Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Pathog ; 15(12): e1008192, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31809522

RESUMEN

The hypoxia-inducible factor 1 alpha (HIF1α) protein and the hypoxic microenvironment are critical for infection and pathogenesis by the oncogenic gammaherpesviruses (γHV), Kaposi sarcoma herpes virus (KSHV) and Epstein-Barr virus (EBV). However, understanding the role of HIF1α during the virus life cycle and its biological relevance in the context of host has been challenging due to the lack of animal models for human γHV. To study the role of HIF1α, we employed the murine gammaherpesvirus 68 (MHV68), a rodent pathogen that readily infects laboratory mice. We show that MHV68 infection induces HIF1α protein and HIF1α-responsive gene expression in permissive cells. siRNA silencing or drug-inhibition of HIF1α reduce virus production due to a global downregulation of viral gene expression. Most notable was the marked decrease in many viral genes bearing hypoxia-responsive elements (HREs) such as the viral G-Protein Coupled Receptor (vGPCR), which is known to activate HIF1α transcriptional activity during KSHV infection. We found that the promoter of MHV68 ORF74 is responsive to HIF1α and MHV-68 RTA. Moreover, Intranasal infection of HIF1αLoxP/LoxP mice with MHV68 expressing Cre- recombinase impaired virus expansion during early acute infection and affected lytic reactivation in the splenocytes explanted from mice. Low oxygen concentrations accelerated lytic reactivation and enhanced virus production in MHV68 infected splenocytes. Thus, we conclude that HIF1α plays a critical role in promoting virus replication and reactivation from latency by impacting viral gene expression. Our results highlight the importance of the mutual interactions of the oxygen-sensing machinery and gammaherpesviruses in viral replication and pathogenesis.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Infecciones por Herpesviridae/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Latencia del Virus/fisiología , Replicación Viral/fisiología , Animales , Ratones , Rhadinovirus/metabolismo
2.
J Neuroinflammation ; 13(1): 125, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27245318

RESUMEN

BACKGROUND: Individuals suffering from spinal cord injury (SCI) are at higher risk for respiratory-related viral infections such as influenza. In a previous study (Zha et al., J Neuroinflammation 11:65, 2014), we demonstrated that chronic spinal cord injury caused impairment in CD8(+)T cell function with increased expression of the immunosuppressive protein, programmed cell death 1 (PD-1). The present study was undertaken to establish whether chronic SCI-induced immune deficits would affect antiviral immunity directed against primary and secondary infections. METHODS: Six to seven weeks following a SCI contusion at thoracic level T9, mice were infected intranasally with influenza virus. Virus-specific immunity was analyzed at various time points post-infection and compared to uninjured controls. RESULTS: We report that chronic thoracic SCI impairs the ability of the animals to mount an adequate antiviral immune response. While all uninjured control mice cleared the virus from their lungs by day 10 post-infection, a significant number (approximately 70 %) of chronic SCI mice did not clear the virus and succumbed to infection-induced mortality. This was attributed to severe deficits in both virus-specific antibody production and CD8(+) T cell response in injured mice after primary infection. We also determined that previously acquired humoral immunity was maintained after spinal cord injury as vaccination against influenza A prior to injury-protected mice from a homologous viral challenge. In contrast, prior immunization did not protect mice from a heterotypic challenge with a different strain of influenza virus. CONCLUSIONS: Taken together, our data demonstrate that chronic SCI attenuates virus-specific humoral and cellular immunity during the establishment of primary response and impairs the development of memory CD8(+) T cells. In contrast, B cell memory acquired through vaccination prior to SCI is preserved after injury which demonstrates that antigen-specific memory cells are refractory following injury. Our study defines important parameters of the deficits of chronic SCI-induced immune depression during a viral respiratory infection. Our objective is to better understand the mechanisms of spinal cord injury-induced immune depression with the goal of developing more effective therapies and reduce mortality due to complications from influenza and other infections.


