Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Transl Med ; 17(1): 82, 2019 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-30871636

RESUMO

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.


Assuntos
Interferon alfa-2/farmacologia , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Monócitos/metabolismo , Polietilenoglicóis/farmacologia , Animais , Contagem de Células , Morte Celular/efeitos dos fármacos , Separação Celular , Sobrevivência Celular/efeitos dos fármacos , Criopreservação , Feminino , Humanos , Interferon alfa-2/toxicidade , Interferon-alfa/toxicidade , Interferon gama/toxicidade , Camundongos , Monócitos/efeitos dos fármacos , Polietilenoglicóis/toxicidade , Estabilidade Proteica/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/toxicidade
2.
J Virol ; 91(5)2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27974568

RESUMO

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.


Assuntos
Vírus da Dengue/fisiologia , Imunidade Inata , RNA Helicases/metabolismo , Proteínas de Ligação a RNA/fisiologia , Ativação Transcricional/imunologia , Células A549 , Aedes , Animais , Chlorocebus aethiops , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Transporte Proteico , Regulação para Cima , Células Vero , Replicação Viral
3.
J Transl Med ; 16(1): 196, 2018 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-30012146

RESUMO

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.


Assuntos
Interferon alfa-2/administração & dosagem , Interferon alfa-2/uso terapêutico , Interferon-alfa/administração & dosagem , Interferon-alfa/uso terapêutico , Interferon gama/administração & dosagem , Interferon gama/uso terapêutico , Monócitos/metabolismo , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/uso terapêutico , Relação Dose-Resposta a Droga , Feminino , Humanos , Injeções Intraperitoneais , Interferon alfa-2/farmacologia , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Monócitos/efeitos dos fármacos , Polietilenoglicóis/farmacologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Resultado do Tratamento
4.
J Infect Dis ; 214(suppl 3): S164-S168, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27707892

RESUMO

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.


Assuntos
Serviços de Laboratório Clínico/organização & administração , Surtos de Doenças , Ebolavirus/isolamento & purificação , Doença pelo Vírus Ebola/diagnóstico , Ebolavirus/genética , Guiné , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/virologia , Humanos , Mali/epidemiologia , Manejo de Espécimes
5.
J Infect Dis ; 214(suppl 3): S303-S307, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27471319

RESUMO

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.


Assuntos
Surtos de Doenças , Epidemias , Doença pelo Vírus Ebola/diagnóstico , Doença pelo Vírus Ebola/epidemiologia , Malária/diagnóstico , Adolescente , Alanina Transaminase/análise , Fosfatase Alcalina/análise , Aspartato Aminotransferases/análise , Química Clínica , Serviços de Laboratório Clínico , Ebolavirus/imunologia , Ebolavirus/isolamento & purificação , Doença pelo Vírus Ebola/virologia , Humanos , Libéria/epidemiologia , Testes de Função Hepática , Malária/epidemiologia , Malária/parasitologia , Masculino , Plasmodium/isolamento & purificação , Plasmodium/metabolismo , Sistemas Automatizados de Assistência Junto ao Leito , gama-Glutamiltransferase/análise
6.
J Infect Dis ; 214(suppl 3): S169-S176, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27333914

RESUMO

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.


Assuntos
Serviços de Laboratório Clínico/organização & administração , Ebolavirus/isolamento & purificação , Epidemias/prevenção & controle , Doença pelo Vírus Ebola/epidemiologia , África Ocidental/epidemiologia , Centers for Disease Control and Prevention, U.S. , Feminino , Guiné/epidemiologia , Doença pelo Vírus Ebola/diagnóstico , Doença pelo Vírus Ebola/transmissão , Doença pelo Vírus Ebola/virologia , Humanos , Cooperação Internacional , Libéria/epidemiologia , Masculino , National Institute of Allergy and Infectious Diseases (U.S.) , Segurança , Serra Leoa/epidemiologia , Estados Unidos
7.
Clin Infect Dis ; 63(8): 1026-33, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27531847

RESUMO

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.


Assuntos
Coinfecção , Ebolavirus , Doença pelo Vírus Ebola/complicações , Doença pelo Vírus Ebola/mortalidade , Malária/complicações , Malária/parasitologia , Parasitemia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Criança , Pré-Escolar , Modelos Animais de Doenças , Ebolavirus/genética , Feminino , Doença pelo Vírus Ebola/diagnóstico , Doença pelo Vírus Ebola/epidemiologia , Humanos , Lactente , Recém-Nascido , Malária/diagnóstico , Malária/epidemiologia , Masculino , Camundongos , Pessoa de Meia-Idade , Carga Parasitária , Plasmodium/genética , Taxa de Sobrevida , Carga Viral , Adulto Jovem
8.
Emerg Infect Dis ; 22(2): 331-4, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26812583

RESUMO

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.


Assuntos
Ebolavirus/genética , Doença pelo Vírus Ebola/microbiologia , Nanoporos , Análise de Sequência de DNA/métodos , Surtos de Doenças , Genoma Viral , Doença pelo Vírus Ebola/epidemiologia , Humanos , Mutação
9.
Emerg Infect Dis ; 22(2): 323-6, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26814608

RESUMO

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.


Assuntos
Coinfecção , Surtos de Doenças , Ebolavirus , Doença pelo Vírus Ebola/epidemiologia , Malária/diagnóstico , Malária/parasitologia , Humanos , Malária Falciparum/diagnóstico , Malária Falciparum/parasitologia , Carga Parasitária , Plasmodium falciparum/classificação , Plasmodium falciparum/genética , Prevalência
10.
J Virol ; 89(22): 11534-48, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26355085

RESUMO

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.


Assuntos
Interferon-alfa/metabolismo , Interferon beta/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Infecções por Respirovirus/imunologia , Vírus Sendai/imunologia , Anticorpos Neutralizantes/farmacologia , Linhagem Celular Tumoral , Humanos , Proteínas I-kappa B/metabolismo , Interferon-alfa/classificação , Interferon beta/classificação , Janus Quinases/metabolismo , NF-kappa B/antagonistas & inibidores , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/farmacologia , Receptor de Interferon alfa e beta/imunologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Sulfonas/farmacologia , Tiofenos/farmacologia , Células U937 , Replicação Viral/genética
11.
J Virol ; 87(24): 13094-106, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24027323

RESUMO

Dengue virus (DENV) is a mosquito-transmitted flavivirus that can cause severe disease in humans and is considered a reemerging pathogen of significant importance to public health. The DENV capsid (C) protein functions as a structural component of the infectious virion; however, it may have additional functions in the virus replicative cycle. Here, we show that the DENV C protein interacts and colocalizes with the multifunctional host protein nucleolin (NCL). Furthermore, we demonstrate that this interaction can be disrupted by the addition of an NCL binding aptamer (AS1411). Knockdown of NCL with small interfering RNA (siRNA) or treatment of cells with AS1411 results in a significant reduction of viral titers after DENV infection. Western blotting and quantitative RT-PCR (qRT-PCR) analysis revealed no differences in viral RNA or protein levels at early time points postinfection, suggesting a role for NCL in viral morphogenesis. We support this hypothesis by showing that treatment with AS1411 alters the migration characteristics of the viral capsid, as visualized by native electrophoresis. Here, we identify a critical interaction between DENV C protein and NCL that represents a potential new target for the development of antiviral therapeutics.


Assuntos
Proteínas do Capsídeo/metabolismo , Vírus da Dengue/fisiologia , Dengue/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Replicação Viral , Proteínas do Capsídeo/genética , Dengue/genética , Dengue/virologia , Vírus da Dengue/genética , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Fosfoproteínas/genética , Ligação Proteica , Transporte Proteico , Proteínas de Ligação a RNA/genética , Liberação de Vírus , Nucleolina
12.
Biochim Biophys Acta ; 1823(8): 1378-88, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22683989

RESUMO

Previously we have shown that interferon (IFN)-α induced apoptosis is predominantly mediated by the upregulation of tumor necrosis factor related apoptosis-inducing ligand (TRAIL) via the caspase-8 pathway. It was also shown that recruitment of mitochondria in IFN-α induced apoptosis involves the cleavage of BH3 interacting domain death agonist (Bid) to truncated Bid (tBid). In the present study, we demonstrate that tBid induced by IFN-α2a activates mitochondrial Bak to trigger the loss of mitochondrial membrane integrity, consequently causing release of apoptosis-inducing factor (AIF) in ovarian cancer cells, OVCAR3. AIF translocates from the mitochondria to the nucleus and induces nuclear fragmentation and cell death. Both a small molecule Bid inhibitor (BI-6C9) or Bid-RNA interference (RNAi) preserved mitochondrial membrane potential, prevented nuclear translocation of AIF, and abrogated IFN-α2a-induced cell death. Cell death induced by tBid was inhibited by AIF-RNAi, indicating that caspase-independent AIF signaling is the main pathway through which Bid mediates cell death. This was further supported by experiments showing that BI-6C9 did not prevent the release of cytochrome c from mitochondria to cytosol, while the release of AIF was prevented. In conclusion, IFN-α2a-induced apoptosis is mediated via the mitochondria-associated pathway involving the cleavage of Bid followed by AIF release that involves Bak activation and translocation of AIF from the mitochondria to the nucleus in OVCAR3 cells.


Assuntos
Fator de Indução de Apoptose/metabolismo , Apoptose , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Interferon-alfa/fisiologia , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Caspase 9/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Citocromos c/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Interferon-alfa/farmacologia , Potencial da Membrana Mitocondrial , Neoplasias Ovarianas , Multimerização Proteica , Transporte Proteico , Interferência de RNA , Proteína X Associada a bcl-2/metabolismo
13.
Blood ; 118(9): 2567-77, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21757613

RESUMO

IFNα, a cytokine with multiple functions in innate and adaptive immunity and a potent inhibitor of HIV, exerts antiviral activity, in part, by enhancing apolipoprotein B mRNA-editing enzyme-catalytic polypeptide-like 3 (APOBEC3) family members. Although IFNα therapy is associated with reduced viral burden, this cytokine also mediates immune dysfunction and toxicities. Through detailed mapping of IFNα receptor binding sites, we generated IFNα hybrids and mutants and determined that structural changes in the C-helix alter the ability of IFN to limit retroviral activity. Selective IFNα constructs differentially block HIV replication and their directional magnitude of inhibition correlates with APOBEC3 levels. Importantly, certain mutants exhibited reduced toxicity as reflected by induced indoleamine 2,3-dioxygenase (IDO), suggesting discreet and shared intracellular signaling pathways. Defining IFN structure and function relative to APOBEC and other antiviral genes may enable design of novel IFN-related molecules preserving beneficial antiviral roles while minimizing negative effects.


Assuntos
Citosina Desaminase/biossíntese , HIV-1/fisiologia , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Interferon-alfa/química , Macrófagos/imunologia , Replicação Viral/fisiologia , Desaminases APOBEC , Calmodulina/fisiologia , Citidina Desaminase , Citosina Desaminase/genética , Regulação da Expressão Gênica , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Interferon-alfa/genética , Interferon-alfa/fisiologia , Macrófagos/virologia , Modelos Moleculares , NF-kappa B/fisiologia , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Receptor de Interferon alfa e beta/química , Receptor de Interferon alfa e beta/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Relação Estrutura-Atividade
14.
J Immunol ; 186(3): 1685-93, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21178011

RESUMO

Type I (e.g., IFN-α, IFN-ß) and type II IFNs (IFN-γ) have antiviral, antiproliferative, and immunomodulatory properties. Both types of IFN signal through the Jak/STAT pathway to elicit antiviral activity, yet IFN-γ is thought to do so only through STAT1 homodimers, whereas type I IFNs activate both STAT1- and STAT2-containing complexes such as IFN-stimulated gene factor 3. In this study, we show that IFN-stimulated gene factor 3 containing unphosphorylated STAT2 (ISGF3(II)) also plays a role in IFN-γ-mediated antiviral activity in humans. Using phosphorylated STAT1 as a marker for IFN signaling, Western blot analysis of IFN-α2a-treated human A549 cells revealed that phospho-STAT1 (Y701) levels peaked at 1 h, decreased by 6 h, and remained at low levels for up to 48 h. Cells treated with IFN-γ showed a biphasic phospho-STAT1 response with an early peak at 1-2 h and a second peak at 15-24 h. Gene expression microarray following IFN-γ treatment for 24 h indicated an induction of antiviral genes that are induced by IFN-stimulated gene factor 3 and associated with a type I IFN response. Induction of these genes by autocrine type I and type III IFN signaling was ruled out using neutralizing Abs to these IFNs in biological assays and by quantitative RT-PCR. Despite the absence of autocrine IFNs, IFN-γ treatment induced formation of ISGF3(II). This novel transcription factor complex binds to IFN-stimulated response element promoter sequences, as shown by chromatin immunoprecipitation analysis of the protein kinase R promoter. STAT2 and IFN regulatory factor 9 knockdown in A549 cells reversed IFN-γ-mediated IFN-stimulated response element induction and antiviral activity, implicating ISGF3(II) formation as a significant component of the cellular response and biological activity of IFN-γ.


Assuntos
Vírus da Encefalomiocardite/imunologia , Regulação Viral da Expressão Gênica/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/fisiologia , Interferon gama/fisiologia , Transdução de Sinais/imunologia , Animais , Linhagem Celular Tumoral , Vírus da Encefalomiocardite/patogenicidade , Perfilação da Expressão Gênica , Humanos , Interferon-alfa/farmacologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação/imunologia , Fator de Transcrição STAT1/biossíntese , Fator de Transcrição STAT1/metabolismo
15.
Nat Genet ; 32 Suppl: 474-9, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12454641

RESUMO

The potential medical applications of microarrays have generated much excitement, and some skepticism, within the biomedical community. Some researchers have suggested that within the decade microarrays will be routinely used in the selection, assessment, and quality control of the best drugs for pharmaceutical development, as well as for disease diagnosis and for monitoring desired and adverse outcomes of therapeutic interventions. Realizing this potential will be a challenge for the whole scientific community, as breakthroughs that show great promise at the bench often fail to meet the requirements of clinicians and regulatory scientists. The development of a cooperative framework among regulators, product sponsors, and technology experts will be essential for realizing the revolutionary promise that microarrays hold for drug development, regulatory science, medical practice and public health.


Assuntos
Análise de Sequência com Séries de Oligonucleotídeos/tendências , Previsões , Genética Médica/tendências , Genômica/tendências , Humanos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Formulação de Políticas , Proteômica/tendências , Pesquisa/tendências , Avaliação da Tecnologia Biomédica , Estados Unidos , United States Food and Drug Administration
16.
Clin Cancer Res ; 29(2): 349-363, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36099324

RESUMO

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.


Assuntos
Monócitos , Neoplasias Ovarianas , Humanos , Feminino , Monócitos/metabolismo , Apoptose , Interferon-alfa/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Imunoterapia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
17.
Cancer Immunol Immunother ; 61(7): 1081-92, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22159517

RESUMO

Interferon-activated monocytes are known to exert cytocidal activity against tumor cells in vitro. Here, we have examined whether a combination of IFN-α2a and IFN-γ and human monocytes mediate significant antitumor effects against human ovarian and melanoma tumor xenografts in mouse models. OVCAR-3 tumors were treated i.t. with monocytes alone, IFN-α2a and IFN-γ alone or combination of all three on day 0, 15 or 30 post-tumor implantation. Mice receiving combination therapy beginning day 15 showed significantly reduced tumor growth and prolonged survival including complete regression in 40% mice. Tumor volumes measured on day 80 in mice receiving combination therapy (206 mm(3)) were significantly smaller than those of mice receiving the IFNs alone (1,041 mm(3)), monocytes alone (1,111 mm(3)) or untreated controls (1,728 mm(3)). Similarly, combination therapy with monocytes and IFNs of much larger tumor also inhibited OVCAR-3 tumor growth. Immunohistochemistry studies showed a large number of activated macrophages (CD31(+)/CD68(+)) infiltrating into OVCAR-3 tumors and higher densities of IL-12, IP10 and NOS2, markers of M1 (classical) macrophages in tumors treated with combination therapy compared to the controls. Interestingly, IFNs-activated macrophages induced apoptosis of OVCAR-3 tumor cells as monocytes alone or IFNs alone did not mediate significant apoptosis. Similar antitumor activity was observed in the LOX melanoma mouse model, but not as profound as seen with the OVCAR-3 tumors. Administration of either mixture of monocytes and IFN-α2a or monocytes and IFN-γ did not inhibit Lox melanoma growth; however, a significant inhibition was observed when tumors were treated with a mixture of monocytes, IFN-α2a and IFN-γ. These results indicate that monocytes and both IFN-α2a and IFN-γ may be required to mediate profound antitumor effect against human ovarian and melanoma tumors in mouse models.


Assuntos
Imunoterapia Adotiva/métodos , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Melanoma/terapia , Monócitos/imunologia , Neoplasias Ovarianas/terapia , Adulto , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Humanos , Imuno-Histoquímica , Interferon alfa-2 , Interferon-alfa/imunologia , Interferon gama/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Masculino , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Ovarianas/imunologia , Distribuição Aleatória , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
FEMS Yeast Res ; 11(2): 168-78, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21166768

RESUMO

Secretion of recombinant proteins is a common strategy for heterologous protein expression using the yeast Kluyveromyces lactis. However, a common problem is degradation of a target recombinant protein by secretory pathway aspartyl proteases. In this study, we identified five putative pfam00026 aspartyl proteases encoded by the K. lactis genome. A set of selectable marker-free protease deletion mutants was constructed in the prototrophic K. lactis GG799 industrial expression strain background using a PCR-based dominant marker recycling method based on the Aspergillus nidulans acetamidase gene (amdS). Each mutant was assessed for its secretion of protease activity, its health and growth characteristics, and its ability to efficiently produce heterologous proteins. In particular, despite having a longer lag phase and slower growth compared with the other mutants, a Δyps1 mutant demonstrated marked improvement in both the yield and the quality of Gaussia princeps luciferase and the human chimeric interferon Hy3, two proteins that experienced significant proteolysis when secreted from the wild-type parent strain.


Assuntos
Ácido Aspártico Proteases/deficiência , Expressão Gênica , Kluyveromyces/enzimologia , Kluyveromyces/metabolismo , Proteínas Recombinantes/metabolismo , Arecaceae/enzimologia , DNA Fúngico/química , DNA Fúngico/genética , Proteínas Fúngicas/genética , Deleção de Genes , Kluyveromyces/genética , Luciferases/metabolismo , Dados de Sequência Molecular , Mutagênese , Análise de Sequência de DNA
19.
Immunotherapy ; 11(6): 483-496, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30860437

RESUMO

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.


Assuntos
Imunoterapia/métodos , Interferon-alfa/uso terapêutico , Interferon gama/uso terapêutico , Neoplasias Ovarianas/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , ADP Ribose Transferases/genética , Animais , Apoptose , Toxinas Bacterianas/genética , Linhagem Celular Tumoral , Terapia Combinada , Modelos Animais de Doenças , Exotoxinas/genética , Feminino , Humanos , Interleucina-4/genética , Camundongos , Camundongos Nus , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais , Fatores de Virulência/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa