Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 18(8): 1906-1916, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28228257

RESUMO

Long-lived plasma cells (PCs) in the bone marrow (BM) are a critical source of antibodies after infection or vaccination, but questions remain about the factors that control PCs. We found that systemic infection alters the BM, greatly reducing PCs and regulatory T (Treg) cells, a population that contributes to immune privilege in the BM. The use of intravital imaging revealed that BM Treg cells display a distinct behavior characterized by sustained co-localization with PCs and CD11c-YFP+ cells. Gene expression profiling indicated that BM Treg cells express high levels of Treg effector molecules, and CTLA-4 deletion in these cells resulted in elevated PCs. Furthermore, preservation of Treg cells during systemic infection prevents PC loss, while Treg cell depletion in uninfected mice reduced PC populations. These studies suggest a role for Treg cells in PC biology and provide a potential target for the modulation of PCs during vaccine-induced humoral responses or autoimmunity.


Assuntos
Células da Medula Óssea/imunologia , Medula Óssea/imunologia , Plasmócitos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Autoimunidade/imunologia , Antígeno CTLA-4/imunologia , Imunidade Humoral , Imunofenotipagem/métodos , Camundongos , Camundongos Endogâmicos C57BL
2.
Semin Immunol ; 26(5): 421-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25443579

RESUMO

Complex interactions govern the interplay of central nervous and immune systems, including the generation, homeostatic maintenance, and activation of B cells. Accordingly, spinal cord injury will likely impact all of these processes. Several laboratories have recently explored this possibility, and their observations in aggregate reveal both acute and chronic consequences that can vary based on the injury location. Acute effects include a transient cessation of bone marrow B lymphopoiesis, with a corresponding drop in the peripheral follicular and transitional B cell subsets, whereas the marginal zone subset is preserved. Despite recovery of B lymphopoiesis by 28 days post injury, follicular B cell numbers remain depressed; this may reflect reduced levels of the homeostatic cytokine BLyS. In general, the ability to mount T dependent antibody responses after injury are intact, as are pre-existing memory B cell pools and antibody levels. In contrast, T-independent responses are chronically compromised. Both glucocorticoid-dependent and -independent processes mediate these effects, but a detailed understanding of the mechanisms involved awaits further study. Nonetheless, these observations in toto strengthen the growing appreciation for bidirectional interactions between the CNS and immune system, highlighting the need for further basic and translational efforts.


Assuntos
Subpopulações de Linfócitos B/imunologia , Sistema Nervoso Central/imunologia , Citocinas/metabolismo , Linfopoese/imunologia , Traumatismos da Medula Espinal/imunologia , Animais , Fator Ativador de Células B/biossíntese , Fator Ativador de Células B/imunologia , Subpopulações de Linfócitos B/patologia , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Citocinas/imunologia , Modelos Animais de Doenças , Humanos , Imunidade Humoral , Imunidade Inata , Ativação Linfocitária , Camundongos , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Linfócitos T/imunologia , Linfócitos T/patologia
3.
Cytokine Growth Factor Rev ; 25(2): 107-13, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24507939

RESUMO

BLyS family members govern selection and survival of cells in the pre-immune B cell compartment, and emerging evidence suggests similar roles in antigen-experienced B cell pools. We review the features of this family, with particular emphasis on recent findings of how BLyS influences affinity maturation in germinal centers, which lie at the intersection of the pre-immune and antigen-experienced B cell compartments. We propose a model whereby tolerogenic selection at the transitional stage and affinity maturation in the germinal center employ the same BLyS driven mechanism.


Assuntos
Apresentação de Antígeno/imunologia , Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Sobrevivência Celular/imunologia , Centro Germinativo/imunologia , Humanos , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
4.
J Exp Med ; 211(1): 45-56, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24367004

RESUMO

We have assessed the role of B lymphocyte stimulator (BLyS) and its receptors in the germinal center (GC) reaction and affinity maturation. Despite ample BLyS retention on B cells in follicular (FO) regions, the GC microenvironment lacks substantial BLyS. This reflects IL-21-mediated down-regulation of the BLyS receptor TACI (transmembrane activator and calcium modulator and cyclophilin ligand interactor) on GC B cells, thus limiting their capacity for BLyS binding and retention. Within the GC, FO helper T cells (TFH cells) provide a local source of BLyS. Whereas T cell-derived BLyS is dispensable for normal GC cellularity and somatic hypermutation, it is required for the efficient selection of high affinity GC B cell clones. These findings suggest that during affinity maturation, high affinity clones rely on TFH-derived BLyS for their persistence.


Assuntos
Formação de Anticorpos/imunologia , Fator Ativador de Células B/biossíntese , Linfócitos B/metabolismo , Regulação da Expressão Gênica/imunologia , Centro Germinativo/fisiologia , Linfócitos T Auxiliares-Indutores/metabolismo , Animais , Afinidade de Anticorpos , Linfócitos B/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Linfócitos T Auxiliares-Indutores/imunologia , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo
5.
Oncoimmunology ; 2(8): e25752, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24083082

RESUMO

The ability of Listeria monocytogenes-based anticancer vaccines to induce tumor regression depends on the responsiveness of malignant cells to interferon γ (IFNγ). Inhibition of IFNγ limits the recruitment of T cells to the tumors of vaccinated mice. We hypothesized that vaccination with immunotherapeutic L. monocytogenes induces the IFNγ-dependent production of chemokines that regulate the migration of tumor-infiltrating T cells. To gain further insights into this issue, we examined the chemokine responses of a transplantable, human papillomavirus (HPV)-immortalized murine tumor model (TC-1) following the administration of a L. monocytogenes-based immunotherapeutic agent that expresses E7 from HPV-16. Here, we report that the administration of L. monocytogenes-based anticancer vaccines increases the secretion of chemokine (C-X-C motif) ligand 9 (CXCL9), and CXCL10 by tumors, hence favoring the recruitment of T cells bearing the cognate chemokine (C-X-C motif) receptor 3 (CXCR3). Furthermore, the expression of CXCL9, but not CXCL10, in TC-1 tumors was significantly reduced upon anti-IFNγ antibody treatment. CXCL9 was highly expressed by TC-1 cells following the administration of IFNγ and tumor necrosis factor α (TNFα), in vitro. Moreover, the inhibition of CXCL9 in TC-1 cells reduced the proportion of CD8+ T cells infiltrating tumors in vaccinated mice, while increasing that of CD4+ T cells, thus altering T-cell subset distribution. We conclude that the administration of L. monocytogenes-based anticancer vaccines regulates TH1 chemokine responses and that malignant cells are an important source of these chemokines.

6.
J Infect Dis ; 206(1): 91-8, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22561364

RESUMO

BACKGROUND: Brucella spp. are intracellular bacteria that establish lifelong infections whose mechanisms of chronicity are poorly understood. Notably, B cells facilitate the establishment of the high infection plateau that persists for months. METHODS: We evaluated the contribution of murine B cells toward providing infection niches for Brucella by using flow cytometry and microscopy and by determining live bacterial counts associated with B cells both in vivo and in vitro. RESULTS: Herein we demonstrate that immunoglobulin M and complement-opsonized Brucella abortus infects and survives inside primary murine B cells protected from bactericidal effects of gentamicin. The entry was dependent on microfilaments for internalization and subsequently brucellae reside in a late endosomal/lysosomal compartment. Throughout the infection, 10% of colony-forming units from infected mice was associated with B cells, and these cells transferred disease to naive hosts. Furthermore, Brucella-positive cells were positive for transforming growth factor (TGF) ß1, and about 10% of such cells were B cells, similar to rates found for other intracellular pathogens that induce their hosts cells to produce TGF-ß1. CONCLUSIONS: To conclude, infected B cells contribute to chronic bacterial infections by providing an intracellular niche that may exert an immunoregulatory role. Although professional phagocytic cells harbor intracellular bacteria including Brucella, infection of lymphocytes by bacteria has not been previously appreciated.


Assuntos
Linfócitos B/imunologia , Linfócitos B/microbiologia , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/imunologia , Brucelose/imunologia , Brucelose/microbiologia , Animais , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/metabolismo , Linfócitos B/metabolismo , Brucella abortus/genética , Brucella abortus/metabolismo , Brucelose/genética , Brucelose/metabolismo , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Replicação do DNA/genética , Replicação do DNA/imunologia , Endossomos/genética , Endossomos/imunologia , Endossomos/metabolismo , Endossomos/microbiologia , Imunoglobulina M/imunologia , Imunoglobulina M/metabolismo , Lisossomos/genética , Lisossomos/imunologia , Lisossomos/metabolismo , Lisossomos/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/microbiologia , Fagocitose/genética , Fagocitose/imunologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Células-Tronco/microbiologia , Sobrevida , Fator de Crescimento Transformador beta1/imunologia , Fator de Crescimento Transformador beta1/metabolismo
7.
J Immunol ; 188(11): 5257-66, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22523388

RESUMO

Spinal cord injury (SCI) results in immune depression. To better understand how injury inhibits humoral immunity, the effects of chronic thoracic SCI on B cell development and immune responses to thymus-independent type 2 and thymus-dependent Ags were determined. Mice received complete crush injury or control laminectomy at either thoracic level 3, which disrupts descending autonomic control of the spleen, or at thoracic level 9, which conserves most splenic sympathetic activity. Although mature B cell numbers were only mildly reduced, bone marrow B cell production was transiently but profoundly depressed immediately after injury. Despite the return of normal B cell production 4 wk after SCI, mice receiving thoracic level 3 injury showed a significant reduction in their ability to mount primary thymus-independent type 2 or thymus-dependent immune responses. The latter were marked by decreases in germinal center B cells as well as class-switched high-affinity Ab-secreting cells. Importantly, injury did not affect affinity maturation per se, pre-existing B cell memory, or secondary humoral immune responses. Taken together, these findings show that chronic high thoracic SCI impairs the ability to mount optimal Ab responses to new antigenic challenges, but spares previously established humoral immunity.


Assuntos
Formação de Anticorpos/imunologia , Traumatismos da Medula Espinal/imunologia , Doença Aguda , Animais , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Subpopulações de Linfócitos B/patologia , Doença Crônica , Feminino , Contagem de Linfócitos , Linfopoese/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Baço/citologia , Baço/imunologia , Baço/patologia , Timo/citologia , Timo/imunologia , Timo/patologia
8.
Science ; 335(6066): 342-4, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22174128

RESUMO

Lifelong antibody responses to vaccination require reorganization of lymphoid tissue and dynamic intercellular communication called the germinal center reaction. B lymphocytes undergo cellular polarization during antigen stimulation, acquisition, and presentation, which are critical steps for initiating humoral immunity. Here, we show that germinal center B lymphocytes asymmetrically segregate the transcriptional regulator Bcl6, the receptor for interleukin-21, and the ancestral polarity protein atypical protein kinase C to one side of the plane of division, generating unequal inheritance of fate-altering molecules by daughter cells. Germinal center B lymphocytes from mice with a defect in leukocyte adhesion fail to divide asymmetrically. These results suggest that motile cells lacking constitutive attachment can receive provisional polarity cues from the microenvironment to generate daughter cell diversity and self-renewal.


Assuntos
Divisão Celular Assimétrica , Linfócitos B/citologia , Linfócitos B/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Animais , Linfócitos B/metabolismo , Antígenos CD40/metabolismo , Adesão Celular , Comunicação Celular , Polaridade Celular , Microambiente Celular , Sinais (Psicologia) , Proteínas de Ligação a DNA/metabolismo , Imunização , Camundongos , Camundongos Endogâmicos C57BL , Mitose , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6 , Receptores de Interleucina-21/metabolismo
9.
PLoS Pathog ; 7(6): e1002089, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21738467

RESUMO

African trypanosomes of the Trypanosoma brucei species are extracellular protozoan parasites that cause the deadly disease African trypanosomiasis in humans and contribute to the animal counterpart, Nagana. Trypanosome clearance from the bloodstream is mediated by antibodies specific for their Variant Surface Glycoprotein (VSG) coat antigens. However, T. brucei infection induces polyclonal B cell activation, B cell clonal exhaustion, sustained depletion of mature splenic Marginal Zone B (MZB) and Follicular B (FoB) cells, and destruction of the B-cell memory compartment. To determine how trypanosome infection compromises the humoral immune defense system we used a C57BL/6 T. brucei AnTat 1.1 mouse model and multicolor flow cytometry to document B cell development and maturation during infection. Our results show a more than 95% reduction in B cell precursor numbers from the CLP, pre-pro-B, pro-B, pre-B and immature B cell stages in the bone marrow. In the spleen, T. brucei induces extramedullary B lymphopoiesis as evidenced by significant increases in HSC-LMPP, CLP, pre-pro-B, pro-B and pre-B cell populations. However, final B cell maturation is abrogated by infection-induced apoptosis of transitional B cells of both the T1 and T2 populations which is not uniquely dependent on TNF-, Fas-, or prostaglandin-dependent death pathways. Results obtained from ex vivo co-cultures of living bloodstream form trypanosomes and splenocytes demonstrate that trypanosome surface coat-dependent contact with T1/2 B cells triggers their deletion. We conclude that infection-induced and possibly parasite-contact dependent deletion of transitional B cells prevents replenishment of mature B cell compartments during infection thus contributing to a loss of the host's capacity to sustain antibody responses against recurring parasitemic waves.


Assuntos
Apoptose , Linfopoese/imunologia , Células Precursoras de Linfócitos B/imunologia , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/imunologia , Glicoproteínas Variantes de Superfície de Trypanosoma/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Formação de Anticorpos/imunologia , Variação Antigênica/imunologia , Medula Óssea/imunologia , Células Cultivadas , Memória Imunológica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Linfócitos B/citologia , Baço/citologia , Baço/imunologia , Trypanosoma brucei brucei/metabolismo
10.
Proc Natl Acad Sci U S A ; 105(40): 15517-22, 2008 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-18832171

RESUMO

We have used an inhibiting antibody to determine whether preimmune versus antigen-experienced B cells differ in their requisites for BLyS, a cytokine that controls differentiation and survival. Whereas in vivo BLyS inhibition profoundly reduced naïve B cell numbers and primary immune responses, it had a markedly smaller effect on memory B cells and long-lived plasma cells, as well as secondary immune responses. There was heterogeneity within the memory pools, because IgM-bearing memory cells were sensitive to BLyS depletion whereas IgG-bearing memory cells were not, although both were more resistant than naïve cells. There was also heterogeneity within B1 pools, as splenic but not peritoneal B1 cells were diminished by anti-BLyS treatment, yet the number of natural antibody-secreting cells remained constant. Together, these findings show that memory B cells and natural antibody-secreting cells are BLyS-independent and suggest that these pools can be separately manipulated.


Assuntos
Formação de Anticorpos/imunologia , Fator Ativador de Células B/antagonistas & inibidores , Fator Ativador de Células B/fisiologia , Linfócitos B/imunologia , Imunidade Inata/imunologia , Animais , Fator Ativador de Células B/metabolismo , Linfócitos B/citologia , Feminino , Memória Imunológica/imunologia , Camundongos , Camundongos Endogâmicos C57BL
11.
Immunol Res ; 42(1-3): 75-83, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18668213

RESUMO

Lymphocyte homeostasis poses a multi-faceted biological puzzle, because steady pre-immune populations must be maintained at an acceptable steady state to yield effective protection, despite stringent selective events during their generation. In addition, activated, memory and both short- and long-term effectors must be governed by independent homeostatic mechanisms. Finally, advancing age is accompanied by substantial changes that impact the dynamics and behavior of these pools, leading to cumulative homeostatic perturbations and compensation. Our laboratory has focused on the over-arching role of BLyS family ligands and receptors in these processes. These studies have led to a conceptual framework within which distinct homeostatic niches are specified by BLyS receptor signatures, which define the BLyS family ligands that can afford survival. The cues for establishing these receptor signatures, as well as the downstream survival mechanisms involved, are integrated with cell extrinsic inputs via cross talk among downstream mediators. A refined understanding of these relationships should yield insight into the selection and maintenance of B cell subsets, as well as an appreciation of how homeostatic mechanisms may contribute to immunosenescence.


Assuntos
Subpopulações de Linfócitos B/citologia , Homeostase/fisiologia , Animais , Fator Ativador de Células B/fisiologia , Receptor do Fator Ativador de Células B/fisiologia , Antígeno de Maturação de Linfócitos B/fisiologia , Senescência Celular/fisiologia , Receptores de Antígenos de Linfócitos B/fisiologia , Proteína Transmembrana Ativadora e Interagente do CAML/fisiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
12.
Microbes Infect ; 9(1): 55-62, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17196866

RESUMO

The inner-membrane protein BacA affects Brucella LPS structure. A bacA deletion mutant of Brucella abortus, known as KL7 (bacA(mut)-KL7), is attenuated in BALB/c mice and protects against challenge. Thus, bacA mutation was a candidate for incorporation into live attenuated vaccines. We assessed bacA(mut)-KL7 in 2 additional mouse strains: the more resistant C57BL/6 that produces interferon-gamma throughout the infection and the highly susceptible interferon-gamma-deficient C57BL/6 in which brucellae exhibit continual exponential growth. While it was hypothesized that bacA(mut)-KL7 would exhibit even greater attenuation relative to its parent strain B. abortus 2308 in C57BL/6 mice than it did in BALB/c mice, this was not the case. Moreover, it was more pathogenic in C57BL/6 interferon-gamma-deficient mice than 2308 causing abscesses and wasting even though the splenic loads of bacA(mut)-KL7 were significantly lower. These 2 observations were correlated, respectively, with an ability of IFNgamma-activated macrophages to equivalently control strains 2308 and bacA(mut)-KL7 and the ability of bacA(mut)-KL7 organism and its LPS to induce greater amounts of pro-inflammatory cytokines than 2308. We conclude that attenuation properties of bacA mutation are dependent upon the nature of the host but more importantly that bacterial gene deletion can result in increased host pathology without an increase in bacterial load, crucial considerations for vaccine design.


Assuntos
Proteínas de Bactérias/genética , Brucella abortus/genética , Brucella abortus/imunologia , Brucelose/imunologia , Citocinas/biossíntese , Proteínas de Membrana Transportadoras/genética , Animais , Proteínas de Bactérias/imunologia , Vacina contra Brucelose/genética , Vacina contra Brucelose/imunologia , Brucella abortus/patogenicidade , Brucelose/microbiologia , Proteínas do Sistema Complemento/imunologia , Citocinas/imunologia , Deleção de Genes , Interferon gama/deficiência , Interferon gama/imunologia , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos , Macrófagos/imunologia , Proteínas de Membrana Transportadoras/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
13.
Crit Rev Immunol ; 26(5): 407-42, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17341186

RESUMO

Brucella spp. are intracellular gram-negative bacteria that include a number of virulent species that cause chronic infections in a variety of mammalian hosts. Human infections are proportional to the level of disease in domestic animals because humans are infected zoonotically after contact with infected animals or their products. The chronicity of infection results from the ability of some brucellae to survive reactive oxygen intermediate and nitric oxide killing in host phagocytes, following which they activate bacterial genes in response to the acidic phagosome environment, prevent phagolysosomal fusion by remodeling the intracellular compartment, and subsequently replicate intracellularly. The crucial component of immunity that results in survival of the host and thus maintenance of this chronic infective state is interferon-gamma (IFN-gamma). Production of IFN-gamma results from the ability of brucella components, including lipid A, to interact with Toll-like receptors for the production of IL-12 and TNF-alpha, although the regulatory cytokine IL-10 is also produced and decreases control of the infection. Although CD4 and CD8 T cells are clearly involved in the production of IFN-gamma, and CD8 T cells may be cytotoxic, a role for NK cells and cytotoxicity in protective immunity to brucellosis has not been substantiated experimentally. Moreover, antibodies have been shown to have a limited role in passive transfer studies.


Assuntos
Brucella/imunologia , Brucella/patogenicidade , Brucelose/imunologia , Brucelose/microbiologia , Animais , Anticorpos/imunologia , Brucelose/metabolismo , Brucelose/patologia , Doença Crônica , Humanos , Linfócitos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA