Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 12: 694588, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34489940

RESUMO

Reactivation of latent cytomegalovirus (CMV) endangers the therapeutic success of hematopoietic cell transplantation (HCT) in tumor patients due to cytopathogenic virus spread that leads to organ manifestations of CMV disease, to interstitial pneumonia in particular. In cases of virus variants that are refractory to standard antiviral pharmacotherapy, immunotherapy by adoptive cell transfer (ACT) of virus-specific CD8+ T cells is the last resort to bridge the "protection gap" between hematoablative conditioning for HCT and endogenous reconstitution of antiviral immunity. We have used the well-established mouse model of CD8+ T-cell immunotherapy by ACT in a setting of experimental HCT and murine CMV (mCMV) infection to pursue the concept of improving the efficacy of ACT by therapeutic vaccination (TherVac) post-HCT. TherVac aims at restimulation and expansion of limited numbers of transferred antiviral CD8+ T cells within the recipient. Syngeneic HCT was performed with C57BL/6 mice as donors and recipients. Recipients were infected with recombinant mCMV (mCMV-SIINFEKL) that expresses antigenic peptide SIINFEKL presented to CD8+ T cells by the MHC class-I molecule Kb. ACT was performed with transgenic OT-I CD8+ T cells expressing a T-cell receptor specific for SIINFEKL-Kb. Recombinant human CMV dense bodies (DB-SIINFEKL), engineered to contain SIINFEKL within tegument protein pUL83/pp65, served for vaccination. DBs were chosen as they represent non-infectious, enveloped, and thus fusion-competent subviral particles capable of activating dendritic cells and delivering antigens directly into the cytosol for processing and presentation in the MHC class-I pathway. One set of our experiments documents the power of vaccination with DBs in protecting the immunocompetent host against a challenge infection. A further set of experiments revealed a significant improvement of antiviral control in HCT recipients by combining ACT with TherVac. In both settings, the benefit from vaccination with DBs proved to be strictly epitope-specific. The capacity to protect was lost when DBs included the peptide sequence SIINFEKA lacking immunogenicity and antigenicity due to C-terminal residue point mutation L8A, which prevents efficient proteasomal peptide processing and binding to Kb. Our preclinical research data thus provide an argument for using pre-emptive TherVac to enhance antiviral protection by ACT in HCT recipients with diagnosed CMV reactivation.


Assuntos
Transferência Adotiva , Linfócitos T CD8-Positivos/transplante , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/administração & dosagem , Citomegalovirus/patogenicidade , Transplante de Células-Tronco Hematopoéticas , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Proliferação de Células , Células Cultivadas , Citomegalovirus/imunologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Vacinas contra Citomegalovirus/imunologia , Modelos Animais de Doenças , Feminino , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Interações Hospedeiro-Patógeno , Hospedeiro Imunocomprometido , Ativação Linfocitária , Camundongos Endogâmicos C57BL , Vacinação , Ativação Viral
2.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31189713

RESUMO

The development of a vaccine against human cytomegalovirus infection (HCMV) is a high-priority medical goal. The viral pentameric protein complex consisting of glycoprotein H (gH)/gL/UL128-131A (PC) is considered to be an important vaccine component. Its relevance to the induction of a protective antibody response is, however, still a matter of debate. We addressed this issue by using subviral dense bodies (DBs) of HCMV. DBs are exceptionally immunogenic. Laboratory HCMV strain DBs harbor important neutralizing antibody targets, like the glycoproteins B, H, L, M, and N, but they are devoid of the PC. To be able to directly compare the impact of the PC on the levels of neutralizing antibody (NT-abs) responses, a PC-positive variant of the HCMV laboratory strain Towne was established by bacterial artificial chromosome (BAC) mutagenesis (Towne-UL130rep). This strain synthesized PC-positive DBs upon infection of fibroblasts. These DBs were used in side-by-side immunizations with PC-negative Towne DBs. Mouse and rabbit sera were tested to address the impact of the PC on DB immunogenicity. The neutralizing antibody response to PC-positive DBs was superior to that of PC-negative DBs, as tested on fibroblasts, epithelial cells, and endothelial cells and for both animal species used. The experiments revealed the potential of the PC to enhance the antibody response against HCMV. Of particular interest was the finding that PC-positive DBs induced an antibody response that blocked the infection of fibroblasts by a PC-positive viral strain more efficiently than sera following immunizations with PC-negative particles.IMPORTANCE Infections with the human cytomegalovirus (HCMV) may cause severe and even life-threatening disease manifestations in newborns and immunosuppressed individuals. Several strategies for the development of a vaccine against this virus are currently pursued. A critical question in this respect refers to the antigenic composition of a successful vaccine. Using a subviral particle vaccine candidate, we show here that one protein complex of HCMV, termed the pentameric complex (PC), enhances the neutralizing antibody response against viral infection of different cell types. We further show for the first time that this not only relates to the infection of epithelial or endothelial cells; the presence of the PC in the particles also enhanced the neutralizing antibody response against the infection of fibroblasts by HCMV. Together, these findings argue in favor of including the PC in strategies for HCMV vaccine development.


Assuntos
Anticorpos Neutralizantes/metabolismo , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Células Cultivadas , Vacinas contra Citomegalovirus/imunologia , Prepúcio do Pênis/citologia , Prepúcio do Pênis/virologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Glicoproteínas de Membrana/imunologia , Camundongos , Complexos Multiproteicos/imunologia , Coelhos
3.
J Biol Chem ; 294(15): 6188-6203, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30782840

RESUMO

Human cytomegalovirus (HCMV) is a common ß-herpesvirus causing life-long latent infections. HCMV replication interferes with cell cycle regulation in host cells because the HCMV-encoded cyclin-dependent kinase (CDK) ortholog pUL97 extensively phosphorylates the checkpoint regulator retinoblastoma protein. pUL97 also interacts with cyclins B1, T1, and H, and recent findings have strongly suggested that these interactions influence pUL97 substrate recognition. Interestingly, here we detected profound mechanistic differences among these pUL97-cyclin interactions. Our study revealed the following. (i) pUL97 interacts with cyclins B1 and H in a manner dependent on pUL97 activity and HCMV-specific cyclin modulation, respectively. (ii) The phosphorylated state of both proteins is an important determinant of the pUL97-cyclin B1 interaction. (iii) Activated phospho-Thr-315 cyclin H is up-regulated during HCMV replication. (iv) Thr-315 phosphorylation is independent of intracellular pUL97 or CDK7 activity. (v) pUL97-mediated in vitro phosphorylation is detectable for cyclin B1 but not H. (vi) Mutual transphosphorylation between pUL97 and CDK7 is not detectable, and an MS-based phosphosite analysis indicated that pUL97 might unexpectedly not be phosphorylated in its T-loop. (vii) The binary complexes pUL97-cyclin H and CDK7-cyclin H as well as the ternary complex pUL97-cyclin-H-CDK7 are detectable in an assembly-based CoIP approach. (viii) pUL97 self-interaction can be bridged by the transcriptional cyclins T1 or H but not by the classical cell cycle-regulating B1 cyclin. Combined, our findings unravel a number of cyclin type-specific differences in pUL97 interactions and suggest a multifaceted regulatory impact of cyclins on HCMV replication.


Assuntos
Ciclina B1/metabolismo , Ciclina H/metabolismo , Ciclina T/metabolismo , Citomegalovirus/fisiologia , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Ciclina B1/genética , Ciclina H/genética , Ciclina T/genética , Células HEK293 , Humanos , Fosforilação , Domínios Proteicos , Estrutura Quaternária de Proteína , Proteínas Virais/genética
4.
Viruses ; 8(2)2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26848680

RESUMO

The human cytomegalovirus (HCMV) replicates to high titers in primary human fibroblast cell cultures. A variety of primary human cells and some tumor-derived cell lines do also support permissive HCMV replication, yet at low levels. Cell lines established by transfection of the transforming functions of adenoviruses have been notoriously resistant to HCMV replication and progeny production. Here, we provide first-time evidence that a permanent cell line immortalized by adenovirus type 5 E1A and E1B (CAP) is supporting the full HCMV replication cycle and is releasing infectious progeny. The CAP cell line had previously been established from amniotic fluid cells which were likely derived from membranes of the developing fetus. These cells can be grown under serum-free conditions. HCMV efficiently penetrated CAP cells, expressed its immediate-early proteins and dispersed restrictive PML-bodies. Viral DNA replication was initiated and viral progeny became detectable by electron microscopy in CAP cells. Furthermore, infectious virus was released from CAP cells, yet to lower levels compared to fibroblasts. Subviral dense bodies were also secreted from CAP cells. The results show that E1A/E1B expression in transformed cells is not generally repressive to HCMV replication and that CAP cells may be a good substrate for dense body based vaccine production.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Proteínas E1B de Adenovirus/metabolismo , Infecções por Adenovirus Humanos/virologia , Adenovírus Humanos/metabolismo , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Replicação Viral , Proteínas E1A de Adenovirus/genética , Proteínas E1B de Adenovirus/genética , Adenovírus Humanos/genética , Transformação Celular Viral , Citomegalovirus/genética , Humanos
5.
PLoS Pathog ; 11(7): e1005049, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26181057

RESUMO

Reactivation of human cytomegalovirus (HCMV) can cause severe disease in recipients of hematopoietic stem cell transplantation. Although preclinical research in murine models as well as clinical trials have provided 'proof of concept' for infection control by pre-emptive CD8 T-cell immunotherapy, there exists no predictive model to experimentally evaluate parameters that determine antiviral efficacy of human T cells in terms of virus control in functional organs, prevention of organ disease, and host survival benefit. We here introduce a novel mouse model for testing HCMV epitope-specific human T cells. The HCMV UL83/pp65-derived NLV-peptide was presented by transgenic HLA-A2.1 in the context of a lethal infection of NOD/SCID/IL-2rg-/- mice with a chimeric murine CMV, mCMV-NLV. Scenarios of HCMV-seropositive and -seronegative human T-cell donors were modeled by testing peptide-restimulated and T-cell receptor-transduced human T cells, respectively. Upon transfer, the T cells infiltrated host tissues in an epitope-specific manner, confining the infection to nodular inflammatory foci. This resulted in a significant reduction of viral load, diminished organ pathology, and prolonged survival. The model has thus proven its potential for a preclinical testing of the protective antiviral efficacy of HCMV epitope-specific human T cells in the evaluation of new approaches to an immunotherapy of CMV disease.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Infecções por Citomegalovirus/terapia , Citomegalovirus/imunologia , Epitopos de Linfócito T/imunologia , Carga Viral/imunologia , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Modelos Animais de Doenças , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Proteínas da Matriz Viral/imunologia
6.
PLoS Pathog ; 9(5): e1003383, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23717207

RESUMO

Control of human cytomegalovirus (HCMV) depends on CD8+ T cell responses that are shaped by an individual's repertoire of MHC molecules. MHC class I presentation is modulated by a set of HCMV-encoded proteins. Here we show that HCMV immunoevasins differentially impair T cell recognition of epitopes from the same viral antigen, immediate-early 1 (IE-1), that are presented by different MHC class I allotypes. In the presence of immunoevasins, HLA-A- and HLA-B-restricted T cell clones were ineffective, but HLA-C*0702-restricted T cell clones recognized and killed infected cells. Resistance of HLA-C*0702 to viral immunoevasins US2 and US11 was mediated by the alpha3 domain and C-terminal region of the HLA heavy chain. In healthy donors, HLA-C*0702-restricted T cells dominated the T cell response to IE-1. The same HLA-C allotype specifically protected infected cells from attack by NK cells that expressed a corresponding HLA-C-specific KIR. Thus, allotype-specific viral immunoevasion allows HCMV to escape control by NK cells and HLA-A- and HLA-B-restricted T cells, while the virus becomes selectively vulnerable to an immunodominant population of HLA-C-restricted T cells. Our work identifies a T cell population that may be of particular efficiency in HCMV-specific immunotherapy.


Assuntos
Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Epitopos de Linfócito T/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Evasão da Resposta Imune , Linfócitos T CD8-Positivos/patologia , Infecções por Citomegalovirus/patologia , Feminino , Humanos , Proteínas Imediatamente Precoces/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Masculino , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas Virais/imunologia
7.
J Virol ; 87(9): 5229-38, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23449799

RESUMO

Suppression of major histocompatibility complex (MHC) class I-mediated presentation of human cytomegalovirus (HCMV) peptides is an important mechanism to avoid CD8 T lymphocyte recognition and killing of infected cells. Of particular interest is how MHC class I presentation of essential regulatory immediate early (IE) proteins of HCMV can be effectively compromised at times when known viral immunoevasins are not abundantly expressed. The tegument protein pp71 had been suggested to be involved in MHC class I downregulation. Intriguingly, this polypeptide is also critically engaged in the initial derepression of the major IE gene locus, leading to enhanced expression of IE proteins IE1-pp72 and IE2-pp86. Using a set of viral mutants, we addressed the role of pp71 in MHC class I presentation of IE1-pp72-derived peptides. We show that the amount of "incoming" pp71 positively correlates with IE1-pp72 protein levels and with the presentation of IE1-derived peptides. This indicates that the amount of the IE1 protein, induced by pp71, rather than a putative immunoevasive function of the tegument protein, determines MHC class I antigen presentation of IE1-derived peptides. This process proved to be independent of the presence of pp65, which had been reported to interfere with IE1 presentation. It may thus be beneficial for the success of HCMV replication to limit the level of pp71 delivered from infecting particles in order to avoid critical levels of MHC class I presentation of IE protein-derived peptides.


Assuntos
Apresentação de Antígeno , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Proteínas Imediatamente Precoces/imunologia , Proteínas Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Citomegalovirus/genética , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Proteínas Imediatamente Precoces/genética , Peptídeos/genética , Peptídeos/imunologia , Regulação para Cima , Proteínas Virais/genética
8.
J Gen Virol ; 94(Pt 2): 376-386, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23100361

RESUMO

Human cytomegalovirus (HCMV) interferes with MHC class I-restricted antigen presentation and thereby reduces recognition by CD8(+) T-cells. This interference is mediated primarily by endoplasmic reticulum-resident glycoproteins that are encoded in the US2-11 region of the viral genome. Such a suppression of recognition would be of particular importance immediately after infection, because several immunodominant viral antigens are already present in the cell in this phase. However, which of the evasion proteins gpUS2-11 interfere(s) with antigen presentation to CD8(+) T-cells at this time of infection is not known. Here we address this question, using recombinant viruses (RV) that express only one of the immunoevasins gpUS2, gpUS3 or gpUS11. Infection with RV-US3 had only a limited impact on the presentation of peptides from the CD8(+) T-cell antigens IE1 and pp65 under immediate-early (IE) conditions imposed by cycloheximide/actinomycin D blocking. Unexpectedly, both RV-US2 and RV-US11 considerably impaired the recognition of IE1 and pp65 by CD8(+) T-cells, and both US2 and, to a lesser extent, US11 were transcribed under IE conditions. Thus, gpUS2 and gpUS11 are key effectors of MHC class I immunoevasion immediately after HCMV infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citomegalovirus/imunologia , Citomegalovirus/patogenicidade , Evasão da Resposta Imune , Tolerância Imunológica , Proteínas de Ligação a RNA/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas Virais/imunologia , Linhagem Celular , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Fatores de Virulência/imunologia
9.
Eur J Immunol ; 42(12): 3442-53, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22930221

RESUMO

Current protocols used to select CMV-specific T cells for adoptive immunotherapy focus on virus-specific memory T cells from seropositive donors. However, this strategy is not feasible in patients undergoing allogeneic haematopoietic stem-cell transplantation (HSCT) from CMV-seronegative donors. Here, we redirected T cells of CMV-seronegative donors with a human genetically engineered TCR recognizing an HLA-A*0201-binding peptide epitope of CMVpp65. To facilitate clinical translation of this approach, we used a non-viral expression system based on in vitro transcribed RNA and electroporation. Although memory and naïve-derived T-cell subsets were both efficiently transfected by TCR-RNA, memory-derived T cells showed much stronger levels of HLA-A*0201-restricted cytolytic activity to CMV-infected fibroblasts and maintained acquired function for 5-10 days. In addition to redirection of CD8(+) cytotoxic T cells, TCR-RNA transfection was capable of redirecting CD4(+) T cells into potent Ag-specific Th cells that efficiently triggered maturation of DCs. Our data suggest that memory rather than naïve-derived T cells are the preferred subset for transient TCR expression by RNA electroporation, providing more efficient and sustained virus-specific CD4(+) and CD8(+) T-cell function. CMV TCR-RNA may represent a suitable therapeutic 'off-the-shelf' reagent to be used in severe CMV infections of HSCT patients when endogenous CMV-specific T-cell immunity is insufficient.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Terapia Baseada em Transplante de Células e Tecidos , Citomegalovirus/imunologia , Memória Imunológica/efeitos dos fármacos , Fosfoproteínas/imunologia , RNA/farmacologia , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas da Matriz Viral/imunologia , Citomegalovirus/genética , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/terapia , Eletroporação/métodos , Fibroblastos/imunologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/terapia , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Memória Imunológica/genética , Fosfoproteínas/genética , RNA/genética , RNA/imunologia , Receptores de Antígenos de Linfócitos T/genética , Transfecção/métodos , Transplante Homólogo , Proteínas da Matriz Viral/genética
10.
Antimicrob Agents Chemother ; 56(1): 75-82, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21968369

RESUMO

Polyethylenimines are cationic polymers with potential as delivery vectors in gene therapy and with proven antimicrobial activity. However, the antiviral activity of polyethylenimines has not been addressed in detail thus far. We have studied the inhibitory effects of a linear 25-kDa polyethylenimine on infections with human papillomaviruses and human cytomegaloviruses. Preincubation of cells with polyethylenimine blocked primary attachment of both viruses to cells, resulting in a significant reduction of infection. In addition, the dissemination of human cytomegalovirus in culture cells was efficiently reduced by recurrent administration of polyethylenimine. Polyethylenimine concentrations required for inhibition of human papillomavirus and cytomegalovirus did not cause any cytotoxic effects. Polyethylenimines and their derivatives may thus be attractive molecules for the development of antiviral microbicides.


Assuntos
Antivirais/farmacologia , Infecções por Citomegalovirus , Citomegalovirus/efeitos dos fármacos , Papillomaviridae/efeitos dos fármacos , Polietilenoimina/farmacologia , Ligação Viral/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Células COS , Cátions , Chlorocebus aethiops , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/prevenção & controle , Infecções por Citomegalovirus/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Células HEK293 , Células HeLa , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/virologia , Microscopia de Fluorescência , Especificidade de Órgãos , Papillomaviridae/fisiologia , Infecções por Papillomavirus/tratamento farmacológico , Infecções por Papillomavirus/prevenção & controle , Infecções por Papillomavirus/virologia , Polietilenoimina/uso terapêutico
11.
Haematologica ; 97(6): 874-82, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22207687

RESUMO

BACKGROUND: After allogeneic hematopoietic stem-cell transplantation patients are at increased risk for herpes zoster as long as varicella-zoster virus specific T-cell reconstitution is impaired. This study aimed to identify immunodominant varicella-zoster virus antigens that drive recovery of virus-specific T cells after transplantation. DESIGN AND METHODS: Antigens were purified from a varicella-zoster virus infected cell lysate by high-performance liquid chromatography and were identified by quantitative mass spectrometric analysis. To approximate in vivo immunogenicity for memory T cells, antigen preparations were consistently screened with ex vivo PBMC of varicella-zoster virus immune healthy individuals in sensitive interferon-γ ELISpot assays. Candidate virus antigens identified by the approach were genetically expressed in PBMC using electroporation of in vitro transcribed RNA encoding full-length proteins and were then analyzed for recognition by CD4(+) and CD8(+) memory T cells. RESULTS: Varicella-zoster virus encoded glycoproteins B and E, and immediate early protein 62 were identified in immunoreactive lysate material. Predominant CD4(+) T-cell reactivity to these proteins was observed in healthy virus carriers. Furthermore, longitudinal screening in allogeneic stem-cell transplantation patients showed strong expansions of memory T cells recognizing glycoproteins B and E after onset of herpes zoster, while immediate early protein 62 reactivity remained moderate. Reactivity to viral glycoproteins boosted by acute zoster was mediated by both CD4(+) and CD8(+) T cells. CONCLUSIONS: Our data demonstrate that glycoproteins B and E are major targets of varicella-zoster virus specific CD4(+) and CD8(+) T-cell reconstitution occurring during herpes zoster after allogeneic stem-cell transplantation. Varicella-zoster virus glycoproteins B and E might form the basis for novel non-hazardous zoster subunit vaccines suitable for immunocompromised transplant patients.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Transplante de Células-Tronco Hematopoéticas , Herpesvirus Humano 3/imunologia , Condicionamento Pré-Transplante/métodos , Proteínas do Envelope Viral/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Eletroporação , Feminino , Herpes Zoster/imunologia , Herpes Zoster/virologia , Herpesvirus Humano 3/química , Humanos , Masculino , Plasmídeos , Espectrometria de Massas por Ionização por Electrospray , Transfecção , Transplante Homólogo , Proteínas do Envelope Viral/química
12.
Expert Rev Vaccines ; 8(8): 999-1021, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19627184

RESUMO

Impairment of cellular immunity upon hematopoietic stem cell transplantation (HSCT) may lead to serious clinical manifestations induced by human cytomegalovirus (HCMV) and varicella-zoster virus (VZV) infections. Although the clinical presentations, preferential organ involvement and clinical courses are different, infections with both herpesviruses are similar with respect to many pathophysiological aspects and the therapeutic strategies that are employed to combat them. Antiviral drug prophylaxis and therapy are successfully used to limit the risk of reactivated HCMV and VZV infections, but are unable to absolutely prevent episodes of virus disease in long-term follow-up after HSCT. Control of infection requires the re-establishment of protective antiviral cellular immunity in the host. Here we review the most recent developments in the field of HCMV and VZV immunotherapy with specific emphasis on the question of how vaccines against HCMV and VZV may aid in enhancing the reconstitution of antiviral immunity after HSCT, and thereby support the control of these two agents by transplant recipients.


Assuntos
Vacina contra Varicela/imunologia , Varicela/prevenção & controle , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Hospedeiro Imunocomprometido
13.
Biol Blood Marrow Transplant ; 14(12): 1417-24, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19041065

RESUMO

Reactivated varicella-zoster virus (VZV) infection causes herpes zoster and commonly occurs after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Because VZV-specific T cell immunity is essential to prevent virus reactivation, we developed an interferon-gamma enzyme-linked immunosorbent spot (ELISPOT) assay for the sensitive detection of VZV-reactive T cells at the single-cell level ex vivo. We used this assay to monitor the frequency of VZV-reactive T cells in 17 seropositive patients during the first year after T cell-depleted allo-HSCT. The patients did not receive anti-herpesvirus prophylaxis after stem cell engraftment. Independent of the magnitude of transferred donor immunity, VZV-reactive T cell numbers decreased to low levels (median, 2/mL; range, 0 to 35/mL) in peripheral blood early after transplantation. Only patients with subsequent zoster (n = 5) exhibited a dramatic boost in VZV-reactive T cells (median, 366/mL; range, 158 to 756/mL), which was induced by the reactivation event. The postzoster VZV-reactive T cell levels were similar to those seen in healthy virus carriers. In contrast, antiviral T cell levels remained low in patients without VZV disease. Our results demonstrate that VZV-specific T cell immunity recovered efficiently during zoster in T cell-depleted allo-HSCT recipients. It did not reconstitute spontaneously in nonzoster patients, even in the absence of antiviral prophylaxis. Prospective studies should investigate whether VZV vaccination can substitute for natural resensitization by virus disease.


Assuntos
Herpes Zoster/imunologia , Herpesvirus Humano 3/imunologia , Recuperação de Função Fisiológica/imunologia , Linfócitos T/imunologia , Ativação Viral/imunologia , Feminino , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/terapia , Neoplasias Hematológicas/virologia , Transplante de Células-Tronco Hematopoéticas , Herpes Zoster/prevenção & controle , Humanos , Imunidade Celular/imunologia , Depleção Linfocítica , Masculino , Linfócitos T/virologia , Transplante Homólogo , Vacinação , Vacinas Virais/imunologia , Vacinas Virais/farmacologia
14.
Nat Immunol ; 6(5): 515-23, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15821739

RESUMO

Human cytomegalovirus, a chief pathogen in immunocompromised people, can persist in a healthy immunocompetent host throughout life without being eliminated by the immune system. Here we show that pp65, the main tegument protein of human cytomegalovirus, inhibited natural killer cell cytotoxicity by an interaction with the activating receptor NKp30. This interaction was direct and specific, leading to dissociation of the linked CD3zeta from NKp30 and, consequently, to reduced killing. Thus, pp65 is a ligand for the NKp30 receptor and demonstrates a unique mechanism by which an intracellular viral protein causes general suppression of natural killer cell cytotoxicity by specific interaction with an activating receptor.


Assuntos
Citomegalovirus/fisiologia , Células Matadoras Naturais/imunologia , Glicoproteínas de Membrana/antagonistas & inibidores , Fosfoproteínas/metabolismo , Receptores Imunológicos/antagonistas & inibidores , Proteínas da Matriz Viral/metabolismo , Animais , Células Cultivadas , Citotoxicidade Imunológica/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Cinética , Glicoproteínas de Membrana/metabolismo , Camundongos , Receptor 3 Desencadeador da Citotoxicidade Natural , Fosfoproteínas/farmacologia , Ligação Proteica , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/metabolismo , Proteínas da Matriz Viral/farmacologia
15.
Blood ; 101(12): 4870-7, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12609847

RESUMO

Human cytomegalovirus (HCMV) has developed multiple strategies to escape immune recognition. Here, we demonstrate that HCMV down-regulates HLA-DR expression in infected interferon gamma (IFN-gamma)-stimulated fibroblasts at 1 day after infection. Decreased HLA-DR expression was not observed on cells infected with an HCMV strain lacking the pp65 gene (RVAD65), but was observed on cells transfected with the pp65 gene. HLA-DR expression accumulated in vacuoles near the nucleus in HCMV-infected, but not in uninfected or RVAD65-infected cells. In addition, the HLA-DR alpha-chain, but not the beta-chain or HLA-DM, was degraded in HCMV-infected but not in RVAD65-infected cells. Thus, the HCMV protein pp65 mediates decreased expression of HLA-DR, by mediating an accumulation of HLA class II molecules in lysosomes that results in degradation of the HLA-DR alpha-chain.


Assuntos
Citomegalovirus/imunologia , Antígenos HLA-DR/metabolismo , Lisossomos/metabolismo , Fosfoproteínas/fisiologia , Proteínas Proto-Oncogênicas , Proteínas da Matriz Viral/fisiologia , Transporte Biológico , Citomegalovirus/química , Citomegalovirus/genética , Células Dendríticas/imunologia , Fibroblastos/ultraestrutura , Fibroblastos/virologia , Citometria de Fluxo , Deleção de Genes , Expressão Gênica , Antígenos HLA-D/metabolismo , Antígenos HLA-DR/genética , Humanos , Fator Gênico 3 Estimulado por Interferon , Interferon gama/farmacologia , Janus Quinase 1 , Janus Quinase 2 , Leucócitos Mononucleares/imunologia , Microscopia Confocal , Fosfoproteínas/genética , Proteínas Tirosina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transfecção , Proteínas da Matriz Viral/genética
16.
J Clin Virol ; 25 Suppl 2: S75-85, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12361759

RESUMO

BACKGROUND: Pre- and perinatal human cytomegalovirus (HCMV) infection remains one of the major causes of mental defects and sensineural hearing loss in children. In addition, it is a prominent infectious complication in immunosuppressed individuals such as AIDS patients or transplant recipients. Therefore, the development of an HCMV vaccine has been given top priority by health care institutions. STUDY DESIGN: Defective subviral particles of HCMV, termed Dense Bodies (DB) contain the dominant target antigens for humoral and cellular immune responses elicited during natural infection. These enveloped particles are released from infected culture cells and can be purified by gradient centrifugation. DB were analyzed for their ability to induce virus neutralizing antibodies and cytotoxic T cells (CTL) after immunization of mice. RESULTS: Purified DB entered human and murine hematopoetic and fibroblast cells very efficiently, thereby delivering their protein content into the cytoplasm. The cellular uptake was abrogated after sonication and freeze-thawing of the particles, indicating that the integrity of the viral envelope was important for this process. DB immunization of HLA-A2.K(b) transgenic mice induced significant CTL responses in the absence of viral gene expression and without the use of adjuvant. Induction of cytolytic cells by DB was sensitive to sonication and freeze-thawing as determined by CD3epsilon -redirected lysis analysis. In accordance with that, induction of virus neutralizing antibodies was much more effective when untreated DB were used as immunogen. CONCLUSIONS: DB provide a promising basis for the development of a subunit vaccine against HCMV infection. The ability to genetically engineer HCMV provides a rationale to optimize such a vaccine and to develop concepts for future multicomponent vaccines.


Assuntos
Vacinas contra Citomegalovirus/imunologia , Citomegalovirus/imunologia , Vírion/imunologia , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Infecções por Citomegalovirus/prevenção & controle , Citotoxicidade Imunológica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Testes de Neutralização , Linfócitos T Citotóxicos/imunologia , Vacinas de Produtos Inativados/imunologia
17.
Virology ; 295(2): 208-16, 2002 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-12033779

RESUMO

Control of human cytomegalovirus (HCMV) infection is predominantly mediated by cytolytic CD8+ T lymphocytes (CTL). Among the roughly 200 HCMV-encoded polypeptides, the tegument protein pp65 (ppUL83) and the nonstructural IE1 protein are considered to be dominant CTL targets. Yet the importance of CTL against IE1 for protective immunity against HCMV reactivation and disease has remained elusive. Analyses have been difficult, as all MHC class I presented peptides of IE1 defined so far are located in parts of the protein that are variable between viral strains. In this study a conserved decameric peptide from IE1 (P6, IE1(354-363)) that bound to HLA-A2 was identified. Using peptide-pulsed, HLA-matched stimulator cells, CTL lines which recognized P6 after exogenous loading as well as after endogenous processing could repeatedly be generated. However, memory CTL directed against P6 were not readily detectable by ex vivo ELISPOT analysis in peripheral blood mononuclear cells of healthy seropositive individuals, indicating that this peptide represents a quantitatively subdominant determinant during latent HCMV infection. Using the conserved HLA-A2 presented peptide P6 will enable more detailed studies on the role of IE1-specific CTL in patients suffering from various HCMV-related disease conditions and investigation of the role of such cells for immune control of HCMV. Since IE1 is the first viral protein to be expressed after reactivation from latency, P6 may also serve as an important component of a future recombinant HCMV vaccine.


Assuntos
Sequência Conservada , Citomegalovirus/imunologia , Antígeno HLA-A2/metabolismo , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/imunologia , Fragmentos de Peptídeos/imunologia , Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas Virais , Sequência de Aminoácidos , Linhagem Celular , Infecções por Citomegalovirus/imunologia , Testes Imunológicos de Citotoxicidade , Epitopos de Linfócito T/imunologia , Humanos , Proteínas Imediatamente Precoces/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Latência Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA