Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 15(678): eabo0205, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36630481

RESUMEN

The common γ chain (γc; IL-2RG) is a subunit of the interleukin (IL) receptors for the γc cytokines IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. The lack of appropriate neutralizing antibodies recognizing IL-2RG has made it difficult to thoroughly interrogate the role of γc cytokines in inflammatory and autoimmune disease settings. Here, we generated a γc cytokine receptor antibody, REGN7257, to determine whether γc cytokines might be targeted for T cell-mediated disease prevention and treatment. Biochemical, structural, and in vitro analysis showed that REGN7257 binds with high affinity to IL-2RG and potently blocks signaling of all γc cytokines. In nonhuman primates, REGN7257 efficiently suppressed T cells without affecting granulocytes, platelets, or red blood cells. Using REGN7257, we showed that γc cytokines drive T cell-mediated disease in mouse models of graft-versus-host disease (GVHD) and multiple sclerosis by affecting multiple aspects of the pathogenic response. We found that our xenogeneic GVHD mouse model recapitulates hallmarks of acute and chronic GVHD, with T cell expansion/infiltration into tissues and liver fibrosis, as well as hallmarks of immune aplastic anemia, with bone marrow aplasia and peripheral cytopenia. Our findings indicate that γc cytokines contribute to GVHD and aplastic anemia pathology by promoting these characteristic features. By demonstrating that broad inhibition of γc cytokine signaling with REGN7257 protects from immune-mediated disorders, our data provide evidence of γc cytokines as key drivers of pathogenic T cell responses, offering a potential strategy for the management of T cell-mediated diseases.


Asunto(s)
Anemia Aplásica , Enfermedad Injerto contra Huésped , Subunidad gamma Común de Receptores de Interleucina , Linfocitos T , Animales , Ratones , Anemia Aplásica/metabolismo , Anticuerpos Monoclonales/metabolismo , Citocinas/metabolismo , Enfermedad Injerto contra Huésped/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/patología , Subunidad gamma Común de Receptores de Interleucina/antagonistas & inhibidores , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Primates
2.
Cytokine ; 162: 156091, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36481478

RESUMEN

RATIONALE: Type 2 (T2) asthma is characterized by airflow limitations and elevated levels of blood and sputum eosinophils, fractional exhaled nitric oxide, IgE, and periostin. While eosinophils are associated with exacerbations, the contribution of eosinophils to lung inflammation, remodeling and function remains largely hypothetical. OBJECTIVES: To determine the effect of T2 cytokines IL-4, IL-13 and IL-5 on eosinophil biology and compare the impact of depleting just eosinophils versus inhibiting all aspects of T2 inflammation on airway inflammation. METHODS: Human eosinophils or endothelial cells stimulated with IL-4, IL-13 or IL-5 were assessed for gene changes or chemokine release.Mice exposed to house dust mite extract received anti-IL-4Rα (dupilumab), anti-IL-5 or control antibodies and were assessed for changes in lung histological and inflammatory endpoints. MEASUREMENTS AND MAIN RESULTS: IL-4 or IL-13 stimulation of human eosinophils and endothelial cells induced gene expression changes related to granulocyte migration; whereas, IL-5 induced changes reflecting granulocyte differentiation.In a mouse model, blocking IL-4Rα improved lung function by impacting multiple effectors of inflammation and remodeling, except peripheral eosinophil counts, thereby disconnecting blood eosinophils from airway inflammation, remodeling and function. Blocking IL-5 globally reduced eosinophil counts but did not impact inflammatory or functional measures of lung pathology. Whole lung transcriptome analysis revealed that IL-5 or IL-4Rα blockade impacted eosinophil associated genes, whereas IL-4Rα blockade also impacted genes associated with multiple cells, cytokines and chemokines, mucus production, cell:cell adhesion and vascular permeability. CONCLUSIONS: Eosinophils are not the sole contributor to asthma pathophysiology or lung function decline and emphasizes the need to block additional mediators to modify lung inflammation and impact lung function.


Asunto(s)
Asma , Neumonía , Animales , Humanos , Ratones , Asma/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Endoteliales/metabolismo , Inflamación/metabolismo , Interleucina-13/metabolismo , Pulmón/metabolismo , Neumonía/metabolismo , Interleucina-4/farmacología
3.
Mol Cell Biol ; 42(1): e0046721, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-34723652

RESUMEN

A subset of hospitalized COVID-19 patients, particularly the aged and those with comorbidities, develop the most severe form of the disease, characterized by acute respiratory disease syndrome (ARDS), coincident with experiencing a "cytokine storm." Here, we demonstrate that cytokines which activate the NF-κB pathway can induce activin A. Patients with elevated activin A, activin B, and FLRG at hospital admission were associated with the most severe outcomes of COVID-19, including the requirement for mechanical ventilation, and all-cause mortality. A prior study showed that activin A could decrease viral load, which indicated there might be a risk to giving COVID-19 patients an inhibitor of activin. To evaluate this, the role for activin A was examined in a hamster model of SARS-CoV-2 infection, via blockade of activin A signaling. The hamster model demonstrated that use of an anti-activin A antibody did not worsen the disease and there was no evidence for increase in lung viral load and pathology. The study indicates blockade of activin signaling may be beneficial in treating COVID-19 patients experiencing ARDS.


Asunto(s)
Activinas/sangre , Anticuerpos Monoclonales Humanizados/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Proteínas Relacionadas con la Folistatina/sangre , SARS-CoV-2/efectos de los fármacos , Adulto , Anciano , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , COVID-19/mortalidad , COVID-19/virología , Línea Celular , Células Cultivadas , Cricetinae , Método Doble Ciego , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud/métodos , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , SARS-CoV-2/fisiología , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Tasa de Supervivencia
5.
Nat Microbiol ; 6(1): 73-86, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33340034

RESUMEN

Non-human primate models will expedite therapeutics and vaccines for coronavirus disease 2019 (COVID-19) to clinical trials. Here, we compare acute severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in young and old rhesus macaques, baboons and old marmosets. Macaques had clinical signs of viral infection, mild to moderate pneumonitis and extra-pulmonary pathologies, and both age groups recovered in two weeks. Baboons had prolonged viral RNA shedding and substantially more lung inflammation compared with macaques. Inflammation in bronchoalveolar lavage was increased in old versus young baboons. Using techniques including computed tomography imaging, immunophenotyping, and alveolar/peripheral cytokine response and immunohistochemical analyses, we delineated cellular immune responses to SARS-CoV-2 infection in macaque and baboon lungs, including innate and adaptive immune cells and a prominent type-I interferon response. Macaques developed T-cell memory phenotypes/responses and bystander cytokine production. Old macaques had lower titres of SARS-CoV-2-specific IgG antibody levels compared with young macaques. Acute respiratory distress in macaques and baboons recapitulates the progression of COVID-19 in humans, making them suitable as models to test vaccines and therapies.


Asunto(s)
COVID-19/veterinaria , Callithrix/inmunología , Pulmón/inmunología , Macaca mulatta/inmunología , Enfermedades de los Monos/virología , Papio/inmunología , SARS-CoV-2/inmunología , Inmunidad Adaptativa , Animales , Anticuerpos Antivirales/inmunología , Lavado Broncoalveolar , Líquido del Lavado Bronquioalveolar , COVID-19/diagnóstico por imagen , COVID-19/inmunología , COVID-19/patología , Femenino , Humanos , Inmunidad Celular/inmunología , Inmunoglobulina G/inmunología , Inflamación/patología , Pulmón/virología , Masculino , Enfermedades de los Monos/inmunología , Células Mieloides/inmunología , Carga Viral , Esparcimiento de Virus
6.
Science ; 370(6520): 1110-1115, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33037066

RESUMEN

An urgent global quest for effective therapies to prevent and treat coronavirus disease 2019 (COVID-19) is ongoing. We previously described REGN-COV2, a cocktail of two potent neutralizing antibodies (REGN10987 and REGN10933) that targets nonoverlapping epitopes on the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. In this report, we evaluate the in vivo efficacy of this antibody cocktail in both rhesus macaques, which may model mild disease, and golden hamsters, which may model more severe disease. We demonstrate that REGN-COV-2 can greatly reduce virus load in the lower and upper airways and decrease virus-induced pathological sequelae when administered prophylactically or therapeutically in rhesus macaques. Similarly, administration in hamsters limits weight loss and decreases lung titers and evidence of pneumonia in the lungs. Our results provide evidence of the therapeutic potential of this antibody cocktail.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , COVID-19/terapia , Animales , COVID-19/prevención & control , Combinación de Medicamentos , Macaca mulatta , Mesocricetus
7.
Sci Transl Med ; 12(549)2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32581132

RESUMEN

Monoclonal antibodies that block the programmed cell death 1 (PD-1) checkpoint have revolutionized cancer immunotherapy. However, many major tumor types remain unresponsive to anti-PD-1 therapy, and even among responsive tumor types, most of the patients do not develop durable antitumor immunity. It has been shown that bispecific antibodies activate T cells by cross-linking the TCR/CD3 complex with a tumor-specific antigen (TSA). The class of TSAxCD3 bispecific antibodies have generated exciting results in early clinical trials. We have recently described another class of "costimulatory bispecifics" that cross-link a TSA to CD28 (TSAxCD28) and cooperate with TSAxCD3 bispecifics. Here, we demonstrate that these TSAxCD28 bispecifics (one specific for prostate cancer and the other for epithelial tumors) can also synergize with the broader anti-PD-1 approach and endow responsiveness-as well as long-term immune memory-against tumors that otherwise do not respond to anti-PD-1 alone. Unlike CD28 superagonists, which broadly activate T cells and induce cytokine storm, TSAxCD28 bispecifics display little or no toxicity when used alone or in combination with a PD-1 blocker in genetically humanized immunocompetent mouse models or in primates and thus may provide a well-tolerated and "off the shelf" combination approach with PD-1 immunotherapy that can markedly enhance antitumor efficacy.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Animales , Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD28 , Humanos , Inmunoterapia , Ratones , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1
8.
Sci Transl Med ; 12(525)2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31915305

RESUMEN

T cell activation is initiated upon binding of the T cell receptor (TCR)/CD3 complex to peptide-major histocompatibility complexes ("signal 1"); activation is enhanced by engagement of a second "costimulatory" receptor, such as the CD28 receptor on T cells binding to its cognate ligand(s) on the target cell ("signal 2"). CD3-based bispecific antibodies act by replacing conventional signal 1, linking T cells to tumor cells by binding a tumor-specific antigen (TSA) with one arm of the bispecific and bridging to TCR/CD3 with the other. Although some of these so-called TSAxCD3 bispecifics have demonstrated promising antitumor efficacy in patients with cancer, their activity remains to be optimized. Here, we introduce a class of bispecific antibodies that mimic signal 2 by bridging TSA to the costimulatory CD28 receptor on T cells. We term these TSAxCD28 bispecifics and describe two such bispecific antibodies: one specific for ovarian and the other for prostate cancer antigens. Unlike CD28 superagonists, which broadly activate T cells and resulted in profound toxicity in early clinical trials, these TSAxCD28 bispecifics show limited activity and no toxicity when used alone in genetically humanized immunocompetent mouse models or in primates. However, when combined with TSAxCD3 bispecifics, they enhance the artificial synapse between a T cell and its target cell, potentiate T cell activation, and markedly improve antitumor activity of CD3 bispecifics in a variety of xenogeneic and syngeneic tumor models. Combining this class of CD28-costimulatory bispecific antibodies with the emerging class of TSAxCD3 bispecifics may provide well-tolerated, off-the-shelf antibody therapies with robust antitumor efficacy.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Neoplasias/inmunología , Animales , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Citotoxicidad Inmunológica , Femenino , Células HEK293 , Humanos , Sinapsis Inmunológicas/metabolismo , Activación de Linfocitos/inmunología , Macaca fascicularis , Ratones , Neoplasias/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Sci Immunol ; 5(54)2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33443029

RESUMEN

Deficiency in interleukin-36R (IL-36R) antagonist caused by loss-of-function mutations in IL-36RN leads to DITRA (deficiency of IL-36 receptor antagonist), a rare inflammatory human disease that belongs to a subgroup of generalized pustular psoriasis (GPP). We report a functional genetic mouse model of DITRA with enhanced IL-36R signaling analogous to that observed in patients with DITRA, which provides new insight into our understanding of the IL-36 family of molecules in regulating barrier integrity across multiple tissues. Humanized DITRA-like mice displayed increased skin inflammation in a preclinical model of psoriasis, and in vivo blockade of IL-36R pathway using anti-human IL-36R antibody ameliorated imiquimod-induced skin pathology as both prophylactic and therapeutic treatments. Deeper characterization of the humanized DITRA-like mice revealed that deregulated IL-36R signaling promoted tissue pathology during intestinal injury and led to impairment in mucosal restoration in the repair phase of chronic dextran sulfate sodium (DSS)-induced colitis. Blockade of IL-36R pathway significantly ameliorated DSS-induced intestinal inflammation and rescued the inability of DITRA-like mice to recover from mucosal damage in vivo. Our results indicate a central role for IL-36 in regulating proinflammatory responses in the skin and epithelial barrier function in the intestine, suggesting a new therapeutic potential for targeting the IL-36R axis in psoriasis and at the later stages of intestinal pathology in inflammatory bowel disease.


Asunto(s)
Dermatitis/etiología , Dermatitis/metabolismo , Gastroenteritis/etiología , Gastroenteritis/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Animales , Biomarcadores , Dermatitis/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Gastroenteritis/patología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Piel/metabolismo , Piel/patología
10.
PLoS One ; 13(9): e0203482, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30188946

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) infection is difficult to control because the virus undergoes antigenic variation during infection and also modulates the protective host immune response. Although current vaccines do not provide full protection, they have provided insight into the mechanisms of protection. Live PRRSV vaccines induce partial protection before the appearance of neutralizing antibody, suggesting cell-mediated immunity or other mechanisms may be involved. Herein, we demonstrate recovery from infection is associated with development of cytotoxic T-lymphocytes (CTL) that can kill PRRSV-infected target cells. Initial experiments showed survival of PRRSV-infected monocyte derived macrophage (MDM) targets is reduced when overlaid with peripheral blood mononuclear cells (PBMC) from gilts that had recovered from PRRSV infection. Further studies with PBMC depleted of either CD4+ or CD8+ T-cells and positively selected subpopulations of CD4+ and CD8+ T-cells showed that both CD4+ and CD8+ T-cells were involved in killing. Examination of killing at different time points revealed killing was biphasic and mediated by CTL of different phenotypes. CD4+CD8+high were associated with killing target cells infected for 3-6 hours. CD4+CD8- CTL were associated with killing at 16-24 hours. Thus, all the anti-PRRSV CTL activity in pigs was attributed to two phenotypes of CD4+ cells which is different from the anti-viral CD4-CD8+ CTL phenotype found in most other animals. These findings will be useful for evaluating CTL responses induced by current and future vaccines, guiding to a novel direction for future vaccine development.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Vacunas Virales/inmunología , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Síndrome Respiratorio y de la Reproducción Porcina/patología , Porcinos
11.
PLoS One ; 9(2): e88245, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24505447

RESUMEN

Lyme disease is the most prevalent arthropod borne disease in the US and it is caused by the bacterial spirochete Borrelia burgdorferi (Bb), which is acquired through the bite of an infected Ixodes tick. Vaccine development efforts focused on the von Willebrand factor A domain of the borrelial protein BB0172 from which four peptides (A, B, C and D) were synthesized and conjugated to Keyhole Limpet Hemocyanin, formulated in Titer Max® adjuvant and used to immunize C3H/HeN mice subcutaneously at days 0, 14 and 21. Sera were collected to evaluate antibody responses and some mice were sacrificed for histopathology to evaluate vaccine safety. Twenty-eight days post-priming, protection was evaluated by needle inoculation of half the mice in each group with 10³ Bb/mouse, whereas the rest were challenged with 105Bb/mouse. Eight weeks post-priming, another four groups of similarly immunized mice were challenged using infected ticks. In both experiments, twenty-one days post-challenge, the mice were sacrificed to determine antibody responses, bacterial burdens and conduct histopathology. Results showed that only mice immunized with peptide B were protected against challenge with Bb. In addition, compared to the other the treatment groups, peptide B-immunized mice showed very limited inflammation in the heart and joint tissues. Peptide B-specific antibody titers peaked at 8 weeks post-priming and surprisingly, the anti-peptide B antibodies did not cross-react with Bb lysates. These findings strongly suggest that peptide B is a promising candidate for the development of a new DIVA vaccine (Differentiate between Infected and Vaccinated Animals) for protection against Lyme disease.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/uso terapéutico , Vacunas Bacterianas/uso terapéutico , Borrelia burgdorferi/inmunología , Enfermedad de Lyme/prevención & control , Péptidos/uso terapéutico , Animales , Anticuerpos Antibacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/química , Vacunas Bacterianas/inmunología , Borrelia burgdorferi/química , Femenino , Inmunización , Ixodes/microbiología , Enfermedad de Lyme/inmunología , Enfermedad de Lyme/microbiología , Enfermedad de Lyme/patología , Ratones , Ratones Endogámicos C3H , Péptidos/química , Péptidos/inmunología
12.
J Zoo Wildl Med ; 44(2): 251-61, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23805542

RESUMEN

Recurring infection of reticuloendotheliosis virus (REV), an avian oncogenic gammaretrovirus, has been a major obstacle in attempts to breed and release the endangered Attwater's prairie chicken (Tympanicus cupido attwateri). The aim of this study was to develop a DNA vaccine that protects the birds against REV infection. A plasmid was constructed expressing fusion proteins of REV envelope (env) and VP22 of Gallid herpesvirus 2 or REV gag and VP22. Birds vaccinated with these recombinant plasmids developed neutralizing antibodies; showed delayed replication of virus; and had significantly less infection of lymphocytes, specifically CD4+ lymphocytes. Although the vaccine did not prevent infection, it offered partial protection. Birds in field conditions and breeding facilities could potentially benefit from increased immunity when vaccinated.


Asunto(s)
Galliformes , Productos del Gen gag/inmunología , Virus de la Reticuloendoteliosis Aviar/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , ADN Viral/genética , ADN Viral/aislamiento & purificación , Femenino , Genes Virales , Masculino , Infecciones por Retroviridae/prevención & control , Infecciones por Retroviridae/veterinaria , Infecciones Tumorales por Virus/prevención & control , Infecciones Tumorales por Virus/veterinaria , Vacunas de ADN/inmunología
13.
J Vet Diagn Invest ; 23(4): 826-31, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21908333

RESUMEN

Canine schistosomiasis due to Heterobilharzia americana is a clinically underdiagnosed disease in dogs, which is found primarily in the Gulf Coast and south Atlantic region of the United States. A 3-year-old dog from Texas with a clinical diagnosis of systemic mineralization of unknown origin in the absence of evidence of hypercalcemia was found at necropsy to have severe disseminated H. americana infection involving the liver, pancreas, small and large intestine, lungs, and kidneys. Calcification of many of the large number of H. americana eggs gave the false impression of soft-tissue mineralization on radiographic and ultrasonographic images. Polymerase chain reaction amplification and sequencing of DNA derived from formalin-fixed sections of small intestine and liver, using primers specific for a 487-base pair segment of the H. americana small subunit ribosomal RNA gene, confirmed the presence of H. americana.


Asunto(s)
Calcinosis/veterinaria , Enfermedades de los Perros/parasitología , Trematodos/clasificación , Trematodos/aislamiento & purificación , Infecciones por Trematodos/veterinaria , Animales , Calcinosis/parasitología , Calcinosis/patología , ADN de Helmintos , Enfermedades de los Perros/diagnóstico , Enfermedades de los Perros/patología , Perros , Datos de Secuencia Molecular , ARN Ribosómico 18S/genética , Trematodos/genética , Infecciones por Trematodos/diagnóstico , Infecciones por Trematodos/patología
14.
J Vet Cardiol ; 13(2): 127-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21641896

RESUMEN

A fibrous band connecting the middle of the free edge (nodulus Arantii) of the non-coronary aortic valve cusp to the ascending aorta just above the level of the non-coronary sinus of Valsalva was observed in an asymptomatic, 11-year-old, male Border Collie. The fibrous band was unrelated to the cause of the death in this dog. Such fibrous bands are usually reported in humans with congenital bicuspid aortic valves. To our knowledge, this is the first report of a fibrous band in the aortic valve in a domestic animal.


Asunto(s)
Válvula Aórtica/anomalías , Animales , Válvula Aórtica/patología , Colágeno , Perros , Masculino
15.
Virology ; 399(2): 312-21, 2010 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-20137800

RESUMEN

Marek' disease virus serotype-1, also know as Gallid herpesvirus 2 (GaHV-2), elicits T-cell lymphomas in chickens. The GaHV-2 genome encodes an oncoprotein, Meq, with similarity to the Jun/Fos family of proteins. We have previously shown that Meq homodimers are not sufficient to induce lymphomas in chickens. In this study, we investigated the role of Meq heterodimers in the pathogenicity of GaHV-2 by generating a chimeric meq gene, which contains the leucine zipper region of Fos (meqFos). A recombinant virus containing the meqFos gene in place of parental meq, rMd5-MeqFos, was not capable of transforming chicken lymphocytes, indicating that heterodimerization of Meq alone is not sufficient for transformation. In addition, the recovery of the oncogenic phenotype by a recombinant virus encoding one copy each of MeqGCN (homodimer) and MeqFos (heterodimer) conclusively demonstrates that both homo and heterodimerization of Meq are required for oncogenesis.


Asunto(s)
Transformación Celular Viral , Pollos/virología , Herpesvirus Gallináceo 2/genética , Linfocitos/virología , Proteínas Oncogénicas Virales/metabolismo , Animales , Células Cultivadas , Herpesvirus Gallináceo 2/metabolismo , Leucina Zippers , Enfermedad de Marek/virología , Mutación , Proteínas Oncogénicas Virales/genética , Multimerización de Proteína
16.
Infect Immun ; 78(1): 527-35, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19901065

RESUMEN

Salmonella enterica serotype Typhi, the etiological agent of typhoid fever, produces the Vi capsular antigen, a virulence factor absent in Salmonella enterica serotype Typhimurium. Previous studies suggest that the capsule-encoding viaB locus reduces inflammatory responses in intestinal tissue; however, there are currently no data regarding the in vivo expression of this locus. Here we implemented direct and indirect methods to localize and detect Vi antigen expression within polarized intestinal epithelial cells and in the bovine ileal mucosa. We report that tviB, a gene necessary for Vi production in S. Typhi, was significantly upregulated during invasion of intestinal epithelial cells in vitro. During infection of bovine ligated loops, tviB was expressed at levels significantly higher in calf tissue than those in the inoculum. The presence of the Vi capsular antigen was detected in calf ileal tissue via fluorescence microscopy. Together, these results support the concept that expression of the Vi capsular antigen is induced when S. Typhi transits from the intestinal lumen into the ileal mucosa.


Asunto(s)
Regulación Bacteriana de la Expresión Génica/fisiología , Íleon/microbiología , Mucosa Intestinal/microbiología , Polisacáridos Bacterianos/metabolismo , Salmonella typhi/metabolismo , Animales , Bovinos , Línea Celular Tumoral , Eliminación de Gen , Humanos , Polisacáridos Bacterianos/genética , Regiones Promotoras Genéticas/fisiología , Regulación hacia Arriba , Equilibrio Hidroelectrolítico
17.
Virus Res ; 142(1-2): 57-67, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19189855

RESUMEN

Gallid herpesvirus 2 (GaHV-2), commonly known as Marek's disease virus serotype-1 (MDV-1), causes T cell lymphomas in chickens. Vaccines prepared from the attenuated CVI988/Rispens MDV-1 strain currently offer the best protection. Although attenuated CVI988/Rispens is non-oncogenic, it codes for at least two forms of the MDV oncoprotein Meq, and these proteins (CVI-Meq and CVI-LMeq) have not been fully characterized. Here, we report that both CVI-Meq proteins, like the Meq protein of Md5 (a very virulent oncogenic strain), were capable of transforming Rat-2 and NIH3T3 cells. Both CVI-Meq and CVI-LMeq proteins activated the meq promoter only in the presence of chicken c-Jun (CK-Jun) whereas Md5-Meq activated the same promoter irrespective of CK-Jun co-expression. However, Meq proteins of both Md5 and CVI988 bound the meq promoter in a ChIP assay regardless of whether CK-Jun was co-expressed. To understand the role of Meq DNA binding and transactivation/repression domains in transcription, we constructed three chimeric Meq proteins, namely, Md5-CVI-Meq, CVI-Md5-Meq, and Md5-CVI-L by exchanging domains between Md5 meq and CVI meq genes. Although these chimeric Meq proteins, unlike CVI-Meq proteins, transactivated the meq promoter, the activation was significantly less than Md5-Meq. To determine the role of individual amino acids, point mutations were introduced corresponding to the amino acid changes of CVI-Meq into Md5-Meq. Amino acid residues at positions 71 and 320 of the Md5-Meq protein were found to be important for transactivation of the meq promoter. All three Meq proteins activated the MDV gB, MMP-3 and Bcl-2 promoters and suppressed transcription from the MDV pp38/pp14 bidirectional promoter. Although no significant differences were observed, decreased transactivation activity was observed with CVI-Meq proteins when compared to Md5-Meq. Collectively, the data presented here indicate that CVI-Meq proteins are generally weak transactivators, which might contribute to the non-oncogenic phenotype of CVI988 virus in chickens.


Asunto(s)
Herpesvirus Gallináceo 2/metabolismo , Vacunas contra la Enfermedad de Marek/metabolismo , Enfermedad de Marek/virología , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Enfermedades de las Aves de Corral/virología , Animales , Línea Celular , Transformación Celular Viral , Embrión de Pollo , Pollos , Herpesvirus Gallináceo 2/genética , Enfermedad de Marek/genética , Enfermedad de Marek/metabolismo , Vacunas contra la Enfermedad de Marek/genética , Ratones , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas
18.
J Virol ; 83(2): 859-69, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18971275

RESUMEN

Marek's disease virus (MDV), the etiologic agent of Marek's disease, is a potent oncogenic herpesvirus. MDV is highly contagious and elicits a rapid onset of malignant T-cell lymphomas in chickens within several weeks after infection. MDV genome codes an oncoprotein, Meq, which shares resemblance with the Jun/Fos family of bZIP transcription factors. Similar to Jun, the leucine zipper region of Meq allows the formation of homo- and heterodimers. Meq homo- and heterodimers have different DNA binding affinities and transcriptional activity; therefore, they may differentially regulate transcription of viral and cellular genes. In this study we investigated the role of Meq homodimers in the pathogenicity of MDV by generating a chimeric meq gene, which contains the leucine zipper region of the yeast transcription factor GCN4 (meqGCN). A recombinant virus (rMd5-MeqGCN) containing the chimeric meqGCN gene in place of parental meq was generated with overlapping cosmid clones of Md5, a very virulent MDV strain. The rMd5-MeqGCN virus replicated in vitro and in vivo but was unable to transform T cells in infected chickens. These data provide the first in vivo evidence that Meq homodimers are not sufficient for MDV-induced transformation.


Asunto(s)
Transformación Celular Viral , Linfocitos/virología , Mardivirus/patogenicidad , Enfermedad de Marek/virología , Enfermedades de las Aves de Corral/virología , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Pollos , Patos , Fibroblastos/virología , Proteínas Oncogénicas/metabolismo , Multimerización de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...