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











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 13: 902840, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36311717

RESUMO

In the wake of the A/California/7/2009 H1N1 influenza pandemic vaccination campaigns in 2009-2010, an increased incidence of the chronic sleep-wake disorder narcolepsy was detected in children and adolescents in several European countries. Over the last decade, in-depth epidemiological and immunological studies have been conducted to investigate this association, which have advanced our understanding of the events underpinning the observed risk. Narcolepsy with cataplexy (defined as type-1 narcolepsy, NT1) is characterized by an irreversible and chronic deficiency of hypocretin peptides in the hypothalamus. The multifactorial etiology is thought to include genetic predisposition, head trauma, environmental triggers, and/or infections (including influenza virus infections), and an increased risk was observed following administration of the A/California/7/2009 H1N1 vaccine Pandemrix (GSK). An autoimmune origin of NT1 is broadly assumed. This is based on its strong association with a predisposing allele (the human leucocyte antigen DQB1*0602) carried by the large majority of NT1 patients, and on links with other immune-related genetic markers affecting the risk of NT1. Presently, hypotheses on the underlying potential immunological mechanisms center on molecular mimicry between hypocretin and peptides within the A/California/7/2009 H1N1 virus antigen. This molecular mimicry may instigate a cross-reactive autoimmune response targeting hypocretin-producing neurons. Local CD4+ T-cell responses recognizing peptides from hypocretin are thought to play a central role in the response. In this model, cross-reactive DQB1*0602-restricted T cells from the periphery would be activated to cross the blood-brain barrier by rare, and possibly pathogen-instigated, inflammatory processes in the brain. Current hypotheses suggest that activation and expansion of cross-reactive T-cells by H1N1/09 influenza infection could have been amplified following the administration of the adjuvanted vaccine, giving rise to a "two-hit" hypothesis. The collective in silico, in vitro, and preclinical in vivo data from recent and ongoing research have progressively refined the hypothetical model of sequential immunological events, and filled multiple knowledge gaps. Though no definitive conclusions can be drawn, the mechanistical model plausibly explains the increased risk of NT1 observed following the 2009-2010 H1N1 pandemic and subsequent vaccination campaign, as outlined in this review.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Influenza Humana , Narcolepsia , Criança , Adolescente , Humanos , Influenza Humana/epidemiologia , Influenza Humana/prevenção & controle , Influenza Humana/complicações , Orexinas , Narcolepsia/etiologia , Narcolepsia/genética , Peptídeos
2.
Cancer Immunol Immunother ; 62(3): 529-39, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23069871

RESUMO

There is a complex interplay between the immune system and a developing tumor that is manifest in the way that the balance of T cell subsets in the local tumor environment reflects clinical outcome. Tumor infiltration by CD8(+) T cells and regulatory T cells (Treg) is associated with improved and reduced survival, respectively, in many cancer types. However, little is known of the prognostic value of immunological parameters measured in peripheral blood. In this study, peripheral CD8(+) T cells and Treg from 43 patients with malignant mesothelioma or advanced non-small-cell lung cancer scheduled to commence palliative chemotherapy were assessed by flow cytometry and evaluated for association with patient survival. Patients had a higher proportion of peripheral Treg, proliferating CD8(+) T cells and CD8(+) T cells with an activated effector phenotype compared with age-matched healthy controls. Higher proportions of Treg and proliferating CD8(+) T cells were both associated with poor survival in univariate analyses (hazard ratio [HR] 3.81, 95 % CI 1.69-8.57; p < 0.01 and HR 2.86, 95 % CI 1.26-6.50; p < 0.05, respectively). CD8(+) T cell proliferation was independently predictive of reduced survival in multivariate analysis (HR 2.58, 95 % CI 1.01-6.61; p < 0.05). These findings suggest that peripheral CD8(+) T cell proliferation can be a useful prognostic marker in patients with thoracic malignancies planned for palliative chemotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Neoplasias Pulmonares/imunologia , Mesotelioma/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Proliferação de Células , Feminino , Humanos , Neoplasias Pulmonares/mortalidade , Ativação Linfocitária , Masculino , Mesotelioma/mortalidade , Pessoa de Meia-Idade , Prognóstico , Linfócitos T Reguladores/imunologia
3.
J Exp Med ; 207(6): 1333-43, 2010 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-20513749

RESUMO

Effective immunity requires the coordinated activation of innate and adaptive immune responses. Natural killer (NK) cells are central innate immune effectors, but can also affect the generation of acquired immune responses to viruses and malignancies. How NK cells influence the efficacy of adaptive immunity, however, is poorly understood. Here, we show that NK cells negatively regulate the duration and effectiveness of virus-specific CD4+ and CD8+ T cell responses by limiting exposure of T cells to infected antigen-presenting cells. This impacts the quality of T cell responses and the ability to limit viral persistence. Our studies provide unexpected insights into novel interplays between innate and adaptive immune effectors, and define the critical requirements for efficient control of viral persistence.


Assuntos
Antivirais/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Imunidade Inata/imunologia , Viroses/imunologia , Animais , Citotoxicidade Imunológica/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Epitopos , Antígenos de Histocompatibilidade/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Subfamília A de Receptores Semelhantes a Lectina de Células NK/deficiência , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Peptídeos/imunologia
4.
J Gen Virol ; 91(Pt 10): 2497-506, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20554798

RESUMO

The severity of respiratory syncytial virus (RSV) infections appears to differ with age in both humans and bovines. A primary RSV infection in naïve infants and in young calves runs a more severe course when they are 1-6 months old than in their first month of life. The relative lack of clinical signs in the first month of age may be due to high levels of maternally derived neutralizing antibodies or low exposure to infectious virus. This study examined whether age-dependent differences in the pathogenesis of bovine RSV (bRSV) between neonatal and young calves may be due to differences in age-dependent immunocompetence. To study the effect of age and immune parameters on bRSV disease in neonatal and young calves, neonatal (1-day-old) calves without maternally derived antibodies were infected experimentally with bRSV and the severity of disease and immune responses were evaluated in comparison with disease in similar 6-week-old infected calves. Neonatal calves had more extensive virus replication and lung consolidation, but lower pro-inflammatory [in particular tumour necrosis factor alpha (TNF-α)] responses, specific humoral immune responses, lung neutrophilic infiltration and clinical signs of disease than 6-week-old calves. The lack of correlation between virus replication and clinical signs suggests an important role of pro-inflammatory cytokines, in particular TNF-α, in the disease. The capacity to produce pro-inflammatory TNF-α appeared to increase with age, and may explain the age-dependent differences in RSV pathogenesis.


Assuntos
Doenças dos Bovinos/imunologia , Doenças dos Bovinos/patologia , Imunocompetência , Infecções por Vírus Respiratório Sincicial/veterinária , Vírus Sincicial Respiratório Bovino/imunologia , Vírus Sincicial Respiratório Bovino/patogenicidade , Fatores Etários , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/imunologia , Bovinos , Citocinas/imunologia , Citocinas/metabolismo , Pulmão/imunologia , Pulmão/patologia , Neutrófilos/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Índice de Gravidade de Doença
5.
J Immunol ; 183(12): 7898-908, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20007574

RESUMO

Tumors have evolved multiple mechanisms to evade immune destruction. One of these is expression of T cell inhibitory ligands such as programmed death-ligand 1 (PD-L1; B7-H1). In this study, we show that PD-L1 is highly expressed on mesothelioma tumor cells and within the tumor stroma. However, PD-L1 blockade only marginally affected tumor growth and was associated with the emergence of activated programmed death-1(+) ICOS(+) CD4 T cells in tumor-draining lymph nodes, whereas few activated CD8 T cells were present. Full activation of antitumor CD8 T cells, characterized as programmed death-1(+) ICOS(+) Ki-67(+) and displaying CTL activity, was only observed when CD4 T cells were depleted, suggesting that a population of suppressive CD4 T cells exists. ICOS(+) foxp3(+) regulatory T cells were found to be regulated through PD-L1, identifying one potentially suppressive CD4 T cell population. Thus, PD-L1 blockade activates antitumor CD8 T cell most potently in the absence of CD4 T cells. These findings have implications for the development of PD-L1-based therapies.


Assuntos
Antígenos de Superfície/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Antígeno B7-1/fisiologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Glicoproteínas de Membrana/fisiologia , Mesotelioma/imunologia , Peptídeos/fisiologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Animais , Antígeno B7-H1 , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Contraindicações , Feminino , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Ativação Linfocitária/imunologia , Depleção Linfocítica , Glicoproteínas de Membrana/antagonistas & inibidores , Mesotelioma/patologia , Mesotelioma/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Peptídeos/antagonistas & inibidores , Receptor de Morte Celular Programada 1 , Células Estromais/imunologia , Células Estromais/metabolismo , Células Estromais/patologia , Linfócitos T Reguladores/metabolismo
6.
PLoS One ; 4(9): e6982, 2009 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-19746156

RESUMO

BACKGROUND: Anti-cancer chemotherapy can be simultaneously lymphodepleting and immunostimulatory. Pre-clinical models clearly demonstrate that chemotherapy can synergize with immunotherapy, raising the question how the immune system can be mobilized to generate anti-tumor immune responses in the context of chemotherapy. METHODS AND FINDINGS: We used a mouse model of malignant mesothelioma, AB1-HA, to investigate T cell-dependent tumor resolution after chemotherapy. Established AB1-HA tumors were cured by a single dose of cyclophosphamide in a CD8 T cell- and NK cell-dependent manner. This treatment was associated with an IFN-alpha/beta response and a profound negative impact on the anti-tumor and total CD8 T cell responses. Despite this negative effect, CD8 T cells were essential for curative responses. The important effector molecules used by the anti-tumor immune response included IFN-gamma and TRAIL. The importance of TRAIL was supported by experiments in nude mice where the lack of functional T cells could be compensated by agonistic anti-TRAIL-receptor (DR5) antibodies. CONCLUSION: The data support a model in which chemotherapy sensitizes tumor cells for T cell-, and possibly NK cell-, mediated apoptosis. A key role of tumor cell sensitization to immune attack is supported by the role of TRAIL in tumor resolution and explains the paradox of successful CD8 T cell-dependent anti-tumor responses in the absence of CD8 T cell expansion.


Assuntos
Antineoplásicos/farmacologia , Linfócitos T CD8-Positivos/imunologia , Ciclofosfamida/farmacologia , Neoplasias/terapia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Apoptose , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Imunoterapia/métodos , Interferon gama/metabolismo , Células Matadoras Naturais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/imunologia
7.
J Immunol ; 182(9): 5217-24, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380767

RESUMO

Topical application of tumors with the TLR7 agonist imiquimod is an effective adjunct treatment for a range of primary dermatological cancers. However, for therapy to be effective against a broad range of solid tumor types, it must promote a strong systemic antitumor response that targets metastases in addition to primary tumor. We therefore investigated the potential of locally delivered imiquimod to stimulate an effective systemic antitumor response in a murine model of malignant mesothelioma (AB1-HA) with primary and distal tumors (dual tumor). Persistent delivery of imiquimod into primary tumor significantly retarded tumor growth in all treated mice compared with vehicle control. This local antitumor immune response required both CD8 T cells and NK cells, but not CD4 T cells, and was reliant on type I IFN induction. In vivo CTL studies and Ly6A/E staining of lymphocytes suggested that local imiquimod treatment had indeed induced a systemic, Ag-specific CD8 response. However, notably this response was not sufficient to retard the growth of an untreated distal tumor. Because local imiquimod treatment did not induce significant CD4 T cell responses, we investigated the efficacy of combining imiquimod with agonistic CD40 Ab (as a surrogate for CD4 T cell help). Combination of locally delivered imiquimod with systemic anti-CD40 immunotherapy not only significantly enhanced the local antitumor response, with 30% complete resolution, but it was also effective at significantly retarding growth of distal tumor. These results demonstrate that antitumor responses induced by locally delivered TLR7 agonists can be harnessed systemically for treating distal tumor.


Assuntos
Aminoquinolinas/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Antígenos CD40/imunologia , Glicoproteínas de Membrana/agonistas , Mesotelioma/imunologia , Mesotelioma/terapia , Receptor 7 Toll-Like/agonistas , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , Aminoquinolinas/uso terapêutico , Animais , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Quimioterapia Combinada , Feminino , Imiquimode , Interferon Tipo I/administração & dosagem , Interferon gama/administração & dosagem , Células Matadoras Naturais/imunologia , Ligantes , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/uso terapêutico , Mesotelioma/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Camundongos Transgênicos , Receptor 7 Toll-Like/metabolismo , Receptor 7 Toll-Like/uso terapêutico
8.
J Virol ; 83(9): 4386-94, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19211743

RESUMO

During some persistent viral infections, virus-specific T-cell responses wane due to the antigen-specific deletion or functional inactivation (i.e., exhaustion) of responding CD8 T cells. T-cell exhaustion often correlates with high viral load and is associated with the expression of the inhibitory receptor PD-1. In other infections, functional T cells are observed despite high levels of pathogen persistence. The reasons for these different T-cell fates during chronic viral infections are not clear. Here, we tracked the fate of virus-specific CD8 T cells in lymphocytic choriomeningitis virus (LCMV)-infected mice during viral clearance, the persistence of wild-type virus, or the selection and persistence of a viral variant that abrogates the presentation of a single epitope. Viral clearance results in PD-1(lo) functional virus-specific CD8 T cells, while the persistence of wild-type LCMV results in high PD-1 levels and T-cell exhaustion. However, following the emergence of a GP35V-->A variant virus that abrogates the presentation of the GP33 epitope, GP33-specific CD8 T cells remained functional, continued to show low levels of PD-1, and reexpressed CD127, a marker of memory T-cell differentiation. In the same animals and under identical environmental conditions, CD8 T cells recognizing nonmutated viral epitopes became physically deleted or were PD-1(hi) and nonfunctional. Thus, the upregulation of PD-1 and the functional inactivation of virus-specific T cells during chronic viral infection is dependent upon continued epitope recognition. These data suggest that optimal strategies for vaccination should induce high-magnitude broadly specific T-cell responses that prevent cytotoxic T-lymphocyte escape and highlight the need to evaluate the function of vaccine-induced T cells in the context of antigens presented during virus persistence.


Assuntos
Antígenos de Diferenciação/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Animais , Antígenos Virais/genética , Antígenos Virais/metabolismo , Sequência de Bases , Doença Crônica , Feminino , Glicoproteínas/genética , Glicoproteínas/metabolismo , Subunidade alfa de Receptor de Interleucina-7/imunologia , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/isolamento & purificação , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Receptor de Morte Celular Programada 1 , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Titulometria , Proteínas Virais/genética , Proteínas Virais/metabolismo
9.
Cancer Immunol Immunother ; 58(8): 1219-28, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19052741

RESUMO

Tumor cell death potentially engages with the immune system. However, the efficacy of anti-tumor chemotherapy may be limited by tumor-driven immunosuppression, e.g., through CD25+ regulatory T cells. We addressed this question in a mouse model of mesothelioma by depleting or reconstituting CD25+ regulatory T cells in combination with two different chemotherapeutic drugs. We found that the efficacy of cyclophosphamide to eradicate established tumors, which has been linked to regulatory T cell depletion, was negated by adoptive transfer of CD25+ regulatory T cells. Analysis of post-chemotherapy regulatory T cell populations revealed that cyclophosphamide depleted cycling (Ki-67(hi)) T cells, including foxp3+ regulatory CD4+ T cells. Ki-67(hi) CD4+ T cells expressed increased levels of two markers, TNFR2 and ICOS, that have been associated with a maximally suppressive phenotype according to recently published studies. This suggest that cyclophosphamide depletes a population of maximally suppressive regulatory T cells, which may explain its superior anti-tumor efficacy in our model. Our data suggest that regulatory T cell depletion could be used to improve the efficacy of anti-cancer chemotherapy regimens. Indeed, we observed that the drug gemcitabine, which does not deplete cycling regulatory T cells, eradicates established tumors in mice only when CD25+ CD4+ T cells are concurrently depleted. Cyclophosphamide could be used to achieve regulatory T cell depletion in combination with chemotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Ciclofosfamida/uso terapêutico , Imunossupressores/uso terapêutico , Depleção Linfocítica , Mesotelioma/tratamento farmacológico , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Transferência Adotiva , Animais , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Proteína Coestimuladora de Linfócitos T Induzíveis , Estimativa de Kaplan-Meier , Antígeno Ki-67/imunologia , Antígeno Ki-67/metabolismo , Selectina L/imunologia , Selectina L/metabolismo , Mesotelioma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Linfócitos T Reguladores/imunologia , Gencitabina
10.
Curr Opin Immunol ; 20(5): 545-57, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18573339

RESUMO

Tumors can acquire mutations or hijack regulatory pathways of the host immune system to render them resistant to immune attack. Standard first line therapies such as chemotherapy and radiation were not thought to provoke natural immunity to cancer, but recent findings demonstrating that dying tumor cells present and release key signals to stimulate or evade neighboring leukocytes are challenging that view. Killing tumor cells in a manner that provides danger signals and tumor antigens in the right context promotes the engagement of innate and adaptive immunity; however, this response alone will not be effective against established cancer. Coincidently driving the immune response with specific monoclonal antibodies and other immunomodulators that activate and mature dendritic cells and co-stimulate T cells and other lymphocytes is one approach. Additionally releasing immune checkpoints and inhibiting tumor-derived molecules that prevent effective tumor immunity is another. Combined these approaches have enormous potential to improve the current outcomes from conventional cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/imunologia , Células Dendríticas/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Terapia Combinada , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Humanos , Fatores Imunológicos/uso terapêutico , Imunoterapia , Receptores de Reconhecimento de Padrão/imunologia , Linfócitos T Citotóxicos/metabolismo
11.
J Immunol ; 180(3): 1535-44, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18209049

RESUMO

Effective antitumor CD8 T cell responses may be activated by directly targeting the innate immune system within tumors. We investigated this response by injecting a range of TLR agonists into established tumors using a mouse model of malignant mesothelioma stably transduced with the hemagglutinin (HA) gene as a marker Ag (AB1-HA). Persistent delivery of the dsRNA mimetic poly(I:C) into established AB1-HA tumors resulted in complete tumor resolution in 40% of mice, with the remaining mice also showing a significant delay in tumor progression. Experiments in athymic nude mice along with CD8 depletion and IFN-alphabeta blocking studies revealed that tumor resolution required both CD8 T cells and type I IFN induction, and was associated with local changes in MHC class I expression. Surprisingly, however, tumor resolution was not associated with systemic dissemination or tumor infiltration of effector CD8 T cells. Instead, the antitumor response was critically dependent on the reactivation of tumor-resident CD8 T cell responses. These studies suggest that, once reactivated, pre-existing local CD8 T cell responses are sufficient to resolve established tumors and that in situ type I IFN is a determining factor.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade Inata , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Mesotelioma/imunologia , Animais , Antivirais/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Ligantes , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Ácidos Nucleicos/imunologia , Poli I-C/farmacologia , RNA de Cadeia Dupla/farmacologia
12.
Virology ; 368(1): 17-25, 2007 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-17632195

RESUMO

Pneumonia virus of mice (PVM) causes bronchiolitis and pneumonia in mice. Infection is associated with high levels of viral replication in the lungs and results in the functional inactivation of pulmonary virus-specific CD8 T cells. Due to its similarity to severe human respiratory syncytial virus (RSV) infection, PVM infection in mice has been proposed as an alternative RSV model. Here, we have delineated the minimal requirements for protective T cell immunity in the PVM model. Immunization with a CD8 T cell epitope from the PVM phosphoprotein P, combined with the ovalbumin (OVA) CD4 T cell epitope, did not confer protective immunity against lethal PVM challenge, suggesting a possible role of cognate CD4 T cell immunity. To determine the role of PVM-specific CD4 T cell responses, we mapped a PVM CD4 T cell epitope in the glycoprotein G, using a panel of overlapping peptides. Although immunization with this epitope provided some protection, solid protective immunity was only observed after immunization with a combination of the PVM-specific CD4 and CD8 T cell epitopes. Analysis of post-challenge T cell responses in immunized mice indicated that G-specific pulmonary CD4 T cells displayed a mixed Th1/Th2 phenotype, which was characterized by the presence of both IL-5 and IFN-gamma secreting cells, in the absence of overt pathology.


Assuntos
Epitopos de Linfócito T/imunologia , Glicoproteínas/imunologia , Vírus da Pneumonia Murina/imunologia , Infecções por Pneumovirus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Mapeamento de Epitopos , Feminino , Interferon gama/biossíntese , Interleucina-5/biossíntese , Pulmão/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida
14.
Virology ; 352(1): 157-68, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16730775

RESUMO

The BALB/c mouse model for human respiratory syncytial virus infection has contributed significantly to our understanding of the relative role for CD4+ and CD8+ T cells to immune protection and pathogenic immune responses. To enable comparison of RSV-specific T cell responses in different mouse strains and allow dissection of immune mechanisms by using transgenic and knockout mice that are mostly available on a C57BL/6 background, we characterized the specificity, level and functional capabilities of CD8+ T cells during primary and secondary responses in lung parenchyma, airways and spleens of C57BL/6 mice. During the primary response, epitopes were recognized originating from the matrix, fusion, nucleo- and attachment proteins, whereas the secondary response focused predominantly on the matrix epitope. C57BL/6 mice are less permissive for hRSV infection than BALB/c mice, yet we found CD8+ T cell responses in the lungs and bronchoalveolar lavage, comparable to the responses described for BALB/c mice.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sincicial Respiratório Humano/patogenicidade , Sequência de Aminoácidos , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Organismos Livres de Patógenos Específicos , Proteínas Virais/química , Proteínas Virais/imunologia
15.
J Immunother ; 29(2): 134-42, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16531814

RESUMO

Coramsine is a novel chemotherapeutic agent isolated from Solanum linnaeanum (devil's apple). Topical treatment provides clinical benefit for skin tumors. To evaluate the potential broader applicability of the drug, its in vivo anticancer efficacy in a murine model of malignant mesothelioma and its mode of action were investigated. Systemic administration of coramsine slowed tumor growth and prolonged survival time. Importantly, the antitumor efficacy of coramsine was enhanced when treatment was combined with stimulation of innate immunity using unmethylated CpG-containing oligodeoxynucleotides (ODNs). Combination treatment further slowed tumor growth and provided a survival benefit. Coramsine seems to kill tumor cells by direct cell lysis. Using 2 different assays to detect apoptosis (caspase activation and DNA fragmentation), we found no evidence that coramsine induces any form of programmed cell death. The fact that the efficacy of coramsine is potentiated by CpG ODNs suggests that coramsine-induced cell death is an immunologic null event.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antineoplásicos/farmacologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Oligodesoxirribonucleotídeos/farmacologia , Preparações de Plantas/farmacologia , Alcaloides de Solanáceas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Sinergismo Farmacológico , Imunidade Inata/efeitos dos fármacos , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Solanum
16.
Cancer Res ; 66(2): 601-4, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16423984

RESUMO

This review shows how tumor antigen cross-presentation is affected by the major therapeutic modalities including chemotherapy, radiotherapy, and surgery. We argue that this process could affect the way that a tumor works as its own cellular vaccine, and that it is differentially modulated by the choice of treatment.


Assuntos
Apresentação de Antígeno , Vacinas Anticâncer/imunologia , Apresentação Cruzada/imunologia , Antígenos de Neoplasias/imunologia , Antineoplásicos/imunologia , Terapia Combinada , Humanos , Neoplasias/imunologia , Neoplasias/terapia
17.
J Immunol ; 175(10): 6597-604, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16272314

RESUMO

Pneumonia virus of mice (PVM) is a natural pathogen of mice and has been proposed as a tractable model for the replication of a pneumovirus in its natural host, which mimics human infection with human respiratory syncytial virus (RSV). PVM infection in mice is highly productive in terms of virus production compared with the situation seen with RSV in mice. Because RSV suppresses CD8 T cell effector function in the lungs of infected mice, we have investigated the nature of PVM-induced CD8 T cell responses to study pneumovirus-induced T cell responses in a natural virus-host setting. PVM infection was associated with a massive influx of activated CD8 T cells into the lungs. After identification of three PVM-specific CD8 T cell epitopes, pulmonary CD8 T cell responses were enumerated. The combined frequency of cytokine-secreting CD8 T cells specific for the three epitopes was much smaller than the total number of activated CD8 T cells. Furthermore, quantitation of the CD8 T cell response against one of these epitopes (residues 261-270 from the phosphoprotein) by MHC class I pentamer staining and by in vitro stimulation followed by intracellular IFN-gamma and TNF-alpha staining indicated that the majority of pulmonary CD8 specific for the P261 epitope were deficient in cytokine production. This deficient phenotype was retained up to 96 days postinfection, similar to the situation in the lungs of human RSV-infected mice. The data suggest that PVM suppresses T cell effector functions in the lungs.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Pneumovirus/imunologia , Pneumovirus/imunologia , Sequência de Aminoácidos , Animais , Antígenos Virais/genética , Linfócitos T CD8-Positivos/patologia , Modelos Animais de Doenças , Epitopos/genética , Feminino , Humanos , Memória Imunológica , Técnicas In Vitro , Interferon gama/biossíntese , Pulmão/imunologia , Pulmão/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Pneumovirus/genética , Infecções por Pneumovirus/patologia , Infecções por Pneumovirus/virologia , Baço/imunologia , Baço/patologia , Fator de Necrose Tumoral alfa/biossíntese
18.
Cancer Res ; 65(17): 7580-4, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16140921

RESUMO

Resection alone is rarely curative for advanced tumors, but the outcome generally improves with adjuvant therapy. We have previously shown that a combination of traditional chemotherapy (gemcitabine) and immunotherapy (anti-CD40/FGK-45) without surgery is synergistic and can lead to long-term cure when applied to small tumors. Such cured animals have immunologic memory and are protected from rechallenge. Here we investigate the effectiveness of combination chemotherapy and immunotherapy after partial or complete surgical debulking of large tumors. We found that complete resection followed by combination chemotherapy/immunotherapy led to a high rate of cure (>80%) but failed to induce a long-term, tumor-specific memory. Partial debulking followed by combination therapy elicited the same proportion of cured animals but in contrast to complete resection, a memory response was invoked. We postulate that chemotherapy induced apoptosis of the residual tumor cells following incomplete resection is absolutely required for the induction of long-term immunologic memory.


Assuntos
Anticorpos/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Antígenos CD40/imunologia , Desoxicitidina/análogos & derivados , Mesotelioma/imunologia , Mesotelioma/terapia , Animais , Linhagem Celular Tumoral , Quimioterapia Adjuvante , Desoxicitidina/farmacologia , Imunização Passiva/métodos , Memória Imunológica , Mesotelioma/tratamento farmacológico , Mesotelioma/cirurgia , Camundongos , Camundongos Endogâmicos BALB C , Gencitabina
19.
Vaccine ; 23(16): 1900-9, 2005 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-15734062

RESUMO

A most essential step in vaccine research and development, ie vaccine studies in animals, seriously suffer from long timespans needed to arrive at effective immunogens. In this report we show how almost immediately after vaccination the antibody inducing potential of low immunogenic 'self' antigens can be accurately assessed. (We expect that this timespan can be reduced even more when 'non self' antigens are used, since such responses should be stronger.) The method takes advantage of the immediate onset after vaccination of the immune response in the spleen. This novel method allows detection of antigen-specific B cells of the spleen as early as 7 days after immunization and at frequencies as low as 10 in 1,000,000 cells. The method depends on sequential staining with PE- and APC-conjugated tetramers, made with the same biotinylated peptide. The antigenic peptides are biotinylated and tetramerized with either PE neutravidin or APC streptavidin. We expect that this method can be generally applied to visualize B cell responses, irrespective of the way they are induced. In addition to the fast selection and development of novel immunogens, this procedure can be used to delineate the kinetics of the B cell response, to phenotypically characterize and to isolate antigen-specific B cells, and, perhaps most importantly, to count them at the clonal level before any circulating antibodies can be detected.


Assuntos
Linfócitos B/imunologia , Vacinas Sintéticas/imunologia , Vacinas/imunologia , Animais , Avaliação Pré-Clínica de Medicamentos , Citometria de Fluxo , Hormônio Liberador de Gonadotropina/imunologia , Camundongos , Ovalbumina/imunologia , Fenótipo , Radioimunoensaio , Baço/citologia , Baço/imunologia
20.
Discov Med ; 5(27): 265-70, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20704886

RESUMO

Extract: Cytotoxic chemotherapy remains one of the most widely available treatment options for cancer. Unfortunately, the efficacy of chemotherapy is limited and, for solid tumors in particular, cures are rarely achieved. Immunotherapy is a more experimental treatment method, aimed at mobilizing the body's immune cells to attack tumor cells, but it is also rarely curative. Few studies have investigated the options for combining chemotherapy and immunotherapy, largely because the two forms of treatment are considered to be antagonistic. Two assumptions have contributed to this view. First, most chemotherapies kill target cells by triggering a process of programmed cell death, or apoptosis, and this mode of cell death has been regarded as non-stimulatory or tolerogenic (induces tolerance). Thus, apoptosis-inducing chemotherapy would be expected to induce a state of non-responsiveness in the cytotoxic T lymphocytes that could otherwise potentially destroy tumor cells. Second, lymphocyte depletion (lymphopenia) is a common side effect of many anti-cancer drugs, and this too has been assumed to be detrimental to any potential immune response. However, recent advances in our thinking on immune regulation now challenge both of these assumptions. Here, we will review how recent data support the case for combining chemotherapy and immunotherapy in cancer treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA