Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Exp Med ; 203(8): 2009-19, 2006 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-16880257

RESUMO

Experimental autoimmune myocarditis (EAM) appears after infectious heart disease, the most common cause of dilated cardiomyopathy in humans. Here we report that mice lacking T-bet, a T-box transcription factor required for T helper (Th)1 cell differentiation and interferon (IFN)-gamma production, develop severe autoimmune heart disease compared to T-bet+/+ control mice. Experiments in T-bet-/- IL-4-/- and T-bet-/- IL-4Ralpha-/- mice, as well as transfer of heart-specific Th1 and Th2 cell lines, showed that autoimmune heart disease develops independently of Th1 or Th2 polarization. Analysis of T-bet-/- IL-12Rbeta1-/- and T-bet-/- IL-12p35-/- mice then identified interleukin (IL)-23 as critical for EAM pathogenesis. In addition, T-bet-/- mice showed a marked increase in production of the IL-23-dependent cytokine IL-17 by heart-infiltrating lymphocytes, and in vivo IL-17 depletion markedly reduced EAM severity in T-bet-/- mice. Heart-infiltrating T-bet-/- CD8+ but not CD8- T cells secrete IFN-gamma, which inhibits IL-17 production and protects against severe EAM. In contrast, T-bet-/- CD8+ lymphocytes completely lost their capacity to release IFN-gamma within the heart. Collectively, these data show that severe IL-17-mediated EAM can develop in the absence of T-bet, and that T-bet can regulate autoimmunity via the control of nonspecific CD8+ T cell bystander functions in the inflamed target organ.


Assuntos
Doenças Autoimunes/imunologia , Interleucina-17/biossíntese , Miocardite/imunologia , Fatores de Transcrição/metabolismo , Animais , Doenças Autoimunes/metabolismo , Autoimunidade/imunologia , Medula Óssea , Complexo CD3/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular , Citocinas/biossíntese , Células Dendríticas/citologia , Células Dendríticas/imunologia , Humanos , Imunização , Interleucina-17/deficiência , Interleucina-23 , Subunidade p19 da Interleucina-23 , Interleucinas/biossíntese , Tecido Linfoide/imunologia , Camundongos , Camundongos SCID , Miocardite/metabolismo , Miocárdio/citologia , Miocárdio/patologia , Receptores de Interleucina-2/imunologia , Proteínas com Domínio T , Linfócitos T Reguladores/imunologia , Células Th1/citologia , Células Th1/imunologia , Células Th2/citologia , Células Th2/imunologia , Fatores de Transcrição/deficiência
2.
Circ Res ; 105(9): 912-20, 2009 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-19762681

RESUMO

RATIONALE: The myeloid differentiation factor (MyD)88/interleukin (IL)-1 axis activates self-antigen-presenting cells and promotes autoreactive CD4(+) T-cell expansion in experimental autoimmune myocarditis, a mouse model of inflammatory heart disease. OBJECTIVE: The aim of this study was to determine the role of MyD88 and IL-1 in the progression of acute myocarditis to an end-stage heart failure. METHODS AND RESULTS: Using alpha-myosin heavy chain peptide (MyHC-alpha)-loaded, activated dendritic cells, we induced myocarditis in wild-type and MyD88(-/-) mice with similar distributions of heart-infiltrating cell subsets and comparable CD4(+) T-cell responses. Injection of complete Freund's adjuvant (CFA) or MyHC-alpha/CFA into diseased mice promoted cardiac fibrosis, induced ventricular dilation, and impaired heart function in wild-type but not in MyD88(-/-) mice. Experiments with chimeric mice confirmed the bone marrow origin of the fibroblasts replacing inflammatory infiltrates and showed that MyD88 and IL-1 receptor type I signaling on bone marrow-derived cells was critical for development of cardiac fibrosis during progression to heart failure. CONCLUSIONS: Our findings indicate a critical role of MyD88/IL-1 signaling in the bone marrow compartment in postinflammatory cardiac fibrosis and heart failure and point to novel therapeutic strategies against inflammatory cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/imunologia , Insuficiência Cardíaca/imunologia , Interleucina-1beta/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Miocardite/imunologia , Miocárdio/imunologia , Transdução de Sinais , Animais , Autoimunidade , Transplante de Medula Óssea , Linfócitos T CD4-Positivos/imunologia , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/transplante , Modelos Animais de Doenças , Progressão da Doença , Fibroblastos/imunologia , Fibrose , Adjuvante de Freund , Proteínas de Fluorescência Verde/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Imunidade Inata , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Miocardite/complicações , Miocardite/patologia , Miocardite/fisiopatologia , Miocárdio/patologia , Cadeias Pesadas de Miosina/imunologia , Fenótipo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Quimeras de Transplante
3.
Nucleic Acids Res ; 34(2): e16, 2006 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-16449199

RESUMO

The straightforward production and dose-controlled administration of protein therapeutics remain major challenges for the biopharmaceutical manufacturing and gene therapy communities. Transgenes linked to HIV-1-derived vpr and pol-based protease cleavage (PC) sequences were co-produced as chimeric fusion proteins in a lentivirus production setting, encapsidated and processed to fusion peptide-free native protein in pseudotyped lentivirions for intracellular delivery and therapeutic action in target cells. Devoid of viral genome sequences, protein-transducing nanoparticles (PTNs) enabled transient and dose-dependent delivery of therapeutic proteins at functional quantities into a variety of mammalian cells in the absence of host chromosome modifications. PTNs delivering Manihot esculenta linamarase into rodent or human, tumor cell lines and spheroids mediated hydrolysis of the innocuous natural prodrug linamarin to cyanide and resulted in efficient cell killing. Following linamarin injection into nude mice, linamarase-transducing nanoparticles impacted solid tumor development through the bystander effect of cyanide.


Assuntos
Antineoplásicos , Terapia Genética/métodos , Lentivirus/genética , Neoplasias/terapia , beta-Glucosidase/genética , Animais , Antineoplásicos/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Terapia Combinada , Cricetinae , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Nanoestruturas , Neoplasias/tratamento farmacológico , Nitrilas/uso terapêutico , Pró-Fármacos/uso terapêutico , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Esferoides Celulares , Transfecção , beta-Glucosidase/metabolismo
4.
Circulation ; 113(2): 258-65, 2006 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-16401773

RESUMO

BACKGROUND: Experimental autoimmune myocarditis (EAM) is a CD4+ T-cell-mediated mouse model of postviral cardiomyopathy. Activation of interleukin-1 type 1 and Toll-like receptors that share the common downstream adaptor molecule MyD88 is required for disease induction. The specific role of MyD88 in myocarditis, however, is not known. METHODS AND RESULTS: In contrast to control littermates, MyD88(-/-) mice were protected from myocarditis after immunization with alpha-myosin heavy chain-derived peptide (MyHC-alpha) and complete Freund's adjuvant. Disease resistance of MyD88(-/-) mice resulted from impaired expansion of heart-specific CD4+ T cells after immunization. Intrinsic defects of MyD88(-/-) CD4+ T cells were excluded. In contrast, MyD88(-/-) but not MyD88(+/+) primary antigen presenting dendritic cells (DCs) were defective in their capacity to prime CD4+ T cells. This defect mainly resulted from the inability of MyD88(-/-) DCs to release tumor necrosis factor-alpha. The critical role of MyD88 signaling in DCs in the peripheral lymphatic compartments was finally proven by repetitive injection of activated, MyHC-alpha-loaded MyD88(+/+) DCs that fully restored T-cell expansion and myocarditis in MyD88(-/-) mice. CONCLUSIONS: Autoimmune myocarditis induction depends on MyD88 signaling in self-antigen presenting cells in the peripheral compartments. We conclude that MyD88 might become a target for prevention of heart-specific autoimmunity and cardiomyopathy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Doenças Autoimunes/etiologia , Miocardite/etiologia , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Autoantígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide , Miocardite/imunologia , Cadeias Pesadas de Miosina/administração & dosagem , Cadeias Pesadas de Miosina/imunologia , Miosinas Ventriculares/administração & dosagem , Miosinas Ventriculares/imunologia
5.
Int J Cardiol ; 112(1): 34-9, 2006 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-16891018

RESUMO

Dilated cardiomyopathy is the most common cause of heart failure in young patients and often results from autoimmunity triggered by viral or bacterial infections. Dendritic cells are professional antigen-presenting cells residing in lymphatic tissue and in the heart. They are involved in both, host defense and maintenance of peripheral tolerance. Animal models suggest an important role for dendritic cells in the induction of autoimmune myocarditis. Activated and self-antigen loaded dendritic cells, for example, induce myocarditis and heart failure in susceptible mice. It appears that the combined presence of tissue damage and innate activation exceeding a certain threshold prompts dendritic cells to prime and amplify potentially autoreactive T cells targeting the heart. The concept of dendritic cell induced myocarditis helps us to understand disease pathogenesis and offers a nice approach to develop novel therapeutic strategies against a devastating heart disease.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade , Células Dendríticas/imunologia , Insuficiência Cardíaca/imunologia , Animais , Doenças Autoimunes/complicações , Doenças Autoimunes/tratamento farmacológico , Cardiomiopatia Dilatada/tratamento farmacológico , Cardiomiopatia Dilatada/imunologia , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Modelos Animais de Doenças , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Imunidade Celular/efeitos dos fármacos , Fatores Imunológicos/uso terapêutico , Camundongos , Miocardite/tratamento farmacológico , Miocardite/imunologia
6.
Nucleic Acids Res ; 31(12): e69, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12799458

RESUMO

Advanced heterologous transcription control systems for adjusting desired transgene expression are essential for gene function assignments, drug discovery, manufacturing of difficult to produce protein pharmaceuticals and precise dosing of gene-based therapeutic interventions. Conversion of the Streptomyces albus heat shock response regulator (RheA) into an artificial eukaryotic transcription factor resulted in a vertebrate thermosensor (CTA; cold-inducible transactivator), which is able to adjust transcription initiation from chimeric target promoters (P(CTA)) in a low-temperature- inducible manner. Evaluation of the temperature-dependent CTA-P(CTA) interaction using a tailored ELISA-like cell-free assay correlated increased affinity of CTA for P(CTA) with temperature downshift. The temperature-inducible gene regulation (TIGR) system enabled tight repression in the chicken bursal B-cell line DT40 at 41 degrees C as well as precise titration of model product proteins up to maximum expression at or below 37 degrees C. Implantation of microencapsulated DT40 cells engineered for TIGR-controlled expression of the human vascular endothelial growth factor A (hVEGF121) provided low-temperature-induced VEGF-mediated vascularization in chicken embryos.


Assuntos
Proteínas de Bactérias , Temperatura Baixa , Fatores de Crescimento Endotelial/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Linfocinas/genética , Neovascularização Fisiológica , Engenharia de Proteínas , Ativação Transcricional , Animais , Vasos Sanguíneos/anatomia & histologia , Vasos Sanguíneos/crescimento & desenvolvimento , Sistema Livre de Células , Embrião de Galinha , Fatores de Crescimento Endotelial/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Linfocinas/metabolismo , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Temperatura , Transativadores/genética , Transativadores/metabolismo , Transfecção , Transgenes , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
7.
Hum Vaccin Immunother ; 11(1): 83-90, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25483519

RESUMO

Today, immune compromised interferon-α-receptor deficient mice expressing hCD46 (IFNARCD46tg) are usually used for measles virus (MV) based vaccine characterization. However, for the development of MV-based recombinant vaccine candidates (rMV), an immune competent mouse model is desirable in order to induce and evaluate meaningful immune response. In this study, humoral and cellular immune response induced by rMV in immune competent mice expressing human MV receptor CD46 (hCD46tg) were compared with those induced in wild-type black/6, and IFNARCD46tg mice.   All three strains developed humoral and cellular response against MV, whereas only hCD46tg and IFNARCD46tg mice developed a humoral response against the transgene. Differences were observed in the magnitude of the response, where the IFNARCD46tg mice displayed the strongest immune responses, followed by the hCD46tg mice and the black/6 mice. Interestingly, hCD46tg and wt black/6 mice showed a predominant CD4(+) T-cell response against MV-N, whereas IFNARCD46tg mice developed both, CD4(+) and CD8(+) T-cell response against MV-N. Analysis of the cytokine profile of MV-N specific CD4(+) T-cells and transgene (SIVgag) specific CD8(+) T-cells revealed qualitative differences of the T-cell responses; noticeably a significant reduction of the frequency of CD4(+)IL-2(+) expressing cells in IFNARCD46tg mice as compared with hCD46tg or wt black/6 mice. We show in this study significant quantitative and qualitative differences in immune responses between immune competent and immune-compromised mice. Our results therefore highlight the importance of the animal model and support the use of hCD46tg mice as mouse model for the characterization of the immunological profile induced by recombinant measles virus vaccine candidates.


Assuntos
Vacina contra Sarampo/imunologia , Vírus do Sarampo/imunologia , Modelos Animais , Animais , Anticorpos Antivirais/sangue , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Feminino , Expressão Gênica , Masculino , Sarampo/imunologia , Sarampo/prevenção & controle , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Vírus do Sarampo/genética , Proteína Cofatora de Membrana/genética , Camundongos , Camundongos Transgênicos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
8.
Hum Vaccin Immunother ; 9(3): 599-606, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23324399

RESUMO

Measles virus (MV) vectors are promising candidates for designing new recombinant vaccines since the parental live vaccines have a well-known safety and efficacy record. Like all viral vectors, the MV vector efficacy in inducing a protecting immune answer could be affected by the pre-existing immunity among the human population. In order to determine the optimal immunization route and regimen, we mimicked a MV pre-immunity by passively administrating MV neutralizing antibodies (MV-nAb) prior intramuscular (i.m.) and/or intranasal (i.n.) immunization with recombinant MV expressing the SIV-gag antigen (rMV-SIVgag). Our results revealed that 500 mIU of MV-nAb allowed the induction of a humoral and cellular immune response against the vector and the transgene, while higher titers of the MV-nAb were significantly inhibitory. In a prime-boost regimen, in the presence of MV-nAb, the intranasal-intramuscular (i.n.-i.m.) or intramuscular-intramuscular (i.m.-i.m.) routes induced higher humoral immune responses against the vector and the transgene (SIV-gag). In naive animals, cellular immune response was significantly higher by i.m. immunization; however, MV pre-immunity did not seem to affect the cellular immune response after an i.n. immunization.   In summary, we show that a pre-existing immunity of up to 500 mIU anti-MV neutralizing antibodies had little effect on the replication of rMV and did not inhibit the induction of significant humoral and cellular immune responses in immune-competent mice.


Assuntos
Anticorpos Antivirais/sangue , Portadores de Fármacos , Vetores Genéticos/imunologia , Imunização/métodos , Vírus do Sarampo/imunologia , Sarampo/imunologia , Vacinas Virais/imunologia , Administração Intranasal , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Humanos , Imunidade Celular , Injeções Intramusculares , Vírus do Sarampo/genética , Camundongos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
9.
Hum Vaccin Immunother ; 9(3): 607-13, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23324616

RESUMO

The measles virus vaccine (MVbv) is a clinically certified and well-tolerated vaccine strain that has been given both parenterally and mucosally. It has been extensively used in children and has proven to be safe and effective in eliciting protective immunity. This specific strain was therefore chosen to generate a measles viral vector. The genome of the commercial MVbv vaccine strain was isolated, sequenced and a plasmid, p(+)MVb, enabling transcription of the viral antigenome and rescue of MVb, was constructed. Phylogenic and phenotypic analysis revealed that MVbv and the rescued MVb constitute another evolutionary branch within the hitherto classified measles vaccines. Plasmid p(+)MVb was modified by insertion of artificial MV-type transcription units (ATUs) for the generation of recombinant viruses (rMVb) expressing additional proteins. Replication characteristics and immunogenicity of rMVb vectors were similar to the parental MVbv and to other vaccine strains. The expression of the additional proteins was stable over 10 serial virus transfers, which corresponds to an amplification greater than 10 ( 20) . The excellent safety record and its efficient application as aerosol may add to the usefulness of the derived vectors.


Assuntos
Vetores Genéticos , Vírus do Sarampo/genética , Vacinas Virais/imunologia , Animais , Chlorocebus aethiops , Análise por Conglomerados , Expressão Gênica , Instabilidade Genômica , Dados de Sequência Molecular , Filogenia , Plasmídeos , Análise de Sequência de DNA , Homologia de Sequência , Células Vero , Vacinas Virais/genética , Replicação Viral
10.
J Immunol ; 180(4): 2686-95, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18250481

RESUMO

Experimental autoimmune myocarditis (EAM) represents a Th17 T cell-mediated mouse model of postinflammatory heart disease. In BALB/c wild-type mice, EAM is a self-limiting disease, peaking 21 days after alpha-myosin H chain peptide (MyHC-alpha)/CFA immunization and largely resolving thereafter. In IFN-gammaR(-/-) mice, however, EAM is exacerbated and shows a chronic progressive disease course. We found that this progressive disease course paralleled persistently elevated IL-17 release from T cells infiltrating the hearts of IFN-gammaR(-/-) mice 30 days after immunization. In fact, IL-17 promoted the recruitment of CD11b(+) monocytes, the major heart-infiltrating cells in EAM. In turn, CD11b(+) monocytes suppressed MyHC-alpha-specific Th17 T cell responses IFN-gamma-dependently in vitro. In vivo, injection of IFN-gammaR(+/+)CD11b(+), but not IFN-gammaR(-/-)CD11b(+), monocytes, suppressed MyHC-alpha-specific T cells, and abrogated the progressive disease course in IFN-gammaR(-/-) mice. Finally, coinjection of MyHC-alpha-specific, but not OVA-transgenic, IFN-gamma-releasing CD4(+) Th1 T cell lines, together with MyHC-alpha-specific Th17 T cells protected RAG2(-/-) mice from EAM. In conclusion, CD11b(+) monocytes play a dual role in EAM: as a major cellular substrate of IL-17-induced inflammation and as mediators of an IFN-gamma-dependent negative feedback loop confining disease progression.


Assuntos
Doenças Autoimunes/prevenção & controle , Antígeno CD11b/biossíntese , Linfócitos T CD4-Positivos/imunologia , Interleucina-17/administração & dosagem , Interleucina-17/antagonistas & inibidores , Monócitos/imunologia , Miocardite/imunologia , Miocardite/prevenção & controle , Sequência de Aminoácidos , Animais , Doenças Autoimunes/patologia , Linfócitos T CD4-Positivos/patologia , Linhagem Celular , Movimento Celular/imunologia , Separação Celular , Progressão da Doença , Retroalimentação Fisiológica/imunologia , Soros Imunes/administração & dosagem , Interleucina-17/imunologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Dados de Sequência Molecular , Monócitos/citologia , Monócitos/metabolismo , Miocardite/patologia , Células Th1/imunologia
11.
Vaccine ; 26(17): 2164-74, 2008 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-18346823

RESUMO

Live attenuated recombinant measles viruses (rMV) expressing a codon-optimised spike glycoprotein (S) or nucleocapsid protein (N) of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) were generated (rMV-S and rMV-N). Both recombinant viruses stably expressed the corresponding SARS-CoV proteins, grew to similar end titres as the parental strain and induced high antibody titres against MV and the vectored SARS-CoV antigens (S and N) in transgenic mice susceptible to measles infection. The antibodies induced by rMV-S had a high neutralising effect on SARS-CoV as well as on MV. Moreover, significant N-specific cellular immune responses were measured by IFN-gamma ELISPOT assays. The pre-existence of anti-MV antibodies induced by the initial immunisation dose did not inhibit boost of anti-S and anti-N antibodies. Immunisations comprising a mixture of rMV-S and rMV-N induced immune responses similar in magnitude to that of vaccine components administered separately. These data support the suitability of MV as a bivalent candidate vaccine vector against MV and emerging viruses such as SARS-CoV.


Assuntos
Vírus do Sarampo/fisiologia , Glicoproteínas de Membrana/imunologia , Síndrome Respiratória Aguda Grave/prevenção & controle , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Animais , Animais Geneticamente Modificados , Vetores Genéticos/química , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Vacina contra Sarampo/imunologia , Vírus do Sarampo/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Testes de Neutralização , Proteínas do Nucleocapsídeo/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Síndrome Respiratória Aguda Grave/imunologia , Glicoproteína da Espícula de Coronavírus , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vacinas Virais/química , Vacinas Virais/genética , Vacinas Virais/imunologia
12.
Eur J Immunol ; 36(2): 494-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16402410

RESUMO

Osteopontin (OPN) and CD44 have been implicated in the development of autoimmune diseases, including arthritis and multiple sclerosis, as well as chronic inflammatory diseases, such as atherosclerosis and colitis. To investigate their roles in autoimmune myocarditis induced by immunization with heart alpha-myosin (MyHC-alpha), a mouse model of human cardiomyopathy, we analyzed mice lacking OPN or CD44v6/v7, a CD44 isoform that binds OPN. Both, OPN(-/-) and CD44v6/v7(-/-) mice developed myocarditis with the same prevalence and severity as BALB/c wild-type controls. Furthermore, treatment of BALB/c mice with a pan-neutralizing anti-CD44 antibody did not affect the disease outcome. Consistently, expansion of MyHC-alpha-specific autoimmune CD4(+) T cells and MyHC-alpha autoantibody responses from either CD44v6/v7(-/-) mice or OPN(-/-) mice was indistinguishable from their wild-type controls. Thus, OPN and CD44v6/v7 are merely spectators rather than protagonists in autoimmune myocarditis.


Assuntos
Doenças Autoimunes/imunologia , Glicoproteínas/imunologia , Receptores de Hialuronatos/imunologia , Miocardite/imunologia , Sialoglicoproteínas/imunologia , Transdução de Sinais/imunologia , Animais , Autoanticorpos/imunologia , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Modelos Animais de Doenças , Glicoproteínas/genética , Receptores de Hialuronatos/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miocardite/genética , Miocardite/patologia , Osteopontina , Sialoglicoproteínas/deficiência , Miosinas Ventriculares/administração & dosagem , Miosinas Ventriculares/efeitos adversos , Miosinas Ventriculares/imunologia
13.
Biotechnol Bioeng ; 80(6): 691-705, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12378611

RESUMO

The novel macrolide-inducible and -repressible mammalian gene regulation systems (E.REX) have been cloned into a variety of sophisticated expression configurations including (1) multi-purpose expression vectors, (2) pTRIDENT-based artificial operons, (3) dual-regulated expression strategies for independent control of two different transgenes, (4) autoregulated vectors for one-step installation of adjustable multigene expression, and (5) oncoretroviral and lentiviral plasmids for transduction of macrolide-, streptogramin- and tetracycline-dependent transactivators and production of cell lines supporting independent control of three different transgenes. This vector portfolio represents a construction kit-like toolbox for efficient installation of adjustable gene expression responsive to clinically licensed antibiotics and enables the design of multiregulated multigene metabolic engineering strategies required for biopharmaceutical manufacturing, gene therapy, and tissue engineering.


Assuntos
Antibacterianos/farmacologia , Células CHO/efeitos dos fármacos , Células CHO/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Engenharia Genética/métodos , Vetores Genéticos , Animais , Clonagem Molecular/métodos , Cricetinae , Terapia Genética/métodos , Macrolídeos/farmacologia , Família Multigênica , Estreptograminas/farmacologia , Tetraciclina/farmacologia , Engenharia Tecidual/métodos , Ativação Transcricional/efeitos dos fármacos , Transgenes/efeitos dos fármacos , Transgenes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA