I IFNs are used as therapeutic agents to treat viral diseases, understanding their antiviral mechanisms can enhance clinical treatments.


Asunto(s)
Interferón-alfa/metabolismo , Interferón beta/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Infecciones por Respirovirus/inmunología , Virus Sendai/inmunología , Anticuerpos Neutralizantes/farmacología , Línea Celular Tumoral , Humanos , Proteínas I-kappa B/metabolismo , Interferón-alfa/clasificación , Interferón beta/clasificación , Quinasas Janus/metabolismo , FN-kappa B/antagonistas & inhibidores , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/farmacología , Receptor de Interferón alfa y beta/inmunología , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Sulfonas/farmacología , Tiofenos/farmacología , Células U937 , Replicación Viral/genética
19.
J Interferon Cytokine Res ; 35(1): 55-62, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25068849

RESUMEN

Interferons (IFNs) play an important role in immune surveillance of tumors; however, their efficacy in the treatment of malignancies has been limited. Monocytes are mononuclear phagocytes that are critical to the generation of an innate immune response to tumors. The authors and others have shown that treatment of tumor cell lines in vitro and in vivo with human monocytes primed with type I and type II IFNs results in killing. We now expand on this work, in an extended panel of ovarian cancer cell lines. In this study, we hypothesized that there would be variable sensitivity amongst cell lines to the killing properties of monocytes and IFNs. To this end, we explored the interactions of IFN primed monocytes in conjunction with the standard of therapy for ovarian cancer, taxane, and platinum-based chemotherapeutics. Using 6 ovarian cancer cell lines, we demonstrated that there is variation from cell line to cell line in the ability of IFN-α2a and IFN-γ primed monocytes to synergistically kill target tumor cells, and further, there is an additive killing effect when target cells are treated with both IFN primed monocytes and chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Interferón-alfa/uso terapéutico , Interferón gamma/uso terapéutico , Monocitos Activados Asesinos/inmunología , Neoplasias Ováricas/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica , Carboplatino/uso terapéutico , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Ováricas/inmunología , Paclitaxel/uso terapéutico
20.
J Interferon Cytokine Res ; 34(11): 876-84, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24955567

RESUMEN

Human interleukin-29 (IL-29), a helical cytokine with interferon-like activities, is currently being developed as a clinical biotherapeutic to treat chronic hepatitis C infection and some cancers. As such, the World Health Organization (WHO) has recognized a need for biological standardization of IL-29 and the establishment of an internationally available reference reagent of IL-29. In order to accomplish this, an international collaborative study that evaluates WHO candidate reference reagents of IL-29 was instigated by the National Institute for Biological Standards and Control (NIBSC) in 2010 and was carried out in the succeeding year. Two preparations of human sequence recombinant IL-29, one expressed in murine NS0 cells and the other in Escherichia coli, were formulated and lyophilized at NIBSC before evaluation in the collaborative study for their suitability to serve as a reference reagent. The preparations were tested by 6 laboratories from 4 countries using in vitro bioassays and also evaluated for thermal stability within the NIBSC laboratory. On the basis of the results of the collaborative study, both preparations, 07/212 (NS0-derived) and 10/176 (E. coli-derived) were judged sufficiently active and stable to serve as a reference reagent. However, since IL-29 produced in E. coli is in development for clinical applications, it was recommended that the preparation coded 10/176 be established as the WHO international reference reagent for human IL-29. This recommendation was accepted, and the IL-29 preparation coded 10/176 was formally established by the WHO ECBS at its meeting in October 2012 as the WHO international reference reagent for IL-29 with an assigned unitage of 5,000 reference units per ampoule.


Asunto(s)
Hepatitis C/terapia , Inmunoterapia/métodos , Interleucinas/normas , Neoplasias/terapia , Proteínas Recombinantes/normas , Animales , Línea Celular , Enfermedad Crónica , Escherichia coli/genética , Hepatitis C/inmunología , Humanos , Interferones , Interleucinas/uso terapéutico , Cooperación Internacional , Ratones , Neoplasias/inmunología , Estabilidad Proteica , Proteínas Recombinantes/uso terapéutico , Estándares de Referencia , Organización Mundial de la Salud
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...