Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 7081, 2023 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-37925420

RESUMO

B cells play a central role in humoral immunity but also have antibody-independent functions. Studies to date have focused on B cells in blood and secondary lymphoid organs but whether B cells reside in non-lymphoid organs (NLO) in homeostasis is unknown. Here we identify, using intravenous labeling and parabiosis, a bona-fide tissue-resident B cell population in lung, liver, kidney and urinary bladder, a substantial proportion of which are B-1a cells. Tissue-resident B cells are present in neonatal tissues and also in germ-free mice NLOs, albeit in lower numbers than in specific pathogen-free mice and following co-housing with 'pet-store' mice. They spatially co-localise with macrophages and regulate their polarization and function, promoting an anti-inflammatory phenotype, in-part via interleukin-10 production, with effects on bacterial clearance during urinary tract infection. Thus, our data reveal a critical role for tissue-resident B cells in determining the homeostatic 'inflammatory set-point' of myeloid cells, with important consequences for tissue immunity.


Assuntos
Linfócitos B , Macrófagos , Camundongos , Animais , Anticorpos , Fígado , Pulmão
2.
Autoimmun Rev ; 22(2): 103236, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36436750

RESUMO

Approximately 5% of the world-wide population is affected by autoimmune diseases. Overall, autoimmune diseases are still difficult to treat, impose a high burden on patients, and have a significant economic impact. Like other complex diseases, e.g., cancer, autoimmune diseases develop over several years. Decisive steps in the development of autoimmune diseases are (i) the development of autoantigen-specific lymphocytes and (often) autoantibodies and (ii) potentially clinical disease manifestation at a later stage. However, not all healthy individuals with autoantibodies develop disease manifestations. Identifying autoantibody-positive healthy individuals and monitoring and inhibiting their switch to inflammatory autoimmune disease conditions are currently in their infancy. The switch from harmless to inflammatory autoantigen-specific T and B-cell and autoantibody responses seems to be the hallmark for the decisive factor in inflammatory autoimmune disease conditions. Accordingly, biomarkers allowing us to predict this progression would have a significant impact. Several factors, such as genetics and the environment, especially diet, smoking, exposure to pollutants, infections, stress, and shift work, might influence the progression from harmless to inflammatory autoimmune conditions. To inspire research directed at defining and ultimately targeting autoimmune predisease, here, we review published evidence underlying the progression from health to autoimmune predisease and ultimately to clinically manifest inflammatory autoimmune disease, addressing the following 3 questions: (i) what is the current status, (ii) what is missing, (iii) and what are the future perspectives for defining and modulating autoimmune predisease.


Assuntos
Doenças Autoimunes , Autoimunidade , Humanos , Doenças Autoimunes/etiologia , Autoanticorpos , Autoantígenos , Linfócitos
3.
Front Immunol ; 9: 258, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29520270

RESUMO

B-1 cells constitute a unique subpopulation of lymphocytes residing mainly in body cavities like the peritoneal cavity (PerC) but are also found in spleen and bone marrow (BM). As innate-like B cells, they mediate first line immune defense through low-affinity natural IgM (nIgM) antibodies. PerC B-1 cells can egress to the spleen and differentiate into nIgM antibody-secreting plasma cells that recognize conserved exogenous and endogenous cellular structures. Homing to and homeostasis within the PerC are regulated by the chemokine CXCL13 released by PerC macrophages and stroma cells. However, the exact mechanisms underlying the regulation of CXCL13 and B-1 homeostasis are not fully explored. B-1 cells play important roles in the inflammatory response to infection, autoimmunity, ischemia/reperfusion injury, obesity, and atherosclerosis. Remarkably, this list of inflammatory entities has a strong overlap with diseases that are regulated by complement suggesting a link between B-1 cells and the complement system. Interestingly, up to now, no data exist regarding the role of complement in B-1 cell biology. Here, we demonstrate for the first time that C5a regulates B-1 cell steady-state dynamics within the peritoneum, the spleen, and the BM. We found decreased B-1a cell numbers in the peritoneum and the spleen of C5aR1-/- mice associated with increased B1-a and B1-b numbers in the spleen and high serum titers of nIgM antibodies directed against phosphorylcholine and several pneumococcal polysaccharides. Similarly, peritoneal B-1a cells were decreased in the peritoneum and splenic B-1a and B-1b cells were increased in C5aR2-/- mice. The decrease in peritoneal B-1 cell numbers was associated with decreased peritoneal CXCL13 levels in C5aR1-/- and C5aR2-/- mice. In search for mechanisms, we found that combined TLR2 and IL-10 receptor activation in PerC macrophages induced strong CXCL13 production, which was significantly reduced in cells from C5aR1- and C5aR2-deficient mice and after combined C5aR-targeting. Such stimulation also induced marked local C5 production by PerC macrophages and C5a generation. Importantly, peritoneal in vivo administration of C5a increased CXCL13 production. Taken together, our findings suggest that local non-canonical C5 activation in PerC macrophages fuels CXCL13 production as a novel mechanism to control B-1 cell homeostasis.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Quimiocina CXCL13/metabolismo , Complemento C5a/metabolismo , Macrófagos/imunologia , Peritônio/imunologia , Baço/imunologia , Animais , Células Cultivadas , Citocinas/metabolismo , Homeostase , Humanos , Imunidade Inata , Imunoglobulina M/sangue , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor da Anafilatoxina C5a/genética , Receptores de Interleucina-10/metabolismo , Células Th1/imunologia
4.
J Autoimmun ; 39(3): 180-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22727274

RESUMO

Antibodies contribute to the pathogenesis of many chronic inflammatory diseases, including autoimmune disorders and allergies. They are secreted by proliferating plasmablasts, short-lived plasma cells and non-proliferating, long-lived memory plasma cells. Memory plasma cells refractory to immunosuppression are critical for the maintenance of both protective and pathogenic antibody titers. Here, we studied the response of plasma cells in spleen, bone marrow and inflamed kidneys of lupus-prone NZB/W mice to high-dose dexamethasone and/or cyclophosphamide. BrdU+, dividing plasmablasts and short-lived plasma cells in the spleen were depleted while BrdU- memory plasma cells survived. In contrast, all bone marrow plasma cells including anti-DNA secreting cells were refractory to both drugs. Unlike bone marrow and spleen, which showed a predominance of IgM-secreting plasma cells, inflamed kidneys mainly accommodated IgG-secreting plasma cells, including anti-DNA secreting cells, some of which survived the treatments. These results indicate that the bone marrow is the major site of memory plasma cells resistant to treatment with glucocorticoids and anti-proliferative drugs, and that inflamed tissues and secondary lymphoid organs can contribute to the autoreactive plasma cell memory. Therefore, new strategies targeting autoreactive plasma cell memory should be considered. This could be the key to finding a curative approach to the treatment of chronic inflammatory autoantibody-mediated diseases.


Assuntos
Medula Óssea/imunologia , Ciclofosfamida/uso terapêutico , Dexametasona/uso terapêutico , Imunossupressores/uso terapêutico , Nefrite Lúpica/tratamento farmacológico , Plasmócitos/imunologia , Animais , Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Autoimunidade/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Bromodesoxiuridina/administração & dosagem , Ciclofosfamida/farmacologia , DNA/imunologia , Dexametasona/farmacologia , Modelos Animais de Doenças , Feminino , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Imunoglobulina M/biossíntese , Imunoglobulina M/imunologia , Memória Imunológica/efeitos dos fármacos , Imunossupressores/farmacologia , Rim/efeitos dos fármacos , Rim/imunologia , Rim/patologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Camundongos , Camundongos Endogâmicos NZB , Especificidade de Órgãos , Plasmócitos/efeitos dos fármacos , Plasmócitos/patologia , Baço/efeitos dos fármacos , Baço/imunologia , Baço/patologia
6.
J Am Soc Nephrol ; 22(2): 336-48, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21233415

RESUMO

Anti-neutrophil cytoplasmic autoantibodies (ANCA) cause vasculitis and necrotizing crescentic glomerulonephritis (NCGN). Steroids and cytotoxic drugs reduce mortality but can cause significant adverse events. The proteasome inhibitor bortezomib (BTZ) prevents glomerulonephritis in mouse models of lupus but its efficacy in ANCA-associated glomerulonephritis is unknown. We induced anti-MPO IgG-mediated NCGN by transplanting wild-type bone marrow (BM) into irradiated MPO-deficient mice immunized with MPO. Four weeks after BM transplantation, we treated mice with steroid/cyclophosphamide (S/CYC) or BTZ. Compared with untreated control mice, both S/CYC and BTZ significantly reduced urine abnormalities, NCGN, and infiltration of neutrophils and macrophages. Response to BTZ depended on timing of administration: BTZ abrogated NCGN if begun 3 weeks, but not 5 weeks, after BM transplantation. BTZ treatment significantly reduced total and MPO-specific plasma cells in both the spleen and bone marrow, resulting in significantly reduced anti-MPO titers. Furthermore, BTZ affected neither B cells nor total CD4 and CD8 T cells, including their naive and effector subsets. In contrast, S/CYC reduced the total number of cells in the spleen, including total and MPO-specific plasma cells and B cells. In contrast to BTZ, S/CYC did not affect total and MPO-specific plasma cells in the bone marrow. Three of 23 BTZ-treated mice died within 36 hours after BTZ administration. In summary, BTZ depletes MPO-specific plasma cells, reduces anti-MPO titers, and prevents NCGN in mice.


Assuntos
Anticorpos Anticitoplasma de Neutrófilos/imunologia , Ácidos Borônicos/farmacologia , Glomerulonefrite/prevenção & controle , Peroxidase/imunologia , Plasmócitos/fisiologia , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Animais , Autoanticorpos/sangue , Células da Medula Óssea/fisiologia , Ácidos Borônicos/toxicidade , Bortezomib , ELISPOT , Glomerulonefrite/etiologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Pirazinas/toxicidade , Baço/imunologia
8.
Blood ; 116(11): 1867-75, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20538807

RESUMO

Long-lived plasma cells in the bone marrow produce memory antibodies that provide immune protection persisting for decades after infection or vaccination but can also contribute to autoimmune and allergic diseases. However, the composition of the microenvironmental niches that are important for the generation and maintenance of these cells is only poorly understood. Here, we demonstrate that, within the bone marrow, plasma cells interact with the platelet precursors (megakaryocytes), which produce the prominent plasma cell survival factors APRIL (a proliferation-inducing ligand) and IL-6 (interleukin-6). Accordingly, reduced numbers of immature and mature plasma cells are found in the bone marrow of mice deficient for the thrombopoietin receptor (c-mpl) that show impaired megakaryopoiesis. After immunization, accumulation of antigen-specific plasma cells in the bone marrow is disturbed in these mice. Vice versa, injection of thrombopoietin allows the accumulation and persistence of a larger number of plasma cells generated in the course of a specific immune response in wild-type mice. These results demonstrate that megakaryocytes constitute an important component of the niche for long-lived plasma cells in the bone marrow.


Assuntos
Células da Medula Óssea/metabolismo , Megacariócitos/metabolismo , Plasmócitos/metabolismo , Nicho de Células-Tronco/metabolismo , Animais , Células da Medula Óssea/citologia , Comunicação Celular/efeitos dos fármacos , Contagem de Células , Células Cultivadas , Feminino , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Ovalbumina/imunologia , Ovalbumina/farmacologia , Plasmócitos/citologia , Plasmócitos/efeitos dos fármacos , Receptores de Trombopoetina/genética , Receptores de Trombopoetina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nicho de Células-Tronco/citologia , Trombopoetina/farmacologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
9.
J Allergy Clin Immunol ; 124(4): 819-26.e4, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19815119

RESUMO

BACKGROUND: Allergen-specific IgE antibodies are responsible for the pathogenesis of type I hypersensitivity. In patients with allergy, IgE titers can persist in the apparent absence of allergen for years. Seasonal allergen exposure triggers clinical symptoms and enhances allergen-specific IgE. Whether allergen-specific plasma cells originating from seasonal allergen exposures can survive and become long-lived is so far unclear. OBJECTIVE: We analyzed the localization and lifetimes of allergen-specific IgE-secreting, IgA-secreting, and IgG(1)-secreting plasma cells after allergen inhalation in an ovalbumin-induced murine model of allergic asthma. METHODS: Ovalbumin-specific IgG(1)-secreting, IgA-secreting, and IgE-secreting cells in lungs, spleen, and bone marrow were isolated and tested for antibody secretion by the ELISpot technique. Longevity of ovalbumin-specific plasma cells was determined by cyclophosphamide treatment, which depletes proliferating plasmablasts but leaves plasma cells untouched. Ovalbumin aerosol-induced infiltrates in lungs were localized by confocal microscopy. RESULTS: Long-lived ovalbumin-specific plasma cells were generated by systemic sensitization and survived in bone marrow and spleen, maintaining systemic ovalbumin-specific titers of IgG, IgA, and IgE. On inhalation of ovalbumin-containing aerosol, sensitized mice developed airway inflammation and more ovalbumin-specific IgG(1)-secreting, IgA-secreting, and IgE-secreting cells in the lungs and in secondary lymphoid organs. These plasma cells joined the pool of ovalbumin-specific plasma cells in the bone marrow and became long-lived-that is, they are resistant to cyclophosphamide. Termination of ovalbumin inhalation depleted ovalbumin-specific plasma cells from the lungs, but they persisted in spleen and bone marrow. CONCLