Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stud Health Technol Inform ; 305: 240-243, 2023 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-37387007

RESUMO

A quantitative approach in the form of an online questionnaire was used to identify challenges and desires related to the Care Records Transmission Process and Care Transition Records (CTR). The questionnaire was sent to nurses, nursing assistants, and trainees working in ambulatory, acute inpatient, or long-term care settings. The survey revealed that creating CTRs is time-consuming, and the lack of standardization of CTRs makes the process even more cumbersome. In addition, most facilities transmit the CTR by physically handing it over to the patient or resident, resulting in little or no preparation time for the individual(s) receiving care. The key findings also suggest that most respondents are only partially satisfied with the completeness of the CTRs and that they must conduct additional interviews to obtain missing information. However, most respondents hoped that digital transmission of CTRs would lead to less administrative burden and that standardization of CTRs would be encouraged.


Assuntos
Hospitais , Transferência de Pacientes , Humanos , Alemanha , Mãos , Pacientes Internados
2.
Immunol Cell Biol ; 95(10): 884-894, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28722018

RESUMO

Intracellular serpins are proposed to inactivate proteases released from lysosome-related organelles into the host cell interior, preventing cell death. Serpinb9 opposes the immune cytotoxic protease, granzyme B, and in a number of settings protects cells against granzyme B-mediated cell death. Using a knockout mouse line engineered to express green fluorescent protein under the serpbinb9 promoter, we demonstrate that serpinb9 is vital for host survival during Ectromelia virus infection by maintaining both mature natural killer NK) cells, and activated CD8+ T cells. Serpinb9 expression parallels granzyme B expression within both populations during infection. Maturing serpinb9-null NK cells exhibit higher levels of granzyme B-mediated apoptosis during infection; hence there are fewer mature NK cells, and these cells also have lower cytotoxic potential. Thus the serpinb9-granzyme B axis is important for homeostasis of both major cytotoxic effector cell populations.


Assuntos
Granzimas/antagonistas & inibidores , Células Matadoras Naturais/imunologia , Proteínas de Membrana/farmacologia , Infecções por Poxviridae/imunologia , Poxviridae/imunologia , Serpinas/farmacologia , Animais , Morte Celular , Sobrevivência Celular , Homeostase , Humanos , Espaço Intracelular , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Virus Res ; 228: 61-65, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27865865

RESUMO

Mousepox is caused by the orthopoxvirus ectromelia virus (ECTV), and is thought to be transmitted via skin abrasions. We studied the ECTV virulence factor N1 following subcutaneous infection of mousepox-susceptible BALB/c mice. In this model, ECTV lacking N1L gene was attenuated more than 1000-fold compared with wild-type virus and replication was profoundly reduced as early as four days after infection. However, in contrast to data from an intranasal model, N1 protein was not required for virus dissemination. Further, neither T cell nor cytokine responses were enhanced in the absence of N1. Together with the early timing of reduced virus titres, this suggests that in a cutaneous model, N1 exerts its function at the level of infected cells or in the inhibition of the very earliest effectors of innate immunity.


Assuntos
Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/virologia , Proteínas Virais/genética , Animais , Interações Hospedeiro-Patógeno , Camundongos , Carga Viral , Proteínas Virais/metabolismo , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Replicação Viral
4.
Immunity ; 41(6): 960-72, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25526309

RESUMO

Granzyme B (GzmB) is a protease with a well-characterized intracellular role in targeted destruction of compromised cells by cytotoxic lymphocytes. However, GzmB also cleaves extracellular matrix components, suggesting that it influences the interplay between cytotoxic lymphocytes and their environment. Here, we show that GzmB-null effector T cells and natural killer (NK) cells exhibited a cell-autonomous homing deficit in mouse models of inflammation and Ectromelia virus infection. Intravital imaging of effector T cells in inflamed cremaster muscle venules revealed that GzmB-null cells adhered normally to the vessel wall and could extend lamellipodia through it but did not cross it efficiently. In vitro migration assays showed that active GzmB was released from migrating cytotoxic lymphocytes and enabled chemokine-driven movement through basement membranes. Finally, proteomic analysis demonstrated that GzmB cleaved basement membrane constituents. Our results highlight an important role for GzmB in expediting cytotoxic lymphocyte diapedesis via basement membrane remodeling.


Assuntos
Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Granzimas/metabolismo , Células Matadoras Naturais/fisiologia , Linfócitos T Citotóxicos/fisiologia , Animais , Membrana Basal/metabolismo , Movimento Celular/genética , Células Cultivadas , Quimiocinas/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Granzimas/genética , Células Matadoras Naturais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteólise , Linfócitos T Citotóxicos/virologia , Migração Transendotelial e Transepitelial/genética
5.
Eur J Immunol ; 43(7): 1789-98, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23568450

RESUMO

Japanese encephalitis, caused by infection with the neurotropic flavivirus, Japanese encephalitis virus (JEV), is among the most important viral encephalitides in Asia. While previous studies established an essential role of Ab and type I IFN, it is still unclear if the cell-mediated immune responses, through their direct antiviral effector functions, contribute to protection against the fatal disease. We report here that mice defective in both the granule exocytosis and death receptor pathways of cytotoxicity display increased susceptibility to JEV. The two cell contact-dependent cytotoxic effector mechanisms act redundantly within the CNS to reduce disease severity. We also demonstrate that IFN-γ is critical in recovery from primary infection with JEV by a mechanism involving suppression of virus growth in the CNS, and that T cells are the main source of the cytokine that promotes viral clearance from the brain. Finally, we show by in vivo depletion of NK cells that this innate immune cell population is dispensable for control of JEV infection in the periphery and in the CNS. Accordingly, cell contact-dependent cytolytic and IFN-γ-dependent noncytolytic clearance of virus mediated by T cells trafficking into the CNS help in recovery from lethal infection in a mouse model of Japanese encephalitis.


Assuntos
Citotoxicidade Imunológica/imunologia , Encefalite Japonesa/imunologia , Interferon gama/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Modelos Animais de Doenças , Imunidade Celular/imunologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
PLoS One ; 7(9): e44834, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028638

RESUMO

Japanese encephalitis is a severe central nervous system (CNS) inflammatory disease caused by the mosquito-borne flavivirus, Japanese encephalitis virus (JEV). In the current study we have investigated the immune responses against JEV in mice lacking expression of the chemokine receptor CCR5, which functions in activation and chemotaxis of leukocytes during infection. We show that CCR5 serves as a host antiviral factor against Japanese encephalitis, with CCR5 deficiency markedly increasing mortality, and viral burden in the CNS. Humoral immune responses, which are essential in recovery from JEV infection, were of similar magnitude in CCR5 sufficient and deficient mice. However, absence of CCR5 resulted in a multifaceted deficiency of cellular immune responses characterized by reduced natural killer and CD8⁺ T cell activity, low splenic cellularity, and impaired trafficking of leukocytes to the brain. Interestingly, adoptive transfer of immune spleen cells, depleted of B lymphocytes, increased resistance of CCR5-deficient recipient mice against JEV regardless of whether the cells were obtained from CCR5-deficient or wild-type donor mice, and only when transferred at one but not at three days post-challenge. This result is consistent with a mechanism by which CCR5 expression enhances lymphocyte activation and thereby promotes host survival in Japanese encephalitis.


Assuntos
Antagonistas dos Receptores CCR5 , Encefalite Japonesa/imunologia , Encefalite Japonesa/metabolismo , Infecções por HIV/tratamento farmacológico , Terapia de Alvo Molecular , Receptores CCR5/metabolismo , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Imunidade Humoral/efeitos dos fármacos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Camundongos , Receptores CCR5/deficiência , Baço/citologia , Análise de Sobrevida , Células Vero
7.
PLoS One ; 6(10): e25765, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21998693

RESUMO

BACKGROUND: We have shown previously in mice, that infection with live viruses, including influenza/A and Semliki Forest virus (SFV), induces systemic partial activation of lymphocytes, characterized by cell surface expression of CD69 and CD86, but not CD25. This partial lymphocytes activation is mediated by type-I interferons (IFN-I). Importantly, we have shown that γ-irradiated SFV does not induce IFN-I and the associated lymphocyte activation. PRINCIPAL FINDINGS: Here we report that, in contrast to SFV, γ-irradiated influenza A virus elicits partial lymphocyte activation in vivo. Furthermore, we show that when using influenza viruses inactivated by a variety of methods (UV, ionising radiation and formalin treatment), as well as commercially available influenza vaccines, only γ-irradiated influenza virus is able to trigger IFN-I-dependent partial lymphocyte activation in the absence of the TLR7/MyD88 signalling pathways. CONCLUSIONS: Our data suggest an important mechanism for the recognition of γ-irradiated influenza vaccine by cytosolic receptors, which correspond with the ability of γ-irradiated influenza virus to induce cross-reactive and cross-protective cytotoxic T cell responses.


Assuntos
Raios gama , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H1N1/efeitos da radiação , Vírus da Influenza A Subtipo H3N2/fisiologia , Vírus da Influenza A Subtipo H3N2/efeitos da radiação , Interferon Tipo I/metabolismo , Linfócitos/virologia , Animais , Linhagem Celular , Cricetinae , Cães , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Influenza A Subtipo H1N1/metabolismo , Vírus da Influenza A Subtipo H3N2/metabolismo , Interferon Tipo I/biossíntese , Linfócitos/citologia , Linfócitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Neuraminidase/metabolismo , Transdução de Sinais , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/virologia , Receptor 7 Toll-Like/metabolismo , Ativação Viral/efeitos da radiação
8.
PLoS One ; 6(8): e23252, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21853094

RESUMO

NK cells kill target cells mainly via exocytosis of granules containing perforin (perf) and granzymes (gzm). In vitro, gzm delivery into the target cell cytosol results in apoptosis, and induction of apoptosis is severely impaired in the absence of gzm A and B. However, their importance for in vivo cytotoxicity by cytotoxic T cells has been questioned. We used an in vivo NK cytotoxicity assay, in which splenocytes from wild-type and ß(2)microglobulin-deficient (MHC-I(neg)) mice are co-injected into recipients whose NK cells were activated by virus infection or synthetic Toll-like receptor ligands. Elimination of adoptively transferred MHC-I(neg) splenocytes was unimpaired in the absence of gzmA and gzmB, but dependent on perforin. This target cell rejection was NK cell dependent, since NK cell depletion abrogated it. Furthermore, target cell elimination in vivo was equally rapid in both wild-type and gzmAxB-deficient recipients, with the majority of specific target cells lost from lymphoid tissue within less than one to two hours after transfer. Thus, similar to T cell cytotoxicity, the contribution of gzmA and B to in vivo target cell elimination remains unresolved.


Assuntos
Antígenos de Histocompatibilidade Classe I/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Animais , Citotoxicidade Imunológica/genética , Feminino , Regulação da Expressão Gênica , Granzimas/deficiência , Granzimas/metabolismo , Células Matadoras Naturais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Perforina/metabolismo , Vírus da Floresta de Semliki/fisiologia
9.
J Virol ; 85(21): 11170-82, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21849445

RESUMO

Ectromelia virus (ECTV) is a natural pathogen of mice that causes mousepox, and many of its genes have been implicated in the modulation of host immune responses. Serine protease inhibitor 2 (SPI-2) is one of these putative ECTV host response modifier proteins. SPI-2 is conserved across orthopoxviruses, but results defining its mechanism of action and in vivo function are lacking or contradictory. We studied the role of SPI-2 in mousepox by deleting the SPI-2 gene or its serine protease inhibitor reactive site. We found that SPI-2 does not affect viral replication or cell-intrinsic apoptosis pathways, since mutant viruses replicate in vitro as efficiently as wild-type virus. However, in the absence of SPI-2 protein, ECTV is attenuated in mousepox-susceptible mice, resulting in lower viral loads in the liver, decreased spleen pathology, and substantially improved host survival. This attenuation correlates with more effective immune responses in the absence of SPI-2, including an earlier serum gamma interferon (IFN-γ) response, raised serum interleukin 18 (IL-18), increased numbers of granzyme B(+) CD8(+) T cells, and, most notably, increased numbers and activation of NK cells. Both virus attenuation and the improved immune responses associated with SPI-2 deletion from ECTV are lost when mice are depleted of NK cells. Consequently, SPI-2 renders mousepox lethal in susceptible strains by preventing protective NK cell defenses.


Assuntos
Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/mortalidade , Interações Hospedeiro-Patógeno , Células Matadoras Naturais/imunologia , Serpinas/metabolismo , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Vírus da Ectromelia/genética , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/virologia , Deleção de Genes , Interferon gama/metabolismo , Interleucina-18/metabolismo , Fígado/virologia , Subpopulações de Linfócitos/química , Subpopulações de Linfócitos/imunologia , Camundongos , Serpinas/genética , Baço/patologia , Análise de Sobrevida , Carga Viral , Proteínas Virais/genética , Replicação Viral
10.
Eur J Immunol ; 41(7): 1948-57, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21590764

RESUMO

Cytotoxic T (Tc) cells play a key role in the defense against virus infections. Tc cells recognize infected cells via the T-cell receptor (TCR) and subsequently kill the target cells by one or more cytotoxic mechanisms. Induction of the cytotoxic mechanisms is finely tuned by the activation signals from the TCR. To determine whether TCR down-regulation affects the cytotoxicity of Tc cells, we studied TCR down-regulation-deficient CD3γLLAA mice. We found that Tc cells from CD3γLLAA mice have reduced cytotoxicity due to a specific deficiency in exocytosis of lytic granules. To determine whether this defect was reflected in an increased susceptibility to virus infections, we studied the course of ectromelia virus (ECTV) infection. We found that the susceptibility to ECTV infection was significantly increased in CD3γLLAA mice with a mortality rate almost as high as in granzyme B knock-out mice. Finally, we found that TCR signaling in CD3γLLAA Tc cells caused highly increased tyrosine phosphorylation and activation of the c-Cbl ubiquitin ligase, and that the impaired exocytosis of lytic granules could be rescued by the knockdown of c-Cbl. Thus, our work demonstrates that TCR down-regulation critically increases Tc cell cytotoxicity and protection against poxvirus infection.


Assuntos
Citotoxicidade Imunológica , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Western Blotting , Complexo CD3/genética , Complexo CD3/imunologia , Linhagem Celular , Exocitose , Granzimas/metabolismo , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Perforina/biossíntese , Perforina/genética , Fosforilação , Proteínas Proto-Oncogênicas c-cbl/genética , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T Citotóxicos/metabolismo , Ubiquitina-Proteína Ligases
11.
J Virol ; 85(11): 5446-55, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21450826

RESUMO

The immunological correlates for recovery from primary Japanese encephalitis virus (JEV) infection in humans and experimental animals remain poorly defined. To investigate the relative importance of the adaptive immune responses, we have established a mouse model for Japanese encephalitis in which a low-dose virus inoculum was administered into the footpads of adult C57BL/6 mice. In this model, ~60% of the mice developed a fatal encephalitis and a virus burden in the central nervous system (CNS). Using mice lacking B cells (µMT(-/-) mice) and immune B cell transfer to wild-type mice, we show a critically important role for humoral immunity in preventing virus spread to the CNS. T cell help played an essential part in the maintenance of an effective antibody response necessary to combat the infection, since mice lacking major histocompatibility complex class II showed truncated IgM and blunted IgG responses and uniformly high lethality. JEV infection resulted in extensive CD8(+) T cell activation, judged by upregulation of surface markers CD69 and CD25 and cytokine production after stimulation with a JEV NS4B protein-derived H-2D(b)-binding peptide and trafficking of virus-immune CD8(+) T cells into the CNS. However, no significant effect of CD8(+) T cells on the survival phenotype was found, which was corroborated in knockout mice lacking key effector molecules (Fas receptor, perforin, or granzymes) of cytolytic pathways triggered by T lymphocytes. Accordingly, CD8(+) T cells are mostly dispensable for recovery from infection with JEV. This finding highlights the conflicting role that CD8(+) T cells play in the pathogenesis of JEV and closely related encephalitic flaviviruses such as West Nile virus.


Assuntos
Anticorpos Antivirais/imunologia , Linfócitos T CD8-Positivos/imunologia , Encefalite Japonesa/imunologia , Animais , Sistema Nervoso Central/virologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite Japonesa/mortalidade , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças dos Roedores/imunologia , Doenças dos Roedores/mortalidade , Análise de Sobrevida
12.
Immunol Cell Biol ; 89(6): 706-15, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21173782

RESUMO

Using Toll-like receptor (TLR) and MyD88 gene knock-out (GKO) mice the effect of TLRs and MyD88 on virus replication, interferon (IFN)-ß production, natural killer (NK) cell and CD8T cell responses were assessed following ectromelia virus (ECTV) and recombinant vaccinia virus (rVV) infection. The capacity for rVVs encoding cytokines to restore immune function in MyD88(-/-) mice was clearly demonstrated. Results showed that TLR2(-/-), TLR4(-/-)and TLR7(-/-) mice survived ECTV infection whereas MyD88(-/-) and TLR9(-/-)mice, in contrast, were highly susceptible. Next, following infection with rVV, MyD88(-/-) mice elicited reduced serum IFN-ß, NK cell and CD8T cell responses compared with wild-type mice, whereas TLR9(-/-) mice showed elevated CD8T cell responses. When MyD88(-/-)mice were infected with rVV co-expressing IFN-ß these mice were able to restore IFN-ß levels and CD8T cell responses but not NK cell activation. Interestingly, even though rVV co-expressing interleukin (IL)-2 enhanced NK cell activation in MyD88(-/-) mice, this was not associated with an antiviral effect, as observed in normal mice. Surprisingly, co-infection with rVV IL-2/rVV IL-12, but not rVV IL-2/rVV IFN-ß, restored the attenuated phenotype of rVV IL-2 in MyD88(-/-) mice indicating that the IL-2/IL-12 combination promotes antiviral responses. Our results clearly show that the CD8T cell defect observed in MyD88(-/-) mice to vaccinia virus infection can be restored by rVV-encoding IFN-ß demonstrating the critical role of this cytokine in T cell mediated immunity and illustrates that the model can provide an effective platform for the elucidation of cytokine immunobiology.


Assuntos
Citocinas/genética , Fator 88 de Diferenciação Mieloide/genética , Receptores Toll-Like/genética , Vaccinia virus/genética , Vaccinia virus/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular , Chlorocebus aethiops , Citocinas/metabolismo , Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/prevenção & controle , Feminino , Regulação Viral da Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Interleucina-12/metabolismo , Interleucina-2/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Replicação Viral/imunologia
13.
Int Immunol ; 22(9): 757-67, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20682547

RESUMO

Type-I IFN (IFN-I) are highly pleiotropic cytokines known to modulate immune responses and play an early central role in mediating antiviral defenses. We have shown that IFN-I mediate transient up-regulation of a distinct subset of lymphocyte surface activation markers on both B and T cells in vivo independent of cognate antigen: a state referred to as 'partial lymphocyte activation'. Here we investigated in vitro the possibility that partial lymphocyte activation may serve to lower the antigen-specific activation thresholds for T cells. We found that the kinetics of Ca(2+) flux in T cells responding to TCR cross-linking was not enhanced in partially activated T cells. Furthermore, following TCR stimulation with anti-cluster of differentiation (CD) 3 epsilon, a lower proportion of partially activated than naive T cells proliferated. In contrast, the proliferation of partially activated and naive ovalbumin peptide (OVAp, SIINFEKL) specific CD8(+) T cells (OT-I CD8(+) T cells) was similar when stimulated with OVAp. Surprisingly, using an enzyme-linked immunospot (ELISPOT) assay for IFN-gamma secretion, we found that a higher number of partially activated OT-I CD8(+) T cells expressed effector functions than did naive OT-I CD8(+) T cells. This is most readily explained by an increased survival of activated antigen-specific CD8(+) T cells from a pool of partially activated T cells than naive T cells. Overall, when examining the effects of early (Ca(2+) flux), intermediate (proliferation) or late events (IFN-gamma secretion) of T-cell activation, we found that partial activation promotes the survival but does not alter the antigen-specific activation thresholds of CD8(+) T cells.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Interferon Tipo I/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Especificidade do Receptor de Antígeno de Linfócitos T , Subpopulações de Linfócitos T/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Antígenos/imunologia , Complexo CD3/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Interferon Tipo I/imunologia , Interferon gama/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/imunologia , Receptores de Antígenos de Linfócitos T/agonistas , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia
14.
J Gen Virol ; 91(Pt 6): 1450-60, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20147516

RESUMO

We have recently shown that intranasal (i.n.) administration of gamma-irradiated A/PR/8 [A/Puerto Rico/8/34 (H1N1)] protects mice against lethal avian influenza A/Vietnam/1203/2004 (H5N1) and other heterosubtypic influenza A infections. Here, we used gamma-irradiated, formalin- and UV-inactivated A/PC [A/Port Chalmers/1/73 (H3N2)] virus preparations and compared their ability to induce both homologous and heterosubtypic protective immunity. Our data show that, in contrast to i.n. vaccination with formalin- or UV-inactivated virus, or the present commercially available trivalent influenza vaccine, a single dose of gamma-ray-inactivated A/PC (gamma-A/PC) conferred significant protection in mice against both homologous and heterosubtypic virus challenges. A multiple immunization regime was required for formalin-inactivated virus preparations to induce protective immunity against a homotypic virus challenge, but did not induce influenza A strain cross-protective immunity. The highly immunogenic gamma-A/PC, but not formalin- or UV-inactivated A/PC, nor the currently available subvirion vaccine, elicited cytotoxic T-cell responses that are most likely responsible for the cross-protective and long-lasting immunity against highly lethal influenza A infections in mice. Finally, freeze-drying of gamma-A/PC did not affect the ability to induce cross-protective immunity.


Assuntos
Proteção Cruzada , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Inativação de Vírus , Animais , Peso Corporal , Feminino , Formaldeído/toxicidade , Raios gama , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Vírus da Influenza A Subtipo H3N2/efeitos da radiação , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia , Raios Ultravioleta , Vacinas de Produtos Inativados/imunologia , Carga Viral
15.
J Virol ; 84(9): 4212-21, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20164231

RESUMO

We previously demonstrated that a single dose of nonadjuvanted intranasal gamma-irradiated influenza A virus can provide robust protection in mice against both homologous and heterosubtypic challenges, including challenge with an H5N1 avian virus strain. We investigated the mechanism behind the observed cross-protection to define which arms of the adaptive immune response are involved in mediating this protection. Studies with gene knockout mice showed the cross-protective immunity to be mediated mainly by T cells and to be dependent on the cytolytic effector molecule perforin. Adoptive transfer of memory T cells from immunized mice, but not of memory B cells, protected naïve recipients against lethal heterosubtypic influenza virus challenge. Furthermore, gamma-irradiated influenza viruses induced cross-reactive Tc-cell responses but not cross-neutralizing or cross-protective antibodies. In addition, histological analysis showed reduced lung inflammation in vaccinated mice compared to that in unvaccinated controls following heterosubtypic challenge. This reduced inflammation was associated with enhanced early recruitment of T cells, both CD4(+) and CD8(+), and with early influenza virus-specific cytotoxic T-cell responses. Therefore, cross-protective immunity induced by vaccination with gamma-irradiated influenza A virus is mediated mainly by Tc-cell responses.


Assuntos
Proteção Cruzada , Raios gama , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/imunologia , Linfócitos T Citotóxicos/imunologia , Transferência Adotiva , Animais , Anticorpos Antivirais/sangue , Peso Corporal , Vírus da Influenza A Subtipo H1N1/efeitos da radiação , Vírus da Influenza A Subtipo H3N2/efeitos da radiação , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Análise de Sobrevida , Vacinas de Produtos Inativados/imunologia
16.
PLoS One ; 4(10): e7512, 2009 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-19838298

RESUMO

BACKGROUND: Ectromelia virus is a natural mouse pathogen, causing mousepox. The cytotoxic T (Tc) cell granule serine-protease, granzyme B, is important for its control, but the underlying mechanism is unknown. Using ex vivo virus immune Tc cells, we have previously shown that granzyme B is able to activate several independent pro-apoptotic pathways, including those mediated by Bid/Bak/Bax and caspases-3/-7, in target cells pulsed with Tc cell determinants. METHODS AND FINDINGS: Here we analysed the physiological relevance of those pro-apoptotic pathways in ectromelia infection, by incubating ectromelia-immune ex vivo Tc cells from granzyme A deficient (GzmB(+) Tc cells) or granzyme A and granzyme B deficient (GzmAxB(-/-) Tc cell) mice with ectromelia-infected target cells. We found that gzmB-induced apoptosis was totally blocked in ectromelia infected or peptide pulsed cells lacking caspases-3/-7. However ectromelia inhibited only partially apoptosis in cells deficient for Bid/Bak/Bax and not at all when both pathways were operative suggesting that the virus is able to interfere with apoptosis induced by gzmB in case not all pathways are activated. Importantly, inhibition of viral replication in vitro, as seen with wild type cells, was not affected by the lack of Bid/Bak/Bax but was significantly reduced in caspase-3/-7-deficient cells. Both caspase dependent processes were strictly dependent on gzmB, since Tc cells, lacking both gzms, neither induced apoptosis nor reduced viral titers. SIGNIFICANCE: Out findings present the first evidence on the biological importance of the independent gzmB-inducible pro-apoptotic pathways in a physiological relevant virus infection model.


Assuntos
Caspases/metabolismo , Vírus da Ectromelia/genética , Ectromelia Infecciosa/virologia , Fibroblastos/metabolismo , Granzimas/metabolismo , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/virologia , Animais , Apoptose , Calreticulina/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína X Associada a bcl-2/metabolismo
17.
J Immunol ; 183(1): 37-40, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19525394

RESUMO

Cytotoxic T (Tc) cells lyse target cells via exocytosis of granules containing perforin (perf) and granzymes (gzm). In vitro, gzm delivery into the target cell cytosol results in apoptosis, and in the absence of gzm A and B the induction of apoptosis is severely impaired. However, using in vivo Tc cell killing assays, we find that virus-immune, gzm A x B-deficient (gzmAxB(-/-)) mice are competent to eliminate adoptively transferred target cells pulsed with an immunodominant Tc cell determinant as rapidly and completely as their wild-type counterparts. Specific target cell elimination occurred with similar kinetics in both spleen and lymph nodes. Thus, neither gzmA nor gzmB are required for rapid and efficient in vivo cytotoxicity by Tc cells.


Assuntos
Citotoxicidade Imunológica , Granzimas/fisiologia , Linfócitos T Citotóxicos/imunologia , Animais , Morte Celular/genética , Morte Celular/imunologia , Citotoxicidade Imunológica/genética , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/enzimologia , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/patologia , Granzimas/deficiência , Granzimas/genética , Vírus da Influenza A/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/enzimologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Linfócitos T Citotóxicos/patologia , Linfócitos T Citotóxicos/virologia , Fatores de Tempo
18.
PLoS One ; 4(4): e5336, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19401775

RESUMO

BACKGROUND: Influenza A (flu) virus causes significant morbidity and mortality worldwide, and current vaccines require annual updating to protect against the rapidly arising antigenic variations due to antigenic shift and drift. In fact, current subunit or split flu vaccines rely exclusively on antibody responses for protection and do not induce cytotoxic T (Tc) cell responses, which are broadly cross-reactive between virus strains. We have previously reported that gamma-ray inactivated flu virus can induce cross-reactive Tc cell responses. METHODOLOGY/PRINCIPAL FINDING: Here, we report that intranasal administration of purified gamma-ray inactivated human influenza A virus preparations (gamma-Flu) effectively induces heterotypic and cross-protective immunity. A single intranasal administration of gamma-A/PR8[H1N1] protects mice against lethal H5N1 and other heterotypic infections. CONCLUSIONS/SIGNIFICANCE: Intranasal gamma-Flu represents a unique approach for a cross-protective vaccine against both seasonal as well as possible future pandemic influenza A virus infections.


Assuntos
Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/patogenicidade , Vacinas contra Influenza/administração & dosagem , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Administração Intranasal , Animais , Reações Cruzadas , Feminino , Raios gama , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Virus da Influenza A Subtipo H5N1/genética , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , RNA Viral/análise , RNA Viral/genética , Linfócitos T Citotóxicos/imunologia , Vacinação/métodos , Vacinas de Produtos Inativados/administração & dosagem
19.
Immunol Cell Biol ; 86(3): 239-45, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18180794

RESUMO

The mammalian host responds to a microbial infection with a rapid innate immune reaction that is dominated by type I interferon (IFN-I) release. Most cells of vertebrates can respond to microbial attack with IFN-I production, but the cell type responsible for most of the systemic IFN-I release is thought to be plasmacytoid dendritic cells (pDCs). Besides its anti-microbial and especially anti-viral properties IFN-I also exerts a regulatory role on many facets of the sequential adaptive immune response. One of these is being the recently described partial, systemic activation of the vast majority of B and T lymphocytes in mice, irrespective of antigen reactivity. The biological significance of this partial activation of lymphocytes is at present speculative. Secondary infections occurring within a short time span of a primary infection fail to elicit a similar lymphocyte activation response due to a refractory period in systemic IFN-I production. This period of exhaustion in IFN-I responses is associated with an increased susceptibility of the host to secondary infections. The latter correlates with well-established clinical observations of heightened susceptibility of patients to secondary microbial infections after viral episodes.


Assuntos
Memória Imunológica , Infecções/fisiopatologia , Interferon Tipo I , Animais , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Suscetibilidade a Doenças , Humanos , Imunidade Ativa , Imunidade Inata , Infecções/imunologia , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Ativação Linfocitária
20.
J Immunol ; 177(5): 3235-41, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16920963

RESUMO

Viral infections often cause a period of heightened susceptibility to a secondary infection but the cause of this phenomenon is unknown. We found that a primary viral infection in mice rapidly triggers an IFN-I-dependent partial activation state in the majority of B and T lymphocytes, which reverts to a resting phenotype within 5 days. When a secondary infection with an unrelated virus occurred 5 to 9 days after the primary infection, no recurrence of marked activation of lymphocytes was observed. This was not due to an inherent inability of the previously activated cells to undergo renewed partial activation, because they responded when challenged with virus after transfer into "naive" recipients. Instead, the failure to respond optimally resided in the original host's incapacity to mount an IFN-I response to the secondary infection during this time period. Thus, transient immunosuppression through exhaustion of IFN-I production during an acute viral infection creates a time period of enhanced susceptibility to secondary infection.


Assuntos
Interferon Tipo I/biossíntese , Viroses/imunologia , Viroses/metabolismo , Doença Aguda , Adenoviridae/fisiologia , Infecções por Adenoviridae/imunologia , Infecções por Adenoviridae/virologia , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/metabolismo , Infecções por Alphavirus/virologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Biomarcadores , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Células Dendríticas/citologia , Células Dendríticas/imunologia , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/virologia , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Camundongos , Vírus da Floresta de Semliki/fisiologia , Linfócitos T/citologia , Linfócitos T/imunologia , Viroses/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...