Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Immunol ; 206(7): 1483-1492, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33685995

RESUMO

Patients with grade III anaplastic astrocytomas (AA) separate into survival cohorts based on the presence or absence of mutations in isocitrate dehydrogenase (IDH). Progression to glioblastoma (GBM), morphologically distinguishable by elevated microvascular proliferation, necrosis, and cell division in tumor tissues, is considerably more rapid in IDH wild-type tumors such that their diagnosis as AA is relatively rare. More often initially presenting as GBM, these contain higher numbers of tumor-associated macrophages (TAMs) than most AA, and GBM patients also have higher levels of circulating M2 monocytes. TAM and M2 monocytes share functional properties inhibitory for antitumor immunity. Yet, although there is a wealth of data implicating TAM in tumor-immune evasion, there has been limited analysis of the impact of the circulating M2 monocytes. In the current study, immune parameters in sera, circulating cells, and tumor tissues from patients with primary gliomas morphologically diagnosed as AA were assessed. Profound differences in serum cytokines, glioma extracellular vesicle cross-reactive Abs, and gene expression by circulating cells identified two distinct patient cohorts. Evidence of type 2-immune bias was most often seen in patients with IDH wild-type AA, whereas a type 1 bias was common in patients with tumors expressing the IDH1R132H mutation. Nevertheless, a patient's immune profile was better correlated with the extent of tumor vascular enhancement on magnetic resonance imaging than IDH mutational status. Regardless of IDH genotype, AA progression appears to be associated with a switch in systemic immune bias from type 1 to type 2 and the loss of tumor vasculature integrity.


Assuntos
Astrocitoma/imunologia , Glioblastoma/imunologia , Macrófagos Associados a Tumor/imunologia , Adulto , Sobreviventes de Câncer , Carcinogênese , Estudos de Coortes , Citocinas/metabolismo , Feminino , Humanos , Isocitrato Desidrogenase/genética , Masculino , Pessoa de Meia-Idade , Mutação/genética , Células Th1/imunologia , Equilíbrio Th1-Th2 , Células Th2/imunologia
2.
Future Virol ; 15(11): 755-761, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33343683

RESUMO

Immune memory cells residing in previously infected, nonlymphoid tissues play a role in immune surveillance. In the event that circulating antibodies fail to prevent virus spread to the tissues in a secondary infection, these memory cells provide an essential defense against tissue reinfection. CNS tissues are isolated from circulating immune cells and antibodies by the blood-brain barrier, making the presence of tissue-resident immune memory cells particularly needed to combat recurrent infection by neurotropic viruses. Wild-type and laboratory-engineered rabies viruses are neurotropic, differ in pathogenicity, and have varying effects on BBB functions. These viruses have proven invaluable tools in demonstrating the importance of tissue-resident immune memory cells in the reinfection of CNS tissues. Only Type 1 immune memory is effective at therapeutically clearing a secondary infection with wild-type rabies viruses from the CNS and does so despite the maintenance of blood-brain barrier integrity.

3.
J Neurovirol ; 24(5): 606-615, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29987584

RESUMO

Rabies virus is a neurotropic lyssavirus which is 100% fatal in its pathogenic form when reaching unprotected CNS tissues. Death can be prevented by mechanisms delivering appropriate immune effectors across the blood-brain barrier which normally remains intact during pathogenic rabies virus infection. One therapeutic approach is to superinfect CNS tissues with attenuated rabies virus which induces blood-brain barrier permeability and immune cell entry. Current thinking is that peripheral rabies immunization is sufficient to protect against a challenge with pathogenic rabies virus. While this is undoubtedly the case if the virus is confined to the periphery, what happens if the virus reaches the CNS is less well-understood. In the current study, we find that peripheral immunization does not fully protect mice long-term against an intranasal challenge with pathogenic rabies virus. Protection is significantly better in mice that have cleared attenuated virus from the CNS and is associated with a more robust CNS recall response evidently due to the presence in CNS tissues of elevated numbers of lymphocytes phenotypically resembling long-term resident immune cells. Adoptive transfer of cells from rabies-immune mice fails to protect against CNS challenge with pathogenic rabies virus further supporting the concept that long-term resident immune cell populations must be established in brain tissues to protect against a subsequent CNS challenge with pathogenic rabies virus.


Assuntos
Encéfalo/virologia , Vacina Antirrábica/imunologia , Raiva/imunologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Raiva , Vacinas Atenuadas/imunologia
4.
Matrix Biol ; 70: 20-35, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29530483

RESUMO

Therapeutic approaches aimed at curing prostate cancer are only partially successful given the occurrence of highly metastatic resistant phenotypes that frequently develop in response to therapies. Recently, we have described αvß6, a surface receptor of the integrin family as a novel therapeutic target for prostate cancer; this epithelial-specific molecule is an ideal target since, unlike other integrins, it is found in different types of cancer but not in normal tissues. We describe a novel αvß6-mediated signaling pathway that has profound effects on the microenvironment. We show that αvß6 is transferred from cancer cells to monocytes, including ß6-null monocytes, by exosomes and that monocytes from prostate cancer patients, but not from healthy volunteers, express αvß6. Cancer cell exosomes, purified via density gradients, promote M2 polarization, whereas αvß6 down-regulation in exosomes inhibits M2 polarization in recipient monocytes. Also, as evaluated by our proteomic analysis, αvß6 down-regulation causes a significant increase in donor cancer cells, and their exosomes, of two molecules that have a tumor suppressive role, STAT1 and MX1/2. Finally, using the Ptenpc-/- prostate cancer mouse model, which carries a prostate epithelial-specific Pten deletion, we demonstrate that αvß6 inhibition in vivo causes up-regulation of STAT1 in cancer cells. Our results provide evidence of a novel mechanism that regulates M2 polarization and prostate cancer progression through transfer of αvß6 from cancer cells to monocytes through exosomes.


Assuntos
Adenocarcinoma/genética , Antígenos de Neoplasias/genética , Exossomos/metabolismo , Regulação Neoplásica da Expressão Gênica , Integrinas/genética , Neoplasias da Próstata/genética , Fator de Transcrição STAT1/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/imunologia , Antineoplásicos Imunológicos/farmacologia , Comunicação Celular , Diferenciação Celular , Técnicas de Cocultura , Exossomos/patologia , Humanos , Integrinas/antagonistas & inibidores , Integrinas/imunologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Masculino , Camundongos , Camundongos Knockout , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/imunologia , Células PC-3 , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/imunologia , Cultura Primária de Células , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Fator de Transcrição STAT1/imunologia , Transdução de Sinais , Células THP-1 , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Trop Med Infect Dis ; 2(3)2017 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-30270881

RESUMO

Rabies remains a major public health issue worldwide, especially in developing countries where access to medical care can represent a real challenge. While there is still no cure for rabies, it is a vaccine-preventable disease with pre- and post-exposure prophylaxis regimens approved by the World Health Organization (WHO). However, many rabies-exposed individuals have limited access to vaccines and virus-neutralizing antibodies approved for post-exposure prophylaxis. Unfortunately, any delay in the administration of these reagents can have lethal consequences. This highlights the need to develop cost-effective immunological reagents with a greater window of efficacy. Live-attenuated vaccine strains of rabies virus presents a potential treatment in filling this gap. We show here that immunization with live-attenuated vaccines provide long-lasting rabies immunity, superior to the protection induced by inactivated vaccines. In the absence of an immunostimulatory adjuvant, vaccination with multiple doses of inactivated rabies virus induces a type-2 immune response. This type of immunity is highly effective at inducing neutralizing antibody but has limited efficacy in clearing the virus from central nervous system (CNS) tissues. In contrast, a single infection with live-attenuated rabies vaccine safely drives a type-1 immune response, associated with both the production of a neutralizing antibody and the clearance of wild-type rabies virus from CNS tissues. These results indicate that live-attenuated rabies strains have the potential to be more effective in post-exposure prophylaxis than conventional inactivated vaccines.

6.
J Immunol ; 195(9): 4358-68, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26408670

RESUMO

Much of our understanding of CNS immunity has been gained from models involving pathological inflammation. Attenuated rabies viruses (RABV) are unique tools to study CNS immunity in the absence of conventional inflammatory mechanisms, as they spread from the site of inoculation to the CNS transaxonally, thereby bypassing the blood-brain barrier (BBB), and are cleared without neutrophil or monocyte infiltration. To better understand the role of CD4 T cell subsets in the clearance of the virus from CNS tissues, we examined the development of antiviral immunity in wild-type (WT) and T-bet knockout mice (T-bet(-/-)), which lack Th1 cells. Early control of RABV replication in the CNS tissues of WT mice is associated with the production of IFN-γ, with antiviral effects likely mediated through the enhanced expression of type I IFNs. Of interest, IFN-α and -γ are overexpressed in the infected T-bet(-/-) by comparison with WT CNS tissues, and the initial control of RABV infection is similar. Ultimately, attenuated RABV are cleared from the CNS tissues of WT mice by Ab locally produced by the activities of infiltrating T and B cells. Although T and B cell infiltration into the CNS of infected T-bet(-/-) mice is comparable, their activities are not, the consequence being delayed, low-level Ab production and prolonged RABV replication. More importantly, neither T-bet(-/-) mice immunized with an attenuated virus, nor WT mice with Th2 RABV-specific immunity induced by immunization with inactivated virus, are protected in the long term against challenge with a pathogenic RABV.


Assuntos
Sistema Nervoso Central/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Proteínas com Domínio T/imunologia , Animais , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/virologia , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/virologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Citometria de Fluxo , Expressão Gênica/imunologia , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/metabolismo , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Raiva/metabolismo , Raiva/virologia , Vacina Antirrábica/imunologia , Vacina Antirrábica/metabolismo , Vírus da Raiva/metabolismo , Vírus da Raiva/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/virologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/virologia , Fatores de Tempo , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/metabolismo
7.
J Virol ; 87(3): 1834-41, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23192867

RESUMO

A single intramuscular application of the live but not UV-inactivated recombinant rabies virus (RABV) variant TriGAS in mice induces the robust and sustained production of RABV-neutralizing antibodies that correlate with long-term protection against challenge with an otherwise lethal dose of the wild-type RABV. To obtain insight into the mechanism by which live TriGAS induces long-lasting protective immunity, quantitative PCR (qPCR) analysis of muscle tissue, draining lymph nodes, spleen, spinal cord, and brain at different times after TriGAS inoculation revealed the presence of significant copy numbers of RABV-specific RNA in muscle, lymph node, and to a lesser extent, spleen for several days postinfection. Notably, no significant amounts of RABV RNA were detected in brain or spinal cord at any time after TriGAS inoculation. Differential qPCR analysis revealed that the RABV-specific RNA detected in muscle is predominantly genomic RNA, whereas RABV RNA detected in draining lymph nodes is predominantly mRNA. Comparison of genomic RNA and mRNA obtained from isolated lymph node cells showed the highest mRNA-to-genomic-RNA ratios in B cells and dendritic cells (DCs), suggesting that these cells represent the major cell population that is infected in the lymph node. Since RABV RNA declined to undetectable levels by 14 days postinoculation of TriGAS, we speculate that a transient infection of DCs with TriGAS may be highly immunostimulatory through mechanisms that enhance antigen presentation. Our results support the superior efficacy and safety of TriGAS and advocate for its utility as a vaccine.


Assuntos
Linfonodos/virologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Animais , Linfócitos B/virologia , Encéfalo/patologia , Encéfalo/virologia , Células Dendríticas/virologia , Feminino , Injeções Intramusculares , Linfonodos/imunologia , Linfonodos/patologia , Camundongos , Músculos/patologia , Músculos/virologia , RNA Viral/análise , RNA Viral/genética , Raiva/virologia , Vacina Antirrábica/administração & dosagem , Vírus da Raiva/patogenicidade , Reação em Cadeia da Polimerase em Tempo Real , Medula Espinal/patologia , Medula Espinal/virologia , Baço/patologia , Baço/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
8.
Future Virol ; 6(3): 387-397, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21686076

RESUMO

The long-held concept that rabies infection is lethal in humans once the causative rabies virus has reached the CNS has been called into question by the recent survival of a number of patients with clinical rabies. Studies in animal models provide insight into why survival from a rabies virus infection that has spread to the CNS is possible and the immune mechanisms involved. In the CNS, both innate mechanisms capable of inhibiting virus replication and the activity of infiltrating rabies virus-specific T and B cells with the capacity to clear the virus are required. Deficiencies in the induction of either aspect of rabies immunity can lead to lethal consequences but may be overcome by novel approaches to active and passive immunization.

9.
Adv Virus Res ; 79: 55-71, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21601042

RESUMO

Rabies, a neurological disease associated with replication in central nervous system (CNS) tissues of any of a number of rabies viruses endemic in nature, is generally fatal. Prophylactic medical intervention is immune mediated and directed at preventing the spread of the virus from a peripheral site of exposure to the CNS. While individuals rarely develop immune responses capable of clearing the virus from CNS tissues, a variety of laboratory-attenuated rabies viruses are readily cleared from the CNS tissues in animal models. By comparing immune responses to wild-type and attenuated rabies viruses in these models, we have discovered that the latter induce processes required for immune effector infiltration into CNS tissues that are absent from lethal infections. Predominant among these are activities of cells of the neurovascular unit (NVU) that promote an interaction with circulating immune cells. In the absence of this interaction, the specialized barrier function of the NVU remains intact and circulating virus-specific immune effectors are largely excluded from infected CNS tissues. Studies of mixed infections with wild-type and attenuated rabies viruses reveal that wild-type rabies viruses fail to trigger, rather than inhibit, the interactions between immune cells and the NVU required for virus clearance from the CNS. These studies provide insights into how immune effectors with the capacity to clear the virus may be delivered into CNS tissues to contain a wild-type rabies virus infection. However, to apply immunotherapeutic strategies beyond the initial stages of CNS infection, further insights into the fate of the infected cells during virus clearance are needed.


Assuntos
Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Evasão da Resposta Imune , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Animais , Modelos Animais de Doenças , Cães , Humanos , Imunidade Celular , Imunidade Humoral , Virulência
10.
Proc Natl Acad Sci U S A ; 106(27): 11300-5, 2009 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-19581599

RESUMO

Rabies remains an important public health problem with more than 95% of all human rabies cases caused by exposure to rabid dogs in areas where effective, inexpensive vaccines are unavailable. Because of their ability to induce strong innate and adaptive immune responses capable of clearing the infection from the CNS after a single immunization, live-attenuated rabies virus (RV) vaccines could be particularly useful not only for the global eradication of canine rabies but also for late-stage rabies postexposure prophylaxis of humans. To overcome concerns regarding the safety of live-attenuated RV vaccines, we developed the highly attenuated triple RV G variant, SPBAANGAS-GAS-GAS. In contrast to most attenuated recombinant RVs generated thus far, SPBAANGAS-GAS-GAS is completely nonpathogenic after intracranial infection of mice that are either developmentally immunocompromised (e.g., 5-day-old mice) or have inherited deficits in immune function (e.g., antibody production or type I IFN signaling), as well as normal adult animals. In addition, SPBAANGAS-GAS-GAS induces immune mechanisms capable of containing a CNS infection with pathogenic RV, thereby preventing lethal rabies encephalopathy. The lack of pathogenicity together with excellent immunogenicity and the capacity to deliver immune effectors to CNS tissues makes SPBAANGAS-GAS-GAS a promising vaccine candidate for both the preexposure and postexposure prophylaxis of rabies.


Assuntos
Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Raiva/prevenção & controle , Envelhecimento/imunologia , Animais , Animais Lactentes , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/virologia , Hospedeiro Imunocomprometido , Camundongos , Permeabilidade , Vírus da Raiva/patogenicidade , Análise de Sobrevida , Resultado do Tratamento , Vacinação , Vacinas Atenuadas
11.
Proc Natl Acad Sci U S A ; 105(40): 15511-6, 2008 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-18829442

RESUMO

CNS tissues are protected from circulating cells and factors by the blood-brain barrier (BBB), a specialization of the neurovasculature. Outcomes of the loss of BBB integrity and cell infiltration into CNS tissues can differ vastly. For example, elevated BBB permeability is closely associated with the development of neurological disease in experimental allergic encephalomyelitis (EAE) but not during clearance of the attenuated rabies virus CVS-F3 from the CNS tissues. To probe whether differences in the nature of BBB permeability changes may contribute to the pathogenesis of acute neuroinflammatory disease, we compared the characteristics of BBB permeability changes in mice with EAE and in mice clearing CVS-F3. BBB permeability changes are largely restricted to the cerebellum and spinal cord in both models but differ in the extent of leakage of markers of different size and in the nature of cell accumulation in the CNS tissues. The accumulation in the CNS tissues of CD4 T cells expressing mRNAs specific for IFN-gamma and IL-17 is common to both, but iNOS-positive cells invade into the CNS parenchyma only in EAE. Mice that have been immunized with myelin basic protein (MBP) and infected exhibit the features of EAE. Treatment with the peroxynitrite-dependent radical scavenger urate inhibits the invasion of iNOS-positive cells into the CNS tissues and the development of clinical signs of EAE without preventing the loss of BBB integrity in immunized/infected animals. These findings indicate that BBB permeability changes can occur in the absence of neuropathology provided that cell invasion is restricted.


Assuntos
Autoimunidade , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/virologia , Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/imunologia , Raiva/imunologia , Animais , Barreira Hematoencefálica/patologia , Movimento Celular , Cerebelo/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Sequestradores de Radicais Livres/metabolismo , Imuno-Histoquímica , Interferon gama/imunologia , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos , Óxido Nítrico Sintase/metabolismo , Ácido Peroxinitroso/metabolismo , Ácido Peroxinitroso/uso terapêutico , Raiva/metabolismo , Vírus da Raiva/imunologia , Vírus da Raiva/metabolismo
12.
J Neuroimmunol ; 196(1-2): 8-15, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18406473

RESUMO

The blood-brain barrier (BBB) is dramatically but transiently compromised in the cerebella of myelin basic protein immunized mice at least 1 week prior to the development of the paralytic phase of experimental allergic encephalomyelitis (EAE). Treatment of mice with the peroxynitrite-dependent radical scavenger uric acid (UA) during the first week after immunization blocks the early increase in cerebellar BBB permeability and the subsequent development of clinical signs of EAE. These results indicate that the early loss of BBB integrity in the cerebellum is likely to be a necessary step in the development of paralytic EAE.


Assuntos
Barreira Hematoencefálica/imunologia , Cerebelo/patologia , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/fisiopatologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/imunologia , Cerebelo/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Feminino , Fluoresceína , Regulação da Expressão Gênica/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/metabolismo , Camundongos , Proteína Básica da Mielina/imunologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Estatísticas não Paramétricas , Fatores de Tempo
13.
J Immunol ; 178(11): 7334-43, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17513784

RESUMO

Elevated blood-brain barrier (BBB) permeability is associated with both the protective and pathological invasion of immune and inflammatory cells into CNS tissues. Although a variety of processes have been implicated in the changes at the BBB that result in the loss of integrity, there has been no consensus as to their induction. TNF-alpha has often been proposed to be responsible for increased BBB permeability but there is accumulating evidence that peroxynitrite (ONOO(-))-dependent radicals may be the direct trigger. We demonstrate here that enhanced BBB permeability in mice, whether associated with rabies virus (RV) clearance or CNS autoimmunity, is unaltered in the absence of TNF-alpha. Moreover, the induction of TNF-alpha expression in CNS tissues by RV infection has no impact on BBB integrity in the absence of T cells. CD4 T cells are required to enhance BBB permeability in response to the CNS infection whereas CD8 T cells and B cells are not. Like CNS autoimmunity, elevated BBB permeability in response to RV infection is evidently mediated by ONOO(-). However, as opposed to the invading cells producing ONOO(-) that have been implicated in the pathogenesis of CNS inflammation, during virus clearance ONOO(-) is produced without pathological sequelae by IFN-gamma-stimulated neurovascular endothelial cells.


Assuntos
Barreira Hematoencefálica/imunologia , Permeabilidade da Membrana Celular/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Ácido Peroxinitroso/fisiologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Permeabilidade da Membrana Celular/genética , Movimento Celular/genética , Movimento Celular/imunologia , Cerebelo/imunologia , Cerebelo/patologia , Cerebelo/virologia , Encefalomielite Autoimune Experimental/fisiopatologia , Encefalomielite Autoimune Experimental/virologia , Feminino , Linfopenia/imunologia , Linfopenia/patologia , Linfopenia/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Raiva/imunologia , Transdução de Sinais/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Subpopulações de Linfócitos T/virologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Tirosina/análogos & derivados , Tirosina/metabolismo , Carga Viral
14.
Proc Natl Acad Sci U S A ; 104(13): 5656-61, 2007 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-17372191

RESUMO

Experimental allergic encephalomyelitis (EAE) is an inflammatory demyelinating disease of the CNS that is used to model certain parameters of multiple sclerosis. To establish the relative contributions of T cell reactivity, the loss of blood-brain barrier (BBB) integrity, CNS inflammation, and lesion formation toward the pathogenesis of EAE, we assessed the incidence of EAE and these parameters in mice lacking NF-kappaB, TNF-alpha, IFN-alphabeta receptors, IFN-gamma receptors, and inducible nitric oxide synthase. Although increased myelin oligodendrocyte glycoprotein-specific T cell reactivity was generally associated with a more rapid onset or increased disease severity, the loss of BBB integrity and cell accumulation in spinal cord tissues was invariably associated with the development of neurological disease signs. Histological and real-time RT-PCR analyses revealed differences in the nature of immune/inflammatory cell accumulation in the spinal cord tissues of the different mouse strains. On the other hand, disease severity during the acute phase of EAE directly correlated with the extent of BBB permeability. Thus, the loss of BBB integrity seems to be a requisite event in the development of EAE and can occur in the absence of important inflammatory mediators.


Assuntos
Barreira Hematoencefálica , Encefalomielite Autoimune Experimental/genética , Medula Espinal/patologia , Animais , Proliferação de Células , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Feminino , Interferon gama/metabolismo , Masculino , Camundongos , Camundongos Knockout , Permeabilidade , Fatores Sexuais , Medula Espinal/metabolismo , Linfócitos T/citologia
15.
J Immunol ; 176(12): 7666-75, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16751414

RESUMO

The loss of blood-brain barrier (BBB) integrity in CNS inflammatory responses triggered by infection and autoimmunity has generally been associated with the development of neurological signs. In the present study, we demonstrate that the clearance of the attenuated rabies virus CVS-F3 from the CNS is an exception; increased BBB permeability and CNS inflammation occurs in the absence of neurological sequelae. We speculate that regionalization of the CNS inflammatory response contributes to its lack of pathogenicity. Despite virus replication and the expression of several chemokines and IL-6 in both regions being similar, the up-regulation of MIP-1beta, TNF-alpha, IFN-gamma, and ICAM-1 and the loss of BBB integrity was more extensive in the cerebellum than in the cerebral cortex. The accumulation of CD4- and CD19-positive cells was higher in the cerebellum than the cerebral cortex. Elevated CD19 levels were paralleled by kappa-L chain expression levels. The timing of BBB permeability changes, kappa-L chain expression in CNS tissues, and Ab production in the periphery suggest that the in situ production of virus-neutralizing Ab may be more important in virus clearance than the infiltration of circulating Ab. The data indicate that, with the possible exception of CD8 T cells, the effectors of rabies virus clearance are more commonly targeted to the cerebellum. This is likely the result of differences in the capacity of the tissues of the cerebellum and cerebral cortex to mediate the events required for BBB permeability changes and cell invasion during virus infection.


Assuntos
Barreira Hematoencefálica/imunologia , Permeabilidade da Membrana Celular/imunologia , Cerebelo/patologia , Cerebelo/virologia , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Vírus da Raiva/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/biossíntese , Cerebelo/fisiopatologia , Córtex Cerebral/fisiopatologia , Quimiocinas/biossíntese , Feminino , Inflamação/imunologia , Inflamação/fisiopatologia , Inflamação/virologia , Molécula 1 de Adesão Intercelular/biossíntese , Interferon gama/biossíntese , Interleucina-6/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos , Raiva/imunologia , Raiva/patologia , Raiva/fisiopatologia , Raiva/virologia , Vírus da Raiva/crescimento & desenvolvimento , Vírus da Raiva/patogenicidade , Fator de Necrose Tumoral alfa/biossíntese
16.
J Neuroimmunol ; 155(1-2): 32-42, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15342194

RESUMO

Urate (UA) selectively scavenges peroxynitrite-dependent radicals and suppresses CNS inflammation through effects that are manifested at the blood-brain barrier (BBB). ICAM-1 upregulation in the spinal cord tissues of myelin basic protein (MBP) immunized mice is selectively inhibited by UA treatment. In contrast, the expression of ICAM-1 and other adhesion molecules by circulating cells is unchanged. Moreover, TNF-alpha expression in the CNS tissues of MBP-immunized mice is suppressed by UA treatment but TNF-alpha-induced ICAM-1 expression on neurovascular endothelial cells is not. Therefore the effect of UA on ICAM-1 upregulation in the CNS tissues is likely due to its known contribution to the maintenance of BBB integrity in MBP-immunized mice which in turn inhibits cell invasion into the CNS and prevents TNF-alpha production in CNS tissues.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Encefalomielite Autoimune Experimental/tratamento farmacológico , Molécula 1 de Adesão Intercelular/metabolismo , Medula Espinal/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Ácido Úrico/farmacologia , Animais , Barreira Hematoencefálica/imunologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/imunologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Feminino , Sequestradores de Radicais Livres/farmacologia , Sequestradores de Radicais Livres/uso terapêutico , Radicais Livres/imunologia , Radicais Livres/metabolismo , Molécula 1 de Adesão Intercelular/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/imunologia , Camundongos , Proteína Básica da Mielina/imunologia , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/metabolismo , Óxido Nítrico Sintase/efeitos dos fármacos , Óxido Nítrico Sintase/imunologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Ácido Peroxinitroso/metabolismo , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/imunologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia , Ácido Úrico/uso terapêutico
17.
J Pharmacol Exp Ther ; 310(3): 1053-61, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15159442

RESUMO

Poly(ADP-ribose) polymerase (PARP) activity has been implicated in the pathogenesis of several central nervous system (CNS) disorders. For example, the presence of extensive poly(ADP)ribosylation in CNS tissues from animals with experimental allergic encephalomyelitis (EAE) indicates that PARP activity may be involved in this inflammatory disease process. Using PJ34 [N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-N, N-dimethylacetamide.HCl], a selective PARP inhibitor, we studied the mechanisms through which PARP activity may contribute to the onset of acute EAE. PLSJL mice immunized with myelin antigens were treated with PJ34, and the effects on the progression of EAE and several other parameters relevant to the disease process were assessed. PJ34 exerted therapeutic effects at the onset of EAE that were associated with reduced CNS inflammation and the maintenance of neurovascular integrity. Expression of genes encoding the intercellular adhesion molecule-1 (ICAM-1) and the inflammatory mediators interferon-gamma, tumor necrosis factor-alpha, and inducible nitric-oxide synthase were decreased in CNS tissues from drug-treated animals. Administration of PJ34 biased the class of myelin basic protein (MBP)-specific antibodies elicited from IgG2a to IgG1 and IgG2b and modulated antigen-specific T-cell reactivity. Therefore, the mode of action of PJ34 at the onset of EAE is likely mediated by a shift in the MBP-specific immune response from a proinflammatory Th1 toward an anti-inflammatory Th2 phenotype.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Fenantrenos/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases , Adjuvantes Imunológicos/farmacologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Modelos Animais de Doenças , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , Proteína Básica da Mielina/administração & dosagem , Fenantrenos/farmacologia , Doenças da Medula Espinal/tratamento farmacológico , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Células Th2/efeitos dos fármacos , Células Th2/imunologia , Fator de Necrose Tumoral alfa/metabolismo
18.
Free Radic Biol Med ; 33(10): 1363-71, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12419468

RESUMO

Serum levels of uric acid (UA), an inhibitor of peroxynitrite- (ONOO-) related chemical reactions, became elevated approximately 30 million years ago in hominid evolution. During a similar time frame, higher mammals lost the ability to synthesize another important radical scavenger, ascorbic acid (AA), leading to the suggestion that UA may have replaced AA as an antioxidant. However, in vivo treatment with AA does not protect against the development of experimental allergic encephalomyelitis (EAE), a disease that has been associated with the activity of ONOO- and is inhibited by UA. When compared in vitro, UA and AA were found to have similar capacities to inhibit the nitrating properties of ONOO-. However UA and AA had different capacities to prevent ONOO- -mediated oxidation, especially in the presence of iron ion (Fe3+). While UA at physiological concentrations effectively blocked dihydrorhodamine-123 oxidation in the presence of Fe3+, AA did not, regardless of whether the source of ONOO- was synthetic ONOO-, SIN-1, or RAW 264.7 cells. AA also potentiated lipid peroxidation in vivo and in vitro. In conclusion, the superior protective properties of UA in EAE may be related to its ability to neutralize the oxidative properties of ONOO- in the presence of free iron ions.


Assuntos
Ácido Ascórbico/farmacologia , Encefalomielite Autoimune Experimental/prevenção & controle , Molsidomina/análogos & derivados , Tirosina/análogos & derivados , Ácido Úrico/farmacologia , Albuminas/metabolismo , Animais , Antioxidantes/farmacologia , Ácido Ascórbico/sangue , Barreira Hematoencefálica , Linhagem Celular , Radicais Livres , Imuno-Histoquímica , Ferro/farmacologia , Peroxidação de Lipídeos , Camundongos , Molsidomina/farmacologia , Bainha de Mielina/metabolismo , Óxido Nítrico/metabolismo , Oxigênio/metabolismo , Ácido Peroxinitroso/antagonistas & inibidores , Rodaminas/farmacologia , Fatores de Tempo , Tirosina/farmacologia , Ácido Úrico/sangue
19.
Proc Natl Acad Sci U S A ; 99(25): 16303-8, 2002 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-12451183

RESUMO

Uric acid (UA) is a purine metabolite that selectively inhibits peroxynitrite-mediated reactions implicated in the pathogenesis of multiple sclerosis (MS) and other neurodegenerative diseases. Serum UA levels are inversely associated with the incidence of MS in humans because MS patients have low serum UA levels and individuals with hyperuricemia (gout) rarely develop the disease. Moreover, the administration of UA is therapeutic in experimental allergic encephalomyelitis (EAE), an animal model of MS. Thus, raising serum UA levels in MS patients, by oral administration of a UA precursor such as inosine, may have therapeutic value. We have assessed the effects of inosine, as well as inosinic acid, on parameters relevant to the chemical reactivity of peroxynitrite and the pathogenesis of EAE. Both had no effect on chemical reactions associated with peroxynitrite, such as tyrosine nitration, or on the activation of inflammatory cells in vitro. Moreover, when mice treated with the urate oxidase inhibitor potassium oxonate were fed inosine or inosinic acid, serum UA levels were elevated markedly for a period of hours, whereas only a minor, transient increase in serum inosine was detected. Administration of inosinic acid suppressed the appearance of clinical signs of EAE and promoted recovery from ongoing disease. The therapeutic effect on animals with active EAE was associated with increased UA, but not inosine, levels in CNS tissue. We, therefore, conclude that the mode of action of inosine and inosinic acid in EAE is via their metabolism to UA.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Inosina Monofosfato/uso terapêutico , Inosina/uso terapêutico , Molsidomina/análogos & derivados , Ácido Úrico/metabolismo , Administração Oral , Animais , Biotransformação , Cromatografia Líquida de Alta Pressão , Avaliação Pré-Clínica de Medicamentos , Encefalomielite Autoimune Experimental/metabolismo , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Inosina/farmacocinética , Inosina Monofosfato/farmacocinética , Camundongos , Molsidomina/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Esclerose Múltipla/metabolismo , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Oxirredução , Estresse Oxidativo , Ácido Oxônico/farmacologia , Ácido Peroxinitroso/metabolismo , Urato Oxidase/antagonistas & inibidores , Ácido Úrico/análogos & derivados , Ácido Úrico/farmacologia , Xantinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA