Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
3.
Theriogenology ; 86(7): 1695-704, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27349138

RESUMO

Parthenogenesis, embryonic development without fertilization, resembles very early embryonic mortality in fertilized eggs. Also, parthenogenesis alters egg albumen characteristics in virgin Chinese Painted quail hens genetically selected for parthenogenesis (PV). When these PV hens are mated (PM), hatchability is reduced versus control mated (CM) hens that were not genetically selected for parthenogenesis. However, it is unclear if parthenogenesis, which occurs in PM hens, reduces hatchability due to infertility and altered albumen characteristics. Sperm-egg penetration (SEP) holes are indicative of true fertilization and may be useful in identifying if eggs from PM hens exhibit a decrease in fertility versus CM hens. Therefore, the objectives of this study were to determine if parthenogenesis in PM hens (1) decreases SEP, (2) alters albumen characteristics similar to parthenogenesis in eggs from PV hens, and (3) yields albumen characteristics similar to fertilized eggs containing early mortality. Daily, PV and PM eggs were collected, labeled, and incubated for 10 days, then broken out to determine the incidence of parthenogenesis and albumen characteristics. Also daily, fresh PM and CM quail eggs were macroscopically examined to determine if an egg was infertile with no embryonic development, parthenogenetic, or fertile. Each of these eggs was then microscopically examined for SEP. For both PV and PM incubated eggs, parthenogenesis decreased albumen pH, O2, and protein concentrations yet increased Ca(2+) and CO2 concentrations versus eggs with no development. For incubated PM eggs, albumen pH and O2 were lower, yet CO2 was higher for eggs containing parthenogens or early dead embryos versus infertile eggs. For SEP, fresh eggs classified as infertile or parthenogenetic from PM and CM hens had similar SEP holes but only one sixth as many SEP holes as eggs classified as fertilized. Eggs from CM hens had 3.5 times as many SEP holes as PM eggs. In conclusion, parthenogenesis that occurs in mated quail hens inhibits fertility and alters albumen characteristics similarly to parthenogenesis in unfertilized eggs and early embryonic mortality in fertilized eggs.


Assuntos
Coturnix/fisiologia , Ovalbumina/química , Partenogênese/fisiologia , Interações Espermatozoide-Óvulo/fisiologia , Animais , Cloretos/química , Copulação , Coturnix/embriologia , Coturnix/genética , Feminino , Masculino , Ovalbumina/fisiologia , Óvulo
4.
Biol Reprod ; 93(3): 71, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26157071

RESUMO

The ovalbumin gene family in Gallus gallus is composed of three homologous genes located within a 46 kb locus on chromosome 2: ovalbumin, ovalbumin-related protein Y (OVAY), and ovalbumin-related protein X (OVAX) genes. The expression of these genes in hen oviduct is under estrogen control, but their relative hormonal responsiveness and subsequent protein concentration in egg, is distinctive. Interestingly, all three proteins lack the classical signal peptide for secretion. Ovalbumin, OVAX, and OVAY belong to the serine protease inhibitor (serpin) family whose members share a common tertiary structure. Ovalbumin and OVAX are one of the few members of this family that do not express any protease inhibition activity whereas OVAY has been predicted to be inhibitory, by comparison with the consensus sequence for inhibitory serpins. In contrast to ovalbumin and OVAY, OVAX interacts with heparin, a negatively charged glycosaminoglycan, via a positively charged domain exposed at the surface of the molecule. Ovalbumin is the major egg white protein and might be a source of amino acids for the developing embryo. The physiological function of OVAY is not known, but recent data have revealed a possible role of this protein in early embryonic development. Considering the antibacterial activities of OVAX, this protein might play a role in egg defense. This review sheds light on the expression, biochemistry, and structural specificities of these three highly similar paralogs. It gives new clues in favor of diverging functions, which are likely to have arisen by duplication events from a common ancestral gene.


Assuntos
Proteínas Aviárias/genética , Proteínas Aviárias/fisiologia , Aves/fisiologia , Proteínas do Ovo/genética , Proteínas do Ovo/fisiologia , Ovalbumina/genética , Ovalbumina/fisiologia , Serpinas/genética , Serpinas/fisiologia , Sequência de Aminoácidos , Animais , Evolução Biológica , Galinhas , Humanos , Dados de Sequência Molecular
5.
Blood ; 124(4): 628-37, 2014 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-24855206

RESUMO

Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) are threatened by potentially lethal viral manifestations like cytomegalovirus (CMV) reactivation. Because the success of today's virostatic treatment is limited by side effects and resistance development, adoptive transfer of virus-specific memory T cells derived from the stem cell donor has been proposed as an alternative therapeutic strategy. In this context, dose minimization of adoptively transferred T cells might be warranted for the avoidance of graft-versus-host disease (GVHD), in particular in prophylactic settings after T-cell-depleting allo-HSCT protocols. To establish a lower limit for successful adoptive T-cell therapy, we conducted low-dose CD8(+) T-cell transfers in the well-established murine Listeria monocytogenes (L.m.) infection model. Major histocompatibility complex-Streptamer-enriched antigen-specific CD62L(hi) but not CD62L(lo) CD8(+) memory T cells proliferated, differentiated, and protected against L.m. infections after prophylactic application. Even progenies derived from a single CD62L(hi) L.m.-specific CD8(+) T cell could be protective against bacterial challenge. In analogy, low-dose transfers of Streptamer-enriched human CMV-specific CD8(+) T cells into allo-HSCT recipients led to strong pathogen-specific T-cell expansion in a compassionate-use setting. In summary, low-dose adoptive T-cell transfer (ACT) could be a promising strategy, particularly for prophylactic treatment of infectious complications after allo-HSCT.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Doença Enxerto-Hospedeiro/imunologia , Imunoterapia Adotiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Imunodeficiência Combinada Severa/imunologia , Adolescente , Animais , Diferenciação Celular , Proliferação de Células , Criança , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/terapia , Doença Enxerto-Hospedeiro/metabolismo , Doença Enxerto-Hospedeiro/terapia , Transplante de Células-Tronco Hematopoéticas , Proteínas de Homeodomínio/fisiologia , Humanos , Imunização , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/fisiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Imunodeficiência Combinada Severa/metabolismo , Imunodeficiência Combinada Severa/terapia , Transplante Homólogo , Ativação Viral
6.
Am J Transplant ; 13(11): 3021-30, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24007441

RESUMO

The use of monoclonal antibodies targeting the CD154 molecule remains one of the most effective means of promoting graft tolerance in animal models, but thromboembolic complications during early clinical trials have precluded their use in humans. Furthermore, the role of Fc-mediated deletion of CD154-expressing cells in the observed efficacy of these reagents remains controversial. Therefore, determining the requirements for anti-CD154-induced tolerance will instruct the development of safer but equally efficacious treatments. To investigate the mechanisms of action of anti-CD154 therapy, two alternative means of targeting the CD40-CD154 pathway were used: a nonagonistic anti-CD40 antibody and an Fc-silent anti-CD154 domain antibody. We compared these therapies to an Fc-intact anti-CD154 antibody in both a fully allogeneic model and a surrogate minor antigen model in which the fate of alloreactive cells could be tracked. Results indicated that anti-CD40 mAbs as well as Fc-silent anti-CD154 domain antibodies were equivalent to Fc-intact anti-CD154 mAbs in their ability to inhibit alloreactive T cell expansion, attenuate cytokine production of antigen-specific T cells and promote the conversion of Foxp3(+) iTreg. Importantly, iTreg conversion observed with Fc-silent anti-CD154 domain antibodies was preserved in the presence of CTLA4-Ig, suggesting that this therapy is a promising candidate for translation to clinical use.


Assuntos
Anticorpos Monoclonais/farmacologia , Ligante de CD40/antagonistas & inibidores , Fatores de Transcrição Forkhead/metabolismo , Sobrevivência de Enxerto/imunologia , Imunoconjugados/farmacologia , Imunossupressores/farmacologia , Linfócitos T Reguladores/imunologia , Abatacepte , Animais , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Ligante de CD40/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/fisiologia , Transplante de Pele , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Doadores de Tecidos , Transplante Homólogo
7.
Am J Physiol Lung Cell Mol Physiol ; 305(5): L364-76, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23831616

RESUMO

Asthma is a chronic inflammatory disease of the small airways, with airway hyperresponsiveness (AHR) and inflammation as hallmarks. Recent studies suggest a role for arginase in asthma pathogenesis, possibly because arginine is the substrate for both arginase and NO synthase and because NO modulates bronchial tone and inflammation. Our objective was to investigate the importance of increased pulmonary arginase 1 expression on methacholine-induced AHR and lung inflammation in a mouse model of allergic asthma. Arginase 1 expression in the lung was ablated by crossing Arg1(fl/fl) with Tie2Cre(tg/-) mice. Mice were sensitized and then challenged with ovalbumin. Lung function was measured with the Flexivent. Adaptive changes in gene expression, chemokine and cytokine secretion, and lung histology were quantified with quantitative PCR, ELISA, and immunohistochemistry. Arg1 deficiency did not affect the allergic response in lungs and large-airway resistance, but it improved peripheral lung function (tissue elastance and resistance) and attenuated adaptive increases in mRNA expression of arginine-catabolizing enzymes Arg2 and Nos2, arginine transporters Slc7a1 and Slc7a7, chemokines Ccl2 and Ccl11, cytokines Tnfa and Ifng, mucus-associated epithelial markers Clca3 and Muc5ac, and lung content of IL-13 and CCL11. However, expression of Il4, Il5, Il10, and Il13 mRNA; lung content of IL-4, IL-5, IL-10, TNF-α, and IFN-γ protein; and lung pathology were not affected. Correlation analysis showed that Arg1 ablation disturbed the coordinated pulmonary response to ovalbumin challenges, suggesting arginine (metabolite) dependence of this response. Arg1 ablation in the lung improved peripheral lung function and affected arginine metabolism but had little effect on airway inflammation.


Assuntos
Arginase/fisiologia , Asma/fisiopatologia , Hiper-Reatividade Brônquica/patologia , Hipersensibilidade/patologia , Pulmão/fisiologia , Pneumonia/patologia , Sistema Respiratório/patologia , Resistência das Vias Respiratórias/fisiologia , Animais , Western Blotting , Hiper-Reatividade Brônquica/induzido quimicamente , Hiper-Reatividade Brônquica/metabolismo , Broncoconstritores/toxicidade , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Perfilação da Expressão Gênica , Hipersensibilidade/metabolismo , Técnicas Imunoenzimáticas , Pulmão/citologia , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Cloreto de Metacolina/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/citologia , Células Mieloides/metabolismo , Ovalbumina/fisiologia , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
J Immunol ; 188(11): 5227-37, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22508931

RESUMO

Plasmacytoid dendritic cells (pDCs) are highly specialized APCs that, in addition to their well-recognized role in anti-viral immunity, also regulate immune responses. Liver-resident pDCs are considerably less immunostimulatory than those from secondary lymphoid tissues and are equipped to promote immune tolerance/regulation through various mechanisms. IL-27 is an IL-12 family cytokine that regulates the function of both APCs and T cells, although little is known about its role in pDC immunobiology. In this study, we show that mouse liver pDCs express higher levels of IL-27p28 and EBV-induced protein 3 (Ebi3) compared with those of splenic pDCs. Both populations of pDCs express the IL-27Rα/WSX-1; however, only liver pDCs significantly upregulate expression of the coregulatory molecule B7 homolog-1 (B7-H1) in response to IL-27. Inhibition of STAT3 activation completely abrogates IL-27-induced upregulation of B7-H1 expression on liver pDCs. Liver pDCs treated with IL-27 increase the percentage of CD4(+)Foxp3(+) T cells in MLR, which is dependent upon expression of B7-H1. pDCs from Ebi3-deficient mice lacking functional IL-27 show increased capacity to stimulate allogeneic T cell proliferation and IFN-γ production in MLR. Liver but not spleen pDCs suppress delayed-type hypersensitivity responses to OVA, an effect that is lost with Ebi3(-/-) and B7-H1(-/-) liver pDCs compared with wild-type liver pDCs. These data suggest that IL-27 signaling in pDCs promotes their immunoregulatory function and that IL-27 produced by pDCs contributes to their capacity to regulate immune responses in vitro and in vivo.


Assuntos
Antígeno B7-H1/biossíntese , Células Dendríticas/imunologia , Interleucinas/biossíntese , Fígado/imunologia , Fator de Transcrição STAT3/fisiologia , Regulação para Cima/imunologia , Animais , Antígeno B7-H1/deficiência , Células Dendríticas/metabolismo , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Humanos , Hipersensibilidade Tardia/genética , Hipersensibilidade Tardia/imunologia , Hipersensibilidade Tardia/patologia , Fígado/citologia , Fígado/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antígenos de Histocompatibilidade Menor , Ovalbumina/fisiologia , Receptores de Citocinas/biossíntese , Regulação para Cima/genética
10.
J Immunol ; 187(12): 6499-507, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22079987

RESUMO

Regulatory T cells (Tregs) play a critical role in the maintenance of airway tolerance. We report that inhaled soluble Ag induces adaptive Foxp3(+) Tregs, as well as a regulatory population of CD4(+) T cells in the lungs and lung-draining lymph nodes that express latency-associated peptide (LAP) on their cell surface but do not express Foxp3. Blocking the cytokine IL-10 or TGF-ß prevented the generation of LAP(+) Tregs and Foxp3(+) Tregs in vivo, and the LAP(+) Tregs could also be generated concomitantly with Foxp3(+) Tregs in vitro by culturing naive CD4(+) T cells with Ag and exogenous TGF-ß. The LAP(+) Tregs strongly suppressed naive CD4(+) T cell proliferation, and transfer of sorted OVA-specific LAP(+) Tregs in vivo inhibited allergic eosinophilia and Th2 cytokine expression in the lung, either when present at the time of Th2 sensitization or when injected after Th2 cells were formed. Furthermore, inflammatory innate stimuli from house dust mite extract, nucleotide-binding oligomerization domain containing 2 ligand, and LPS, which are sufficient for blocking airway tolerance, strongly decreased the induction of LAP(+) Tregs. Taken together, we concluded that inducible Ag-specific LAP(+) Tregs can suppress asthmatic lung inflammation and constitute a mediator of airway tolerance together with Foxp3(+) Tregs.


Assuntos
Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/deficiência , Hipersensibilidade Respiratória/patologia , Hipersensibilidade Respiratória/prevenção & controle , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1/biossíntese , Alérgenos/administração & dosagem , Alérgenos/fisiologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Epitopos de Linfócito T/biossíntese , Epitopos de Linfócito T/genética , Fatores de Transcrição Forkhead/genética , Genes Reporter , Tolerância Imunológica/genética , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Ovalbumina/fisiologia , Hipersensibilidade Respiratória/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta1/genética
11.
PLoS One ; 6(7): e22936, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21829561

RESUMO

Effective control of the intracellular protozoan parasite Toxoplasma gondii depends on the activation of antigen-specific CD8(+) T-cells that manage acute disease and prevent recrudescence during chronic infection. T-cell activation in turn, requires presentation of parasite antigens by MHC-I molecules on the surface of antigen presenting cells. CD8(+) T-cell epitopes have been defined for several T. gondii proteins, but it is unclear how these antigens enter into the presentation pathway. We have exploited the well-characterized model antigen ovalbumin (OVA) to investigate the ability of parasite proteins to enter the MHC-I presentation pathway, by engineering recombinant expression in various organelles. CD8(+) T-cell activation was assayed using 'B3Z' reporter cells in vitro, or adoptively-transferred OVA-specific 'OT-I' CD8(+) T-cells in vivo. As expected, OVA secreted into the parasitophorous vacuole strongly stimulated antigen-presenting cells. Lower levels of activation were observed using glycophosphatidyl inositol (GPI) anchored OVA associated with (or shed from) the parasite surface. Little CD8(+) T-cell activation was detected using parasites expressing intracellular OVA in the cytosol, mitochondrion, or inner membrane complex (IMC). These results indicate that effective presentation of parasite proteins to CD8(+) T-cells is a consequence of active protein secretion by T. gondii and escape from the parasitophorous vacuole, rather than degradation of phagocytosed parasites or parasite products.


Assuntos
Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Ativação Linfocitária/imunologia , Toxoplasmose/imunologia , Toxoplasmose/parasitologia , Vacúolos/imunologia , Transferência Adotiva , Animais , Células Apresentadoras de Antígenos/imunologia , Western Blotting , Células Cultivadas , Feminino , Imunofluorescência , Humanos , Complexo Principal de Histocompatibilidade/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/fisiologia , Frações Subcelulares , Toxoplasma/genética , Toxoplasma/imunologia , Toxoplasmose/metabolismo , Vacúolos/parasitologia
12.
J Immunol ; 187(1): 133-40, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21642544

RESUMO

Memory T cells are distinguished from naive T cells by their rapid production of effector cytokines, although mechanisms for this recall response remain undefined. In this study, we investigated transcriptional mechanisms for rapid IFN-γ production by Ag-specific memory CD4 T cells. In naive CD4 T cells, IFN-γ production only occurred after sustained Ag activation and was associated with high expression of the T-bet transcription factor required for Th1 differentiation and with T-bet binding to the IFN-γ promoter as assessed by chromatin immunoprecipitation analysis. By contrast, immediate IFN-γ production by Ag-stimulated memory CD4 T cells occurred in the absence of significant nuclear T-bet expression or T-bet engagement on the IFN-γ promoter. We identified rapid induction of NF-κB transcriptional activity and increased engagement of NF-κB on the IFN-γ promoter at rapid times after TCR stimulation of memory compared with naive CD4 T cells. Moreover, pharmacologic inhibition of NF-κB activity or peptide-mediated inhibition of NF-κB p50 translocation abrogated early memory T cell signaling and TCR-mediated effector function. Our results reveal a molecular mechanism for memory T cell recall through enhanced NF-κB p50 activation and promoter engagement, with important implications for memory T cell modulation in vaccines, autoimmunity, and transplantation.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Memória Imunológica , Ativação Linfocitária/imunologia , Transcrição Gênica/imunologia , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/citologia , Células Cultivadas , Densitometria , Memória Imunológica/genética , Interferon gama/biossíntese , Interferon gama/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Subunidade p50 de NF-kappa B/biossíntese , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/fisiologia , Ovalbumina/imunologia , Ovalbumina/farmacocinética , Ovalbumina/fisiologia , Fragmentos de Peptídeos/fisiologia , Regiões Promotoras Genéticas/imunologia , Fase de Repouso do Ciclo Celular/genética , Fase de Repouso do Ciclo Celular/imunologia , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/genética , Regulação para Cima/genética , Regulação para Cima/imunologia
13.
Cancer Immunol Immunother ; 60(7): 1029-38, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21519830

RESUMO

Staphylococcal enterotoxins A (SEA) and B (SEB) are classical models of superantigens (SAg), which induce potent T-cell-stimulating activity by forming complexes with MHC class II molecules on antigen-presenting cells. This large-scale activation of T-cells is accompanied by increased production of cytokines such as interferon-γ (IFN-γ). Additionally, as we previously reported, IFN-γ-producing CD8(+) T cells act as "helper cells," supporting the ability of dendritic cells to produce interleukin-12 (IL-12)p70. Here, we show that DC pulsed with SAg promote the enhancement of anti-tumor immunity. Murine bone marrow-derived dendritic cells (DC) were pulsed with OVA(257-264) (SIINFEKL), which is an H-2Kb target epitope of EG7 [ovalbumin (OVA)-expressing EL4] cell lines, in the presence of SEA and SEB and were subcutaneously injected into naïve C57BL/6 mice. SAg plus OVA(257-264)-pulsed DC vaccine strongly enhanced peptide-specific CD8(+) T cells exhibiting OVA(257-264)-specific cytotoxic activity and IFN-γ production, leading to the induction of protective immunity against EG7 tumors. Furthermore, cyclophosphamide (CY) added to SAg plus tumor-antigens (OVA(257-264), tumor lysate, or TRP-2) pulsed DC immunization markedly enhanced tumor-specific T-cell expansion and had a significant therapeutic effect against various tumors (EG7, 2LL, and B16). Superantigens are potential candidates for enhancing tumor immunity in DC vaccines.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Carcinoma Pulmonar de Lewis/imunologia , Células Dendríticas/imunologia , Linfoma/imunologia , Melanoma Experimental/imunologia , Superantígenos/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antineoplásicos Alquilantes/uso terapêutico , Linfócitos T CD8-Positivos , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Ciclofosfamida/uso terapêutico , Citocinas/metabolismo , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/metabolismo , Interferon gama/metabolismo , Interleucina-12/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Ativação Linfocitária , Linfoma/tratamento farmacológico , Linfoma/metabolismo , Masculino , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Taxa de Sobrevida , Linfócitos T Auxiliares-Indutores/imunologia , Células Tumorais Cultivadas , Vacinas de Subunidades Antigênicas/uso terapêutico
14.
Int J Cancer ; 128(6): 1371-83, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20473945

RESUMO

We recently showed that MGL2 specifically binds tumour-associated glycan N-acetylgalactosamine (GalNAc). We here demonstrate that modification of an antigen with tumour-associated glycan GalNAc, targets antigen specifically to the MGL2 on bone marrow derived (BM)-DCs and splenic DCs. Glycan-modification of antigen with GalNAc that mimics tumour-associated glycosylation, promoted antigen internalisation in DCs and presentation to CD4 T cells, as well as differentiation of IFN-γ producing CD4 T cells. Furthermore, GalNAc modified antigen enhanced cross-presentation of both BM-DCs and primary splenic DCs resulting in enhanced antigen specific CD8 T cell responses. Using MyD88-TRIFF(-/-) BM-DCs we demonstrate that the enhanced cross-presentation of the GalNAc modified antigen is TLR independent. Our data strongly suggest that tumour-associated GalNAc modification of antigen targets MGL on DCs and greatly enhances both MHC class II and class I presentation in a TLR independent manner.


Assuntos
Acetilgalactosamina/metabolismo , Antígenos Glicosídicos Associados a Tumores/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Lectinas Tipo C/fisiologia , Animais , Apresentação de Antígeno/imunologia , Western Blotting , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células CHO , Proliferação de Células , Cricetinae , Cricetulus , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Glicosilação , Antígenos de Histocompatibilidade Classe I/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/fisiologia , Ovalbumina/fisiologia , RNA Mensageiro/genética , Receptores de Antígenos de Linfócitos T/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/imunologia , Baço/metabolismo
15.
Poult Sci ; 89(11): 2441-52, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20952708

RESUMO

In the present study, we examined the composition, amount, and uptake of yolk nutrients [fat, protein, water, and carbohydrates (COH)] during incubation of eggs from 30- and 50-wk-old broiler breeder hens. Eggs were sampled at embryonic d 0 (fresh eggs), 13, 15, 17, 19, and 21 (hatch). Egg, embryo, yolk content, and yolk sac membrane were weighed, and the yolk sac (YS; i.e., yolk content + yolk sac membrane) composition was analyzed. From 30 to 50 wk of age, the albumen weight increased by 13.3%, whereas the yolk increased by more than 40%. The proportion of fat in the fresh yolk of the 30-wk-old group was 23.8% compared with 27.4% in the 50-wk-old group, whereas the proportion of protein was 17.9% compared with 15.6%, respectively. During incubation, results indicated that water and protein infiltrated from other egg compartments to the YS. Accordingly, the calculated change in the content of water and protein between fresh yolk and sampled YS does not represent the true uptake of these components from the YS to the embryo, and only fat uptake from the YS can be accurately estimated. By embryonic d 15, fat uptake relative to embryo weight was lower in the 30-wk-old group than in the 50-wk-old group. However, by embryonic d 21, embryos of both groups reached similar relative fat uptake, suggesting that to hatch, embryos must attain a certain amount of fat as a source of energy for the hatching process. The amount of COH in the YS increased similarly during incubation in eggs from hens of both ages, reaching a peak at embryonic d 19, suggesting COH synthesis in the YS. At hatch, the amount of protein, water, and COH in the residual YS, relative to the weight of the yolk-free chick, was similar in eggs from young and old hens. However, chicks from the younger hens had less fat in the YS for their immediate posthatch nutrition compared with those from the older hens.


Assuntos
Embrião de Galinha/fisiologia , Galinhas/fisiologia , Saco Vitelino/fisiologia , Envelhecimento/fisiologia , Animais , Galinhas/crescimento & desenvolvimento , Gema de Ovo/fisiologia , Ovos , Desenvolvimento Embrionário/fisiologia , Feminino , Lipídeos/análise , Fenômenos Fisiológicos da Nutrição/fisiologia , Ovalbumina/fisiologia
16.
Am J Pathol ; 177(4): 1657-64, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20724599

RESUMO

The mechanisms underlying the exaggerated distal airway inflammation and hyperresponsiveness that characterize acute exacerbations of asthma are largely unknown. Using BALB/c mouse experimental models, we demonstrated a potentially important role for alveolar macrophages (AM) in the development of an allergen-induced exacerbation of asthma. To induce features of airway inflammation and remodeling characteristic of mild chronic asthma, animals were systemically sensitized and exposed to low mass concentrations (≈3 mg/m(3)) of aerosolized ovalbumin for 30 minutes per day, 3 days per week, for 4 weeks. A subsequent single moderate-level challenge (≈30 mg/m(3)) was used to trigger an acute exacerbation. In chronically challenged animals, cytokine expression by AM was not increased, whereas after an acute exacerbation, AM exhibited significantly enhanced expression of proinflammatory cytokines, including interleukin (IL) 1ß, IL-6, CXCL-1, and tumor necrosis factor α. In parallel, there was a marked increase in the expression of several cytokines by CD4(+) T-lymphocytes, notably the Th2 cytokines IL-4 and IL-13. Importantly, AM from an acute exacerbation stimulated the expression of Th2 cytokines when cocultured with CD4(+) cells from chronically challenged animals, and their ability to do so was significantly greater than AM from either chronically challenged or naïve controls. Stimulation was partly dependent on interactions involving CD80/86. We conclude that in an acute exacerbation of asthma, enhanced cytokine expression by AM may play a critical role in triggering increased expression of cytokines by pulmonary CD4(+) T-lymphocytes.


Assuntos
Asma/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Pulmão/imunologia , Macrófagos Alveolares/imunologia , Células Th2/imunologia , Alérgenos/imunologia , Animais , Asma/patologia , Hiper-Reatividade Brônquica , Líquido da Lavagem Broncoalveolar , Linfócitos T CD4-Positivos/patologia , Doença Crônica , Citocinas/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Volume Expiratório Forçado , Inflamação/imunologia , Inflamação/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/fisiologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Th2/patologia
17.
Am J Respir Cell Mol Biol ; 43(3): 269-75, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19805483

RESUMO

Allergic asthma, a major cause of morbidity and leading cause of hospitalizations, is an inflammatory disease orchestrated by T helper cells and characterized by the lung migration of eosinophils, which are important asthma effector cells. Lung migration of inflammatory cells requires, among other events, the chemokine receptor transduction of lung-produced inflammatory chemokines. Despite the widespread prevalence of this disease, the molecular mechanisms regulating chemokine production and receptor regulation in asthma are poorly understood. Previous work from our laboratory demonstrated that beta-arrestin-2 positively regulates the development of allergic airway disease in a mouse model, partly through positive regulation of T-lymphocyte chemotaxis to the lung. However, beta-arrestin-2 is expressed in many cell types, including other hematopoietic cells and lung structural cells, which are involved in the development and manifestation of allergic airway disease. To determine the cell types required for beta-arrestin-2-dependent allergic inflammation, we generated bone marrow chimera mice. Using the ovalbumin murine model of allergic airway disease, we show that eosinophilic and lymphocytic inflammation is restored in chimeric mice, with expression of beta-arrestin-2 exclusively on hematopoietic-derived cell types. In contrast, airway hyperresponsiveness is dependent on the expression of beta-arrestin-2 in structural cells. Our data demonstrate that the expression of beta-arrestin-2 in at least two divergent cell types contributes to the pathogenesis of allergic airway disease.


Assuntos
Arrestinas/metabolismo , Asma/metabolismo , Eosinófilos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Pulmão/metabolismo , Sistema Respiratório/metabolismo , Transferência Adotiva , Animais , Medula Óssea/metabolismo , Transplante de Medula Óssea , Citometria de Fluxo , Interleucina-13/farmacologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina/fisiologia , Linfócitos T/metabolismo , beta-Arrestina 2 , beta-Arrestinas
18.
J Immunol ; 184(3): 1419-24, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20038640

RESUMO

The defective ribosomal product (DRiP) hypothesis of endogenous Ag processing posits that rapidly degraded forms of nascent proteins are a major source of peptide ligands for MHC class I molecules. Although there is broad experimental support for the DRiP hypothesis, careful kinetic analysis of the generation of defined peptide class I complexes has been limited to studies of recombinant vaccinia viruses expressing genes derived from other organisms. In this study, we show that insertion of the SIINFEKL peptide into the stalk of influenza A virus neuraminidase (NA) does not detectably modify NA folding, degradation, transport, or sp. act. when expressed in its natural context of influenza A virus infection. Using the 25-D1.16 mAb specific for K(b)-SIINFEKL to precisely quantitate cell surface complexes by flow cytometry, we demonstrate that SIINFEKL is generated in complete lockstep with initiation and abrogation of NA biosynthesis in both L-K(b) fibroblast cells and DC2.4 dendritic/monocyte cells. SIINFEKL presentation requires active proteasomes and TAP, consistent with its generation from a cytosolic DRiP pool. From the difference in the shutoff kinetics of K(b)-SIINFEKL complex expression following protein synthesis versus proteasome inhibition, we estimate that the t(1/2) of the biosynthetic source of NA peptide is approximately 5 min. These observations extend the relevance of the DRiP hypothesis to viral proteins generated in their natural context.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos Virais/biossíntese , Vírus da Influenza A Subtipo H1N1/enzimologia , Vírus da Influenza A Subtipo H1N1/imunologia , Neuraminidase/metabolismo , Biossíntese de Proteínas/imunologia , Proteínas Ribossômicas/biossíntese , Proteínas Ribossômicas/deficiência , Sequência de Aminoácidos , Animais , Antígenos Virais/metabolismo , Linhagem Celular , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Cães , Ativação Enzimática/imunologia , Estabilidade Enzimática/imunologia , Epitopos/biossíntese , Epitopos/metabolismo , Fibroblastos/enzimologia , Fibroblastos/imunologia , Fibroblastos/virologia , Antígenos H-2/biossíntese , Antígenos H-2/metabolismo , Células L , Camundongos , Dados de Sequência Molecular , Monócitos/enzimologia , Monócitos/imunologia , Monócitos/virologia , Neuraminidase/biossíntese , Infecções por Orthomyxoviridae/enzimologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Ovalbumina/metabolismo , Ovalbumina/fisiologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/fisiologia , Dobramento de Proteína , Transporte Proteico/imunologia , Proteínas Ribossômicas/metabolismo
19.
Am J Physiol Lung Cell Mol Physiol ; 297(4): L698-705, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19648284

RESUMO

The effects of remodeling of airway smooth muscle (SM) by hyperplasia on airway SM contractility in vivo are poorly explored. The aim of this study was to investigate the relationship between allergen-induced airway SM hyperplasia and its contractile phenotype. Brown Norway rats were sensitized with ovalbumin (OVA) or saline on day 0 and then either OVA-challenged once on day 14 and killed 24 h later or OVA-challenged 3 times (on days 14, 19, and 24) and killed 2 or 7 days later. Changes in SM mass, expression of total myosin, SM myosin heavy chain fast isoform (SM-B) and myosin light chain kinase (MLCK), tracheal contractions ex vivo, and airway responsiveness to methacholine (MCh) in vivo were assessed. One day after a single OVA challenge, the number of SM cells positive for PCNA was greater than for control animals, whereas the SM mass, contractile phenotype, and tracheal contractility were unchanged. Two days after three challenges, SM mass and PCNA immunoreactive cells were increased (3- and 10-fold, respectively; P < 0.05), but airway responsiveness to MCh was unaffected. Lower expression in total myosin, SM-B, and MLCK was observed at the mRNA level (P < 0.05), and total myosin and MLCK expression were lower at the protein level (P < 0.05) after normalization for SM mass. Normalized tracheal SM force generation was also significantly lower 2 days after repeated challenges (P < 0.05). Seven days after repeated challenges, features of remodeling were restored toward control levels. Allergen-induced hyperplasia of SM cells was associated with a loss of contractile phenotype, which was offset by the increase in mass.


Assuntos
Alérgenos/farmacologia , Asma/imunologia , Hiper-Reatividade Brônquica/imunologia , Modelos Animais de Doenças , Músculo Liso/efeitos dos fármacos , Sistema Respiratório/imunologia , Animais , Asma/tratamento farmacológico , Asma/metabolismo , Western Blotting , Hiper-Reatividade Brônquica/metabolismo , Hiper-Reatividade Brônquica/patologia , Broncoconstritores/farmacologia , Masculino , Cloreto de Metacolina/farmacologia , Músculo Liso/imunologia , Músculo Liso/patologia , Ovalbumina/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos BN , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Sistema Respiratório/metabolismo , Sistema Respiratório/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Immunol ; 183(5): 3130-8, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19675167

RESUMO

Although the pathogenic role of B cells and CD4 T cells has been studied extensively, less is known about the role of CD8 T cells in autoimmunity and self-tolerance. To evaluate the role of CD8 T cells in autoimmunity and its modulation using self-peptides, we used mice expressing soluble OVA (sOVA) under control of the keratin-14 promoter. Spontaneous autoimmunity occurred when sOVA mice were crossed with OT-I mice, whose CD8 T cells carry a Valpha2/Vbeta5-transgenic TCR with specificity for the OVA(257-264) peptide. Eighty-three percent of OVA/OT-I mice died during the first 2 wk of life due to multiple organ inflammation. In contrast, preventive or therapeutic OVA(257-264) peptide injections induced a dose-dependent increase in survival. Healthy survivors exhibited reductions in peripheral CD8 T cells, CD8 coreceptor, and Valpha2 expression. Furthermore, CD8 T cells from healthy mice were anergic and could not be activated by exogenous IL-2. A block in IL-2/IL-7 signaling via the STAT5 pathway provided the basis for low surface expression of the CD8 coreceptor and failure of IL-2 to break CD8 T cell anergy. Thus, the soluble TCR ligand triggered multiple tolerance mechanisms in these sOVA/OT-I mice, making this treatment approach a potential paradigm for modulating human autoimmune diseases.


Assuntos
Doenças Autoimunes/terapia , Regulação para Baixo/imunologia , Interleucina-2/antagonistas & inibidores , Interleucina-7/antagonistas & inibidores , Ovalbumina/fisiologia , Fragmentos de Peptídeos/fisiologia , Receptores de Antígenos de Linfócitos T alfa-beta/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Fator de Transcrição STAT5/antagonistas & inibidores , Transdução de Sinais/imunologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/mortalidade , Antígenos CD8/metabolismo , Galinhas , Anergia Clonal/genética , Anergia Clonal/imunologia , Regulação para Baixo/genética , Tolerância Imunológica/genética , Interleucina-2/fisiologia , Interleucina-7/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Ovalbumina/genética , Ovalbumina/imunologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/fisiologia , Transdução de Sinais/genética , Solubilidade , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...