Asunto(s)
Anticuerpos Antivirales/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular/fisiología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Traumatismos de la Médula Espinal/inmunología , Animales , Anticuerpos Antivirales/metabolismo , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Enfermedad Crónica , Perros , Femenino , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , Orthomyxoviridae/inmunología , Orthomyxoviridae/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Vértebras Torácicas
3.
J Neuroinflammation ; 11: 65, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24690491

RESUMEN

BACKGROUND: Chronic spinal cord injury (SCI) induces immune depression in patients, which contributes to their higher risk of developing infections. While defects in humoral immunity have been reported, complications in T-cell immunity during the chronic phase of SCI have not yet been explored. METHODS: To assess the impact of chronic SCI on peripheral T-cell number and function we used a mouse model of severe spinal cord contusion at thoracic level T9 and performed flow cytometry analysis on the spleen for T-cell markers along with intracellular cytokine staining. Furthermore we identified alterations in sympathetic activity in the spleen of chronic SCI mice by measuring splenic levels of tyrosine hydroxylase (TH) and norepinephrine (NE). To gain insight into the neurogenic mechanism leading to T-cell dysfunction we performed in vitro NE stimulation of T-cells followed by flow cytometry analysis for T-cell exhaustion marker. RESULTS: Chronic SCI impaired both CD4+ and CD8+ T-cell cytokine production. The observed T-cell dysfunction correlated with increased expression of programmed cell death 1 (PD-1) exhaustion marker on these cells. Blocking PD-1 signaling in vitro restored the CD8+ T-cell functional defect. In addition, we showed that chronic SCI mice had higher levels of splenic NE, which contributed to the T-cell exhaustion phenotype, as PD-1 expression on both CD4+ and CD8+ T-cells was up-regulated following sustained exposure to NE in vitro. CONCLUSIONS: These studies indicate that alteration of sympathetic activity following chronic SCI induces CD8+ T-cell exhaustion, which in turn impairs T-cell function and contributes to immune depression. Inhibition of the exhaustion pathway should be considered as a new therapeutic strategy for chronic SCI-induced immune depression.


Asunto(s)
Apoptosis/fisiología , Linfocitos T CD8-positivos/fisiología , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Regulación hacia Arriba/fisiología , Animales , Apoptosis/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Ionóforos de Calcio/farmacología , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Ionomicina/farmacología , Ratones , Ratones Endogámicos C57BL , Norepinefrina/farmacología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Tirosina 3-Monooxigenasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
4.
PLoS One ; 18(7): e0289227, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37506075

RESUMEN

BACKGROUND: Infants with congenital Zika syndrome (CZS) are known to exhibit characteristic brain abnormalities. However, the brain anatomy of Zika virus (ZIKV)-exposed infants, born to ZIKV-positive pregnant mothers, who have normal-appearing head characteristics at birth, has not been evaluated in detail. The aim of this prospective study is, therefore, to compare the cortical and subcortical brain structural volume measures of ZIKV-exposed normocephalic infants to age-matched healthy controls. METHODS AND FINDINGS: We acquired T2-MRI of the whole brain of 18 ZIKV-exposed infants and 8 normal controls on a 3T MRI scanner. The MR images were auto-segmented into eight tissue types and anatomical regions including the white matter, cortical grey matter, deep nuclear grey matter, corticospinal fluid, amygdala, hippocampus, cerebellum, and brainstem. We determined the volumes of these regions and calculated the total intracranial volume (TICV) and head circumference (HC). We compared these measurements between the two groups, controlling for infant age at scan, by first comparing results for all subjects in each group and secondly performing a subgroup analysis for subjects below 8 weeks of postnatal age at scan. ZIKV-exposed infants demonstrated a significant decrease in amygdala volume compared to the control group in both the group and subgroup comparisons (p<0.05, corrected for multiple comparisons using FDR). No significant volume differences were observed in TICV, HC, or any specific brain tissue structures or regions. Study limitations include small sample size, which was due to abrupt cessation of extramural funding as the ZIKV epidemic waned. CONCLUSION: ZIKV-exposed infants exhibited smaller volumes in the amygdala, a brain region primarily involved in emotional and behavioral processing. This brain MRI finding may lead to poorer behavioral outcomes and warrants long-term monitoring of pediatric cases of infants with gestational exposure to Zika virus as well as other neurotropic viruses.


Asunto(s)
Craneosinostosis , Microcefalia , Complicaciones Infecciosas del Embarazo , Infección por el Virus Zika , Virus Zika , Recién Nacido , Embarazo , Femenino , Humanos , Lactante , Niño , Infección por el Virus Zika/epidemiología , Estudios Prospectivos , Complicaciones Infecciosas del Embarazo/epidemiología , Imagen por Resonancia Magnética , Encéfalo/anomalías , Microcefalia/epidemiología
5.
Antimicrob Agents Chemother ; 56(11): 5794-803, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22926574

RESUMEN

Lytic replication of the Kaposi's sarcoma-associated herpesvirus (KSHV) is essential for the maintenance of both the infected state and characteristic angiogenic phenotype of Kaposi's sarcoma and thus represents a desirable therapeutic target. During the peak of herpesvirus lytic replication, viral glycoproteins are mass produced in the endoplasmic reticulum (ER). Normally, this leads to ER stress which, through an unfolded protein response (UPR), triggers phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α), resulting in inhibition of protein synthesis to maintain ER and cellular homeostasis. However, in order to replicate, herpesviruses have acquired the ability to prevent eIF2α phosphorylation. Here we show that clinically achievable nontoxic doses of the glucose analog 2-deoxy-d-glucose (2-DG) stimulate ER stress, thereby shutting down eIF2α and inhibiting KSHV and murine herpesvirus 68 replication and KSHV reactivation from latency. Viral cascade genes that are involved in reactivation, including the master transactivator (RTA) gene, glycoprotein B, K8.1, and angiogenesis-regulating genes are markedly decreased with 2-DG treatment. Overall, our data suggest that activation of UPR by 2-DG elicits an early antiviral response via eIF2α inactivation, which impairs protein synthesis required to drive viral replication and oncogenesis. Thus, induction of ER stress by 2-DG provides a new antiherpesviral strategy that may be applicable to other viruses.


Asunto(s)
Antivirales/farmacología , Desoxiglucosa/farmacología , Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/antagonistas & inhibidores , Herpesvirus Humano 8/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Proteínas Virales/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Retículo Endoplásmico/genética , Retículo Endoplásmico/virología , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Expresión Génica/efectos de los fármacos , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Ratones , Fosforilación , Activación Transcripcional/efectos de los fármacos , Respuesta de Proteína Desplegada/genética , Ensayo de Placa Viral , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
6.
Pediatr Blood Cancer ; 59(7): 1280-3, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22628221

RESUMEN

BACKGROUND: Defects in the immune system may affect vaccine responsiveness. Because of the splenic hypofunction and abnormal opsonic activity, it was unknown whether patients with sickle cell disease (SCD) would respond appropriately to H1N1 vaccination. The objective of this study was to assess seroprotective post-vaccine H1N1 antibody response in children with SCD. PROCEDURE: Serum antibody titers were measured by hemagglutination inhibition and microneutralization (MN) assays. Correlations were established between clinical and treatment parameters and immune response. RESULTS: Twenty-nine of 38 (76.3%) subjects (mean age 11 ± 5.4 years) had durable protective antibody titers 8 ± 1.6 months (range 5-12 months) post-vaccination. Lessened immune response was not associated with time interval from vaccination, splenectomy, or hydroxyurea treatment. Lack of antibody response was associated with age less than 3 years and treatment with chronic transfusions. Of the nine non-responders, seven were on chronic transfusions (39% unresponsiveness rate in the transfused group). The difference in the number of patients with seropositivity between the non-transfused and the transfused groups was statistically significant (P = 0.039). CONCLUSIONS: Most subjects were able to mount an influenza-specific antibody response against the inactivated H1N1 vaccine. Similar to the general population, children less than 3 years were less likely to respond. In addition, patients on chronic transfusions were less likely to respond when compared to non-transfused children. Clinicians should be aware of the possibility of decreased vaccine response in patients with SCD on chronic transfusions. We postulate that transfusion-related immunomodulation (TRIM) may be related to decreased response.


Asunto(s)
Anemia de Células Falciformes/inmunología , Anticuerpos Antivirales/sangre , Transfusión Sanguínea , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Adolescente , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/terapia , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Esplenectomía , Vacunas de Productos Inactivados , Adulto Joven
7.
J Leukoc Biol ; 83(4): 1049-59, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18174364

RESUMEN

CD4(+)CD25(+) regulatory T lymphocytes (Tregs) critically contribute to the mechanisms of cancer-induced tolerance. These cells suppress anti-tumoral CD8(+) and CD4(+) T lymphocytes and can also restrain the function of APCs. We have previously documented the immunostimulatory effects of a chaperone-rich cell lysate (CRCL) anti-cancer vaccine. Tumor-derived CRCL induces tumor immunity in vivo, partly by promoting dendritic cell (DC) and macrophage activation. In the current study, we evaluated the effects of CD4(+)CD25(+)forkhead box P3(+) Tregs isolated from mice bearing 12B1 bcr-abl(+) leukemia on DC and macrophages that had been activated by 12B1-derived CRCL. CRCL-activated DC and macrophages resisted Treg suppression, as the production of proinflammatory cytokines, the activation of transcription factor NF-kappaB, and their immunostimulatory potential was unaffected by Tregs. Our results thus highlight CRCL as a powerful adjuvant endowed with the capacity to overcome tumor-induced Treg-inhibitory effects on APCs.


Asunto(s)
Células Dendríticas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células de la Médula Ósea/inmunología , Línea Celular Tumoral , Terapia de Inmunosupresión , Leucemia Experimental/patología , Activación de Linfocitos , Depleción Linfocítica , Macrófagos Peritoneales/inmunología , Ratones , FN-kappa B/metabolismo
8.
Mol Cancer Ther ; 7(3): 721-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18347157

RESUMEN

We have documented previously that a multiple chaperone protein vaccine termed chaperone-rich cell lysate (CRCL) promotes tumor-specific T-cell responses leading to cancer regression in several mouse tumor models. We report here that CRCL vaccine generated from a mouse breast cancer (TUBO, HER2/neu positive) is also capable of eliciting humoral immunity. Administration of TUBO CRCL triggered anti-HER2/neu antibody production and delayed the progression of established tumors. This antitumor activity can be transferred through the serum isolated from TUBO CRCL-immunized animals and involved both B cells and CD4(+) T lymphocytes. Further evaluation of the mechanisms underlying TUBO CRCL-mediated humoral immunity highlighted the role of antibody-dependent cell-mediated cytotoxicity. These results suggest that tumor-derived CRCL vaccine has a wider applicability as a cancer vaccine because it can target both T-cell- and B-cell-specific responses and may represent a promising approach for the immunotherapy of cancer.


Asunto(s)
Anticuerpos Antineoplásicos/biosíntesis , Vacunas contra el Cáncer/inmunología , Modelos Animales de Enfermedad , Neoplasias Mamarias Experimentales/inmunología , Chaperonas Moleculares/metabolismo , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Sueros Inmunes , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos
9.
Ophthalmic Surg Lasers Imaging Retina ; 49(9): e93-e98, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30222826

RESUMEN

In the spring of 2017, a full-term infant with microcephaly was delivered in South Florida. During first trimester, the mother presented with fever, nausea, and vomiting. She reported no foreign travel for herself or her partner. The infant's neurologic, ophthalmologic, neuroradiologic, and audiologic findings were highly suggestive of congenital Zika syndrome (CZS), confirmed by IgM antibodies and plaque reduction neutralization test. New observations, including peripheral temporal retinal avascularity and peripapillary retinal nerve fiber layer thinning, are presented from this first known case of non-travel-associated CZS in the United States. [Ophthalmic Surg Lasers Imaging Retina. 2018;49:e93-e98.].


Asunto(s)
Distrofias Hereditarias de la Córnea/diagnóstico , Infecciones Virales del Ojo/diagnóstico , Microcefalia/diagnóstico , Enfermedades del Nervio Óptico/diagnóstico , Complicaciones Infecciosas del Embarazo , Infección por el Virus Zika/diagnóstico , Femenino , Edad Gestacional , Humanos , Recién Nacido , Embarazo , ARN Viral/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Ultrasonografía Prenatal , Estados Unidos , Adulto Joven , Virus Zika/genética , Infección por el Virus Zika/congénito
10.
Exp Hematol ; 33(4): 443-51, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15781335

RESUMEN

OBJECTIVE: To quantify the immune response of WASP- mice to three different pathogens: influenza A virus, Streptococcus pneumoniae, and Mycobacterium bovis. METHODS: Primary and secondary T-cell responses to influenza A virus were quantified via tetramer assays. Viral clearance from lung was also measured. Lethality of intranasal inoculation with luminescent S. pneumoniae was assessed by dose escalation and direct luminescence imaging. After intravenous inoculation with M. bovis, residual mycobacteria in lung, liver, and spleen were measured by standard culture methods. RESULTS: The reduced secondary T-cell response to influenza A virus correlates with a relative but not absolute loss of splenic T and B cells similar to that seen in clinical Wiskott-Aldrich Syndrome (WAS), and slower clearance of virus from lung. The reduced magnitude of the secondary T-cell response correlates with a progressive loss of influenza-specific T cells after primary inoculation. WASP- mice show an increased susceptibility to lethal pneumonia after intranasal inoculation with S. pneumoniae, which is among the most frequent causes of clinical complications in WAS patients. WASP- mice clear M. bovis bacille Calmette-Guerin (BCG) more slowly from lung, liver, and spleen. Bone marrow-derived macrophages, however, show normal ex vivo cytokine secretion in response to M. bovis. CONCLUSIONS: These results demonstrate that WASP- mice are functionally immunodeficient in regard to three different pathogens, and provide relevant end points for the study of treatment modalities in this model. They also suggest a specific physiologic mechanism, failure to accumulate memory T cells, for at least one of the defective immune responses.


Asunto(s)
Inmunidad , Proteínas/inmunología , Linfocitos T/inmunología , Animales , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Virus de la Influenza A/inmunología , Hígado/microbiología , Pulmón/microbiología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Mycobacterium bovis/inmunología , Proteínas/genética , Bazo/microbiología , Streptococcus pneumoniae/inmunología , Proteína del Síndrome de Wiskott-Aldrich
11.
Mol Cancer Ther ; 15(11): 2750-2757, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27474152

RESUMEN

The catalytically deficient ERBB3 strongly synergizes with the receptor tyrosine kinase ERBB2, and elevated levels represent an overall risk factor for unfavorable disease outcomes in breast cancer. Although itself not a target of pan-ERBB kinase inhibitors, it contributes to resistance in ERBB2-targeted treatment regiments. The steroidal lactone Withaferin A (WA) has established broad anticancer properties through several modes of action and was shown to be effective against triple-negative breast cancers at elevated concentrations. We found that ERBB2 overexpression does render cells hypersensitive to WA. Although ERBB2 downregulation is one aspect of WA treatment at high concentrations, it is not causal for the elevated sensitivity at lower dosages. Instead, WA targets the ability of ERBB3 to amplify ERBB2 signaling. ERBB3 receptor levels, constitutive phosphorylation of both ERBB3 and ERBB2, as well as signaling through AKT are eliminated by WA treatment. By targeting ERBB2/ERBB3 as a functional unit, it is also effective in cases in which ERBB2-directed inhibitors, such as lapatinib, alone show reduced potency. Hence, WA or derivatives thereof may present a low toxicity addition to ERBB2-targeting therapeutics, especially in cases in which ERBB3 involvement is driving resistance or reduced overall sensitivity. Mol Cancer Ther; 15(11); 2750-7. ©2016 AACR.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/genética , Resistencia a Antineoplásicos/genética , Expresión Génica , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Witanólidos/farmacología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo
12.
Cancer Immunol Immunother ; 56(1): 48-59, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16612596

RESUMEN

CD4(+)CD25(+) regulatory T cells have been characterized as a critical population of immunosuppressive cells. They play a crucial role in cancer progression by inhibiting the effector function of CD4(+) or CD8(+) T lymphocytes. However, whether regulatory T lymphocytes that expand during tumor progression can modulate dendritic cell function is unclear. To address this issue, we have evaluated the inhibitory potential of CD4(+)CD25(+) regulatory T cells from mice bearing a BCR-ABL(+) leukemia on bone marrow-derived dendritic cells. We present data demonstrating that CD4(+)CD25(+)FoxP3(+) regulatory T cells from tumor-bearing animals impede dendritic cell function by down-regulating the activation of the transcription factor NF-kappaB. The expression of the co-stimulatory molecules CD80, CD86 and CD40, the production of TNF-alpha, IL-12, and CCL5/RANTES by the suppressed DC is strongly down-regulated. The suppression mechanism requires TGF-beta and IL-10 and is associated with induction of the Smad signaling pathway and activation of the STAT3 transcription factor.


Asunto(s)
Antígenos CD4/inmunología , Células Dendríticas/fisiología , Terapia de Inmunosupresión , Interleucina-10/metabolismo , Subunidad alfa del Receptor de Interleucina-2/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Células de la Médula Ósea , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/metabolismo , Proteínas de Fusión bcr-abl/metabolismo , Leucemia/inmunología , Leucemia/patología , Leucemia/prevención & control , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Fosforilación , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
13.
Int J Cancer ; 119(11): 2624-31, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16989012

RESUMEN

Tumor derived chaperone-rich cell lysate (CRCL) when isolated from tumor tissues is a potent vaccine that contains at least 4 of the highly immunogenic heat shock proteins (HSP) such as HSP70, HSP90, glucose related protein 94 and calreticulin. We have previously documented that CRCL provides both a source of tumor antigens and danger signals triggering dendritic cell (DC) activation. Immunization with tumor derived CRCL elicits tumor-specific T cell responses leading to tumor regression. In the current study, we further dissect the mechanisms by which CRCL simulates the immune system, and demonstrate that natural killer (NK) cells are required for effective antitumor effects to take place. Our results illustrate that CRCL directly stimulates proinflammatory cytokine and chemokine production by NK cells, which may lead to activation and recruitment of macrophages at the tumor site. Thus, this report provides further insight into the function of CRCL as an immunostimulant against cancer.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Células Asesinas Naturales/inmunología , Neoplasias Experimentales/inmunología , Animales , Vacunas contra el Cáncer/inmunología , Citocinas/biosíntesis , Citocinas/fisiología , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Activación de Macrófagos , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos BALB C
14.
J Virol ; 79(7): 4329-39, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15767433

RESUMEN

The extent to which CD8+ T cells specific for other antigens expand to compensate for the mutational loss of the prominent DbNP366 and DbPA224 epitopes has been investigated using H1N1 and H3N2 influenza A viruses modified by reverse genetics. Significantly increased numbers of CD8+ KbPB1(703)+, CD8+ KbNS2(114)+, and CD8+ DbPB1-F2(62)+ T cells were found in the spleen and in the inflammatory population recovered by bronchoalveolar lavage from mice that were first given the -NP-PA H1N1 virus intraperitoneally and then challenged intranasally with the homologous H3N2 virus. The effect was less consistent when this prime-boost protocol was reversed. Also, though the quality of the response measured by cytokine staining showed some evidence of modification when these minor CD8+-T-cell populations were forced to play a more prominent part, the effects were relatively small and no consistent pattern emerged. The magnitude of the enhanced clonal expansion following secondary challenge suggested that the prime-boost with the -NP-PA viruses gave a response overall that was little different in magnitude from that following comparable exposure to the unmanipulated viruses. This was indeed shown to be the case when the total response was measured by ELISPOT analysis with virus-infected cells as stimulators. More surprisingly, the same effect was seen following primary challenge, though individual analysis of the CD8+ KbPB1(703)+, CD8+ KbNS2(114)+, and CD8+ DbPB1-F2(62)+ sets gave no indication of compensatory expansion. A possible explanation is that novel, as yet undetected epitopes emerge following primary exposure to the -NP-PA deletion viruses. These findings have implications for both natural infections and vaccines.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Proliferación Celular , Epítopos de Linfocito T/genética , Femenino , Memoria Inmunológica , Virus de la Influenza A/genética , Activación de Linfocitos , Ratones , Eliminación de Secuencia , Bazo/inmunología , Subgrupos de Linfocitos T/inmunología
15.
J Gen Virol ; 86(Pt 4): 1121-1130, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15784906

RESUMEN

The severity of disease caused in humans by H5N1 influenza viruses remains unexplained. The NS gene of Hong Kong H5N1/97 viruses was shown to contribute to high pathogenicity of reassortants in a pig model. However, the molecular pathogenesis and host immune response underlying this phenomenon remain unclear. Here, in a mouse model, H1N1 A/Puerto Rico/8/34 (PR/8) reassortants that contained the H5N1/97 NS gene, the H5N1/01 NS gene, or an altered H5N1/97 NS gene encoding a Glu92-->Asp substitution in NS1 was studied. The pathogenicity of reassortant viruses, the induction of cytokines and chemokine CXCL1 (KC) in the lungs and specific B- and T-cell responses was characterized. In mice infected with reassortant virus containing the H5N1/97 NS gene, the mouse lethal dose (50%) and lung virus titres were similar to those of PR/8, which is highly pathogenic to mice. This reassortant virus required two more days than PR/8 to be cleared from the lungs of infected mice. Reassortants containing the altered H5N1/97 NS gene or the H5N1/01 NS gene demonstrated attenuated pathogenicity and lower lung titres in mice. Specific B- and T-cell responses were consistent with viral pathogenicity and did not explain the delayed clearance of the H5N1/97 NS reassortant. The reassortant induced elevated pulmonary concentrations of the inflammatory cytokines IL1alpha, IL1beta, IL6, IFN-gamma and chemokine KC, and decreased concentrations of the anti-inflammatory cytokine IL10. This cytokine imbalance is reminiscent of the clinical findings in two humans who died of H5N1/97 infection and may explain the unusual severity of the disease.


Asunto(s)
Linfocitos B/inmunología , Citocinas/biosíntesis , Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A/patogenicidad , Virus Reordenados/patogenicidad , Linfocitos T/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Virus de la Influenza A/genética , Gripe Humana/inmunología , Gripe Humana/fisiopatología , Gripe Humana/virología , Pulmón/inmunología , Pulmón/fisiopatología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Virus Reordenados/genética , Proteínas no Estructurales Virales/genética , Virulencia
16.
Proc Natl Acad Sci U S A ; 102(17): 6074-9, 2005 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-15831586

RESUMEN

The restriction of influenza A virus replication to mouse respiratory epithelium means that this host response is anatomically compartmentalized, on the one hand, to sites of T cell stimulation and proliferation in the secondary lymphoid tissue and, on the other hand, to the site of effector T cell function and pathology in the pneumonic lung. Thus, it is hardly surprising that virus-specific CD8(+) T cells recovered by bronchoalveolar lavage (BAL) from the infected respiratory tract seem more "activated" in terms of both cytolytic activity and cytokine production than those cells isolated from the spleen. The present analysis uses Affymetrix microarray technology to compare profiles of gene expression in these two lineage-related, yet anatomically separate, lymphocyte populations. Ninety differentially expressed genes were identified for influenza-specific CD8(+)D(b)NP(366)(+) T cells obtained directly ex vivo by BAL or spleen disruption, with nine genes being further analyzed by quantitative, real-time PCR at the population level. Integrin alphaE, for example, was shown by Affymetrix and real-time mRNA analyses and then by single-cell PCR and protein staining to be present at a much higher prevalence on the BAL CD8(+)D(b)NP(366)(+) set. The unpredicted finding, however, was that mRNA expression for 75% of the 90 genes was lower in T cells from the BAL than from the spleen. Apparently, the localization of virus-specific CD8(+) T cells to the site of virus-induced pathology is associated with a narrowing, or "focusing," of gene expression that favors enhanced effector function in the damaged, "high-antigen load" environment of the pneumonic lung.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Regulación de la Expresión Génica/inmunología , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Líquido del Lavado Bronquioalveolar/citología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/fisiología , Diferenciación Celular , Femenino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Orthomyxoviridae/genética , Neumonía Viral/genética , Neumonía Viral/inmunología , Linfocitos T Citotóxicos/inmunología
17.
Virology ; 315(1): 43-55, 2003 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-14592758

RESUMEN

We have investigated the interactions and consequences of superinfecting and coreplication of human papillomavirus (HPV) and herpes simplex virus (HSV) in human epithelial organotypic (raft) culture tissues. In HPV-positive tissues, HSV infection and replication induced significant cytopathic effects (CPE), but the tissues were able to recover and maintain a certain degree of tissue integrity and architecture. HPV31b not only maintained the episomal state of its genomic DNA but also maintained its genomic copy number even during times of extensive HSV-induced CPE. E2 transcripts encoded by HPV31b were undetectable even though HPV31b replication was maintained in HSV- infected raft tissues. Expression of HPV31b oncogenes (E6 and E7) was also repressed but to a lesser degree than was E2 expression. The extent of CPE induced by HSV is dependent on the magnitude of HPV replication and gene expression at the time of HSV infection. During active HSV infection, HPV maintains its genomic copy number even though genes required for its replication were repressed. These studies provide new insight into the complex interaction between two common human sexually transmitted viruses in an in vitro system, modeling their natural host tissue in vivo.


Asunto(s)
Herpesvirus Humano 1/patogenicidad , Herpesvirus Humano 2/patogenicidad , Papillomaviridae/patogenicidad , Replicación Viral , Diferenciación Celular , Línea Celular Transformada , Células Cultivadas , Técnicas de Cultivo , Efecto Citopatogénico Viral , ADN Viral/análisis , Células Epiteliales , Femenino , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/fisiología , Humanos , Inmunohistoquímica , Papillomaviridae/genética , Papillomaviridae/fisiología , Células Tumorales Cultivadas , Proteínas Virales/metabolismo
18.
Proc Natl Acad Sci U S A ; 101(7): 2017-22, 2004 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-14764895

RESUMEN

Can CD4(+) and CD8(+) "memory" T cells that are generated and maintained in the context of low-level virus persistence protect, in the absence of antibody, against a repeat challenge with the same pathogen? Although immune T cells exert effective, long-term control of a persistent gamma-herpesvirus (gammaHV68) in Ig(-/-) microMT mice, subsequent exposure to a high dose of the same virus leads to further low-level replication in the lung. This lytic phase in the respiratory tract is dealt with effectively by the recall of memory T cells induced by a gammaHV68 recombinant (M3LacZ) that does not express the viral M3 chemokine binding protein. At least for the CD8(+) response, greater numbers of memory T cells confer enhanced protection in the M3LacZ-immune mice. However, neither WT gammaHV68 nor the minimally persistent M3LacZ primes the T cell response to the extent that a WT gammaHV68 challenge fails to establish latency in the microMT mice. Memory CD4(+) and CD8(+) T cells thus act together to limit gammaHV68 infection but are unable to provide absolute protection against a high-dose, homologous challenge.


Asunto(s)
Gammaherpesvirinae/inmunología , Inmunoglobulinas/deficiencia , Memoria Inmunológica/inmunología , Sobreinfección/inmunología , Sobreinfección/virología , Linfocitos T/inmunología , Linfocitos T/virología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Inmunización Secundaria , Inmunoglobulinas/genética , Pulmón/virología , Ratones , Bazo/inmunología , Bazo/virología , Latencia del Virus/inmunología
19.
J Gen Virol ; 84(Pt 2): 337-341, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12560565

RESUMEN

The murine gammaherpesvirus-68 kills I-A(b-/-) mice despite the presence of virus-specific CD8+ cytotoxic T lymphocytes (CTL). This has raised the possibility that these CTL are functionally abnormal. Here, no difference was observed between I-A(b-/-) mice and I-A(b+/+) controls in virus-specific CTL function, T cell receptor usage, or surface phenotype. Thus CTL immunity was independent of CD4+ T cells in a chronic herpesvirus infection, but was still inadequate to control virus replication.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Gammaherpesvirinae/patogenicidad , Infecciones por Herpesviridae/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos H-2/metabolismo , Infecciones por Herpesviridae/virología , Antígeno de Histocompatibilidad H-2D , Ratones , Fragmentos de Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo
20.
J Virol ; 77(14): 7756-63, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12829815

RESUMEN

The consequences for the long-term maintenance of virus-specific CD8+-T-cell memory have been analyzed experimentally for sequential respiratory infections with readily eliminated (influenza virus) and persistent (gammaherpesvirus 68 [gammaHV68]) pathogens. Sampling a broad range of tissue sites established that the numbers of CD8+ T cells specific for the prominent influenza virus D(b)NP(366) epitope were reduced by about half in mice that had been challenged 100 days previously with gammaHV68, though the prior presence of a large CD8+ D(b)NP366+ population caused no selective defect in the gammaHV68-specific CD8+ K(b)p79+ response. Conversely, mice that had been primed and boosted to generate substantial gammaHV68-specific CD8+ D(b)p56+ populations did not show any decrease in prevalence for this set of CD8+ memory cytotoxic T lymphocytes (CTL) at 200 days after respiratory exposure to an influenza A virus. However, in both experiments, the total magnitude of the CD8+-T-cell pool was significantly diminished in those that had been infected with gammaHV68 and the influenza A virus. The broader implications of these findings, especially under conditions of repeated exposure to unrelated pathogens, are explored with a mathematical model which emphasizes that the immune effector and memory "phenome" is a function of the overall infection experience of the individual.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Gammaherpesvirinae/patogenicidad , Memoria Inmunológica , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Femenino , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Modelos Inmunológicos , Infecciones por Orthomyxoviridae/virología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA