Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 9: 2532, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30429857

RESUMO

Immunosuppression is one hallmark of sepsis, decreasing the host response to the primary septic pathogens and/or secondary nosocomial infections. CD4 T cells and B cells are among the array of immune cells that experience reductions in number and function during sepsis. "Help" from follicular helper (Tfh) CD4 T cells to B cells is needed for productive and protective humoral immunity, but there is a paucity of data defining the effect of sepsis on a primary CD4 T cell-dependent B cell response. Using the cecal ligation and puncture (CLP) mouse model of sepsis induction, we observed reduced antibody production in mice challenged with influenza A virus or TNP-KLH in alum early (2 days) and late (30 days) after CLP surgery compared to mice subjected to sham surgery. To better understand how these CD4 T cell-dependent B cell responses were altered by a septic event, we immunized mice with a Complete Freund's Adjuvant emulsion containing the MHC II-restricted peptide 2W1S56-68 coupled to the fluorochrome phycoerythrin (PE). Immunization with 2W1S-PE/CFA results in T cell-dependent B cell activation, giving us the ability to track defined populations of antigen-specific CD4 T cells and B cells responding to the same immunogen in the same mouse. Compared to sham mice, differentiation and class switching in PE-specific B cells were blunted in mice subjected to CLP surgery. Similarly, mice subjected to CLP had reduced expansion of 2W1S-specific T cells and Tfh differentiation after immunization. Our data suggest CLP-induced sepsis impacts humoral immunity by affecting the number and function of both antigen-specific B cells and CD4 Tfh cells, further defining the period of chronic immunoparalysis after sepsis induction.


Assuntos
Linfócitos B/imunologia , Coinfecção/imunologia , Vírus da Influenza A/fisiologia , Infecções por Orthomyxoviridae/imunologia , Sepse/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anticorpos/metabolismo , Antígenos de Bactérias/imunologia , Ceco/cirurgia , Diferenciação Celular , Células Cultivadas , Doença Crônica , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Tolerância Imunológica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL
2.
J Biomed Nanotechnol ; 12(6): 1303-11, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27319223

RESUMO

Biodegradable polymeric nanoparticle-based subunit vaccines have shown promising characteristics by enhancing antigen presentation and inducing protective immune responses when compared with soluble protein. Specifically, polyanhydride nanoparticle-based vaccines (i.e., nanovaccines) have been shown to successfully encapsulate and release antigens, activate B and T cells, and induce both antibody- and cell-mediated immunity towards a variety of immunogens. One of the characteristics of strong thymus-dependent antibody responses is the formation of germinal centers (GC) and the generation of GC B cells, which is part of the T helper cell driven cellular response. In order to further understand the role of nanovaccines in the induction of antigen-specific immune responses, their ability to induce germinal center B cell formation and isotype switching and the effects thereof on serum antibody responses were investigated in these studies. Polyanhydride nanovaccines based on 1,6-bis(p-carboxyphenoxy)hexane and 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane were used to subcutaneously administer a viral antigen. GC B cell formation and serum antibody responses induced by the nanovaccines were compared to that induced by alum-based vaccine formulations. It was demonstrated that a single dose of polyanhydride nanovaccines resulted in the formation of robust GCs and serum antibody in comparison to that induced by the alum-based formulation. This was attributed to the sustained release of antigen provided by the nanovaccines. When administered in a multiple dose regimen, the highest post-immunization titer and GC B cell number was enhanced, and the immune response induced by the nanovaccines was further sustained. These studies provide foundational information on the mechanism of action of polyanhydride nanovaccines.


Assuntos
Anticorpos/imunologia , Linfócitos B/imunologia , Centro Germinativo/imunologia , Nanopartículas/química , Polianidridos/química , Vacinas de Subunidades Antigênicas/imunologia , Animais , Anticorpos/sangue , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Nanotecnologia/métodos , Linfócitos T Auxiliares-Indutores/imunologia
3.
Nat Immunol ; 13(2): 188-95, 2011 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-22157630

RESUMO

Infection of erythrocytes with Plasmodium species induces clinical malaria. Parasite-specific CD4(+) T cells correlate with lower parasite burdens and severity of human malaria and are needed to control blood-stage infection in mice. However, the characteristics of CD4(+) T cells that determine protection or parasite persistence remain unknown. Here we show that infection of humans with Plasmodium falciparum resulted in higher expression of the inhibitory receptor PD-1 associated with T cell dysfunction. In vivo blockade of the PD-1 ligand PD-L1 and the inhibitory receptor LAG-3 restored CD4(+) T cell function, amplified the number of follicular helper T cells and germinal-center B cells and plasmablasts, enhanced protective antibodies and rapidly cleared blood-stage malaria in mice. Thus, chronic malaria drives specific T cell dysfunction, and proper function can be restored by inhibitory therapies to enhance parasite control.


Assuntos
Antígenos CD/efeitos dos fármacos , Antígeno B7-H1/antagonistas & inibidores , Linfócitos T CD4-Positivos/efeitos dos fármacos , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Doença Aguda , Animais , Antígenos CD/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/parasitologia , Antígeno B7-H1/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/parasitologia , Criança , Pré-Escolar , Doença Crônica , Eritrócitos/imunologia , Eritrócitos/parasitologia , Feminino , Centro Germinativo/efeitos dos fármacos , Centro Germinativo/imunologia , Centro Germinativo/parasitologia , Humanos , Malária Falciparum/imunologia , Mali , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium falciparum/imunologia , Estados Unidos , Regulação para Cima/efeitos dos fármacos , Proteína do Gene 3 de Ativação de Linfócitos
4.
Immunology ; 133(4): 452-68, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21635248

RESUMO

Germinal centre (GC) reactions are central features of T-cell-driven B-cell responses, and the site where antibody-producing cells and memory B cells are generated. Within GCs, a range of complex cellular and molecular events occur which are critical for the generation of high affinity antibodies. These processes require exquisite regulation not only to ensure the production of desired antibodies, but to minimize unwanted autoreactive or low affinity antibodies. To assess whether T regulatory (Treg) cells participate in the control of GC responses, immunized mice were treated with an anti-glucocorticoid-induced tumour necrosis factor receptor-related protein (GITR) monoclonal antibody (mAb) to disrupt Treg-cell activity. In anti-GITR-treated mice, the GC B-cell pool was significantly larger compared with control-treated animals, with switched GC B cells composing an abnormally high proportion of the response. Dysregulated GCs were also observed regardless of strain, T helper type 1 or 2 polarizing antigens, and were also seen after anti-CD25 mAb treatment. Within the spleens of immunized mice, CXCR5(+) and CCR7(-) Treg cells were documented by flow cytometry and Foxp3(+) cells were found within GCs using immunohistology. Final studies demonstrated administration of either anti-transforming growth factor-ß or anti-interleukin-10 receptor blocking mAb to likewise result in dysregulated GCs, suggesting that generation of inducible Treg cells is important in controlling the GC response. Taken together, these findings indicate that Treg cells contribute to the overall size and quality of the humoral response by controlling homeostasis within GCs.


Assuntos
Centro Germinativo/imunologia , Linfócitos T Reguladores/imunologia , Animais , Centro Germinativo/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T Reguladores/citologia
5.
Free Radic Biol Med ; 50(3): 448-58, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21130157

RESUMO

Reactive oxygen species (ROS) are critical in a broad spectrum of cellular processes including signaling, tumor progression, and innate immunity. The essential nature of ROS signaling in the immune systems of Drosophila and zebrafish has been demonstrated; however, the role of ROS, if any, in mammalian adaptive immune system development and function remains unknown. This work provides the first clear demonstration that thymus-specific elevation of mitochondrial superoxide (O(2)(•-)) disrupts normal T cell development and impairs the function of the mammalian adaptive immune system. To assess the effect of elevated mitochondrial superoxide in the developing thymus, we used a T-cell-specific knockout of manganese superoxide dismutase (i.e., SOD2) and have thus established a murine model to examine the role of mitochondrial superoxide in T cell development. Conditional loss of SOD2 led to increased superoxide, apoptosis, and developmental defects in the T cell population, resulting in immunodeficiency and susceptibility to the influenza A virus H1N1. This phenotype was rescued with mitochondrially targeted superoxide-scavenging drugs. These findings demonstrate that loss of regulated levels of mitochondrial superoxide lead to aberrant T cell development and function, and further suggest that manipulations of mitochondrial superoxide levels may significantly alter clinical outcomes resulting from viral infection.


Assuntos
Imunidade Adaptativa , Vírus da Influenza A Subtipo H1N1/fisiologia , Mitocôndrias/metabolismo , Superóxidos/metabolismo , Linfócitos T/metabolismo , Animais , Apoptose , Feminino , Técnicas de Inativação de Genes , Vírus da Influenza A Subtipo H1N1/imunologia , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/química , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Estresse Oxidativo , Superóxido Dismutase/deficiência , Superóxido Dismutase/genética , Superóxidos/farmacologia , Linfócitos T/imunologia , Linfócitos T/patologia
6.
Methods Mol Biol ; 447: 295-323, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18369926

RESUMO

Chronic alcohol abuse leads to multiple defects in the immune system, leading to an increased risk of infectious disease and malignancy. Immune lesions encompass both the innate and adaptive arms and include deficiencies in the B-cell compartment. Long-term alcoholics exhibit loss of B cells in the periphery and diminished ability to generate protective antibodies. To better mimic the chronic alcoholic patient, our group has used an ethanol-in-drinking-water mouse model. Mice consuming alcohol in this manner progressively develop a range of immune abnormalities, including defects in humoral immunity. To document and explore B-cell lesions in ethanol-consuming mice, our laboratory has used a broad panel of technologies. These include protocols to define the physical state of B cells in the bone marrow and periphery, in vitro approaches to test B-cell activation potential and in vivo experiments to document the humoral competence of the host. These key techniques are detailed in the present chapter.


Assuntos
Consumo de Bebidas Alcoólicas/imunologia , Alcoolismo/imunologia , Formação de Anticorpos , Linfócitos B/imunologia , Etanol/administração & dosagem , Ativação Linfocitária , Animais , Diferenciação Celular , Proliferação de Células , Separação Celular , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunização , Camundongos
7.
J Invest Dermatol ; 128(6): 1386-96, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18094731

RESUMO

Transdifferentiation has become a common claim for somatic stem cells, yet how such cells can be directed toward a specified cell lineage has not been well investigated. We previously demonstrated that when isolated epidermal stem cells were placed into an embryonic environment, their potential was extended beyond the keratinocyte lineage. Here, we present evidence that cells isolated using a modification of our published method for epidermal stem cells can be specifically directed to differentiate into B lymphocyte precursors. We found that these isolated cells co-cultured with S17 bone marrow stromal cells in cytokine-supplemented medium changed their cell surface marker profile and gene expression pattern to one characteristic of B lymphocyte precursors. Such cells also underwent variable, diversity, joining rearrangement at the immunoglobulin heavy-chain locus, a permanent genetic change unique to lymphocytes. This feature is limited to the cells isolated using the modified epidermal stem cell method, as cells isolated using the modified transit amplifying cell method could not be re-directed or reprogrammed. Such results demonstrate that cells from the epidermis can be directed to cross lineage boundaries to become mesodermally derived lymphocytes.


Assuntos
Linfócitos B/citologia , Benzimidazóis/farmacologia , Epiderme/patologia , Células-Tronco Hematopoéticas/citologia , Sistema Hematopoético , Animais , Linfócitos B/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Técnicas Citológicas , Células Epidérmicas , Células-Tronco Hematopoéticas/metabolismo , Cadeias Pesadas de Imunoglobulinas/química , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Células Estromais/citologia
8.
Bone ; 38(1): 41-7, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16122996

RESUMO

Chronic recurrent multifocal osteomyelitis (CRMO) is an autoinflammatory disorder that primarily affects bone but is often accompanied by inflammation of the skin and/or gastrointestinal tract. The etiology is unknown but evidence suggests a genetic component to disease susceptibility. Although most cases of CRMO are sporadic, there is an autosomal recessive syndromic form of the disease, called Majeed syndrome, which is due to homozygous mutations in LPIN2. In addition, there is a phenotypically similar mouse, called cmo (chronic multifocal osteomyelitis) in which the disease is inherited as an autosomal recessive disorder. The cmo locus has been mapped to murine chromosome 18. In this report, we describe phenotypic abnormalities in the cmo mouse that include bone, cartilage and skin inflammation. Utilizing a backcross breeding strategy, we refined the cmo locus to a 1.3 Mb region on murine chromosome 18. Within the refined region was the gene pstpip2, which shares significant sequence homology to the PSTPIP1. Mutations in PSTPIP1 have been shown to cause the autoinflammatory disorder PAPA syndrome (pyogenic arthritis, pyoderma gangrenosum and acne). Mutation analysis, utilizing direct sequencing, revealed a single base pair change c.293T --> C in the pstpip2 gene resulting in a highly conserved leucine at amino acid 98 being replaced by a proline (L98P). No other mutations were found in the coding sequence of the remaining genes in the refined interval, although a 50 kb gap remains unexplored. These data suggest that mutations in pstpip2 may be the genetic explanation for the autoinflammatory phenotype seen in the cmo mouse.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Doenças Autoimunes/genética , Proteínas do Citoesqueleto/genética , Inflamação/genética , Mutação de Sentido Incorreto , Osteomielite/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Mapeamento Cromossômico , Cromossomos de Mamíferos , Doença Crônica , Cruzamentos Genéticos , Proteínas do Citoesqueleto/química , Análise Mutacional de DNA , Éxons , Feminino , Marcadores Genéticos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Repetições de Microssatélites , Dados de Sequência Molecular , Prolina/metabolismo
9.
Blood ; 107(3): 1085-91, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16223771

RESUMO

Stat proteins are latent cytoplasmic transcription factors that are crucial in many aspects of mammalian development. In the immune system, Stat3 has distinct roles in T-cell, neutrophil, and macrophage function, but a role for Stat3 in B-cell development, particularly in the terminal differentiation of B cells into antibody-secreting plasma cells, has never been directly tested. In this study, we used the Cre/lox system to generate a mouse strain in which Stat3 was conditionally deleted in the B-cell lineage (Stat3(fl/fl)CD19(Cre/+)). B-cell development, establishment of the peripheral B-cell compartment, and baseline serum antibody levels were unperturbed in Stat3(fl/fl)CD19(Cre/+) mice. Strikingly, Stat3(fl/fl)CD19(Cre/+) mice displayed profound defects in T-dependent (TD) IgG responses, but normal TD IgM, IgE, and IgA responses and T-independent (TI) IgM and IgG3 responses. In addition, germinal center (GC) formation, isotype switching, and generation of memory B cells, including IgG+ memory cells, were all intact in Stat3(fl/fl)CD19(Cre/+) mice, indicating that the requirement for Stat3 was limited to plasma cell differentiation. These results demonstrate a profound yet highly selective role for Stat3 in TD IgG plasma cell differentiation, and therefore represent a unique example of a transcription factor regulating isotype-specific terminal B-cell differentiation.


Assuntos
Diferenciação Celular/imunologia , Isotipos de Imunoglobulinas/imunologia , Plasmócitos/imunologia , Fator de Transcrição STAT3/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD19/imunologia , Diferenciação Celular/genética , Integrases/genética , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Fator de Transcrição STAT3/deficiência , Proteínas Virais/genética
10.
Immunity ; 21(2): 255-66, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15308105

RESUMO

The oncogenic EBV protein LMP1 mimics a dysregulated CD40 receptor in vitro. To compare CD40 and LMP1-mediated events in vivo, transgenic mice were engineered to express mouse CD40 (mCD40tg) or a protein with extracellular mCD40 and cytoplasmic LMP1 (mCD40-LMP1tg). Transgenic and CD40(-/-) mice were bred so that only the transgenic CD40 molecule is expressed in B cells, macrophages, and dendritic cells. mCD40-LMP1tg mice had normal lymphocyte subsets, and immunization elicited an antibody response featuring normal isotype switching, affinity maturation, and germinal center (GC) formation. However, unimmunized mCD40-LMP1tg mice had expanded immature and germinal center B cells, produced autoantibodies, exhibited marked splenomegaly and lymphadenopathy, and elevated serum IL-6. Thus, signaling through the LMP1 cytoplasmic tail results in amplified and abnormal mimicry of CD40 functions in vivo, indicating possible ways in which LMP1 contributes to the pathogenesis of EBV-associated human disease.


Assuntos
Linfócitos B/metabolismo , Hipersensibilidade/metabolismo , Ativação Linfocitária/imunologia , Baço/citologia , Proteínas da Matriz Viral/metabolismo , Animais , Formação de Anticorpos , Autoimunidade/imunologia , Autoimunidade/fisiologia , Linfócitos B/imunologia , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Infecções por Vírus Epstein-Barr/etiologia , Infecções por Vírus Epstein-Barr/imunologia , Imunofluorescência , Hipersensibilidade/imunologia , Imunoglobulinas/sangue , Imunoglobulinas/imunologia , Interleucina-6/imunologia , Interleucina-6/metabolismo , Camundongos , Transdução de Sinais/imunologia , Baço/imunologia , Baço/metabolismo , Proteínas da Matriz Viral/imunologia
11.
Blood ; 100(13): 4557-64, 2002 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12393436

RESUMO

In allogeneic hematopoietic stem cell transplant recipients, restoration of humoral immunity is delayed and can remain impaired for years. In many severe combined immune deficiency (SCID) patients given haploidentical bone marrow (BM), lesions in humoral immunity are exacerbated by poor engraftment of donor B cells. The nature of these defects is important to understand as they render patients susceptible to infection. Previous work in mice suggested that in utero transplantation (IUT) of allogeneic BM might offer several advantages for the correction of primary immune deficiencies. In SCID mice given fully allogeneic BM in utero, the lymphoid compartment was restored with minimal evidence of graft-versus-host disease (GVHD). The present report examines B-cell reconstitution and function in mice that have received allogeneic IUT. Results are compared with those of adult mice given total body irradiation (TBI) followed by transplantation with allogeneic BM. In addition to enumerating the various B-cell subsets present in BM, spleen, and peritoneal cavity (PC), B-cell competence was assessed by challenging mice with T cell-independent (TI) and T cell-dependent (TD) antigens. The results demonstrated that all B-cell subsets in the BM and periphery were restored in allogeneic IUT and TBI mice, as were antibody responses after TI challenge. Upon immunization with TD antigens, however, IUT and TBI mice exhibited suboptimal responses as measured by the capacity to isotype switch and generate germinal center (GC) B cells. Thus, although allogeneic BM transplantation results in complete recovery of the B-cell compartment, certain elements of the humoral response remain defective.


Assuntos
Subpopulações de Linfócitos B/imunologia , Transplante de Medula Óssea , Imunodeficiência Combinada Severa/terapia , Animais , Animais Congênicos , Formação de Anticorpos , Feminino , Feto/imunologia , Doença Enxerto-Hospedeiro , Sistema Imunitário/embriologia , Sistema Imunitário/crescimento & desenvolvimento , Imunidade Celular , Imunização , Testes Imunológicos , Tecido Linfoide/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Gravidez , Quimera por Radiação , Imunodeficiência Combinada Severa/embriologia , Imunodeficiência Combinada Severa/imunologia , Transplante Homólogo/imunologia , Irradiação Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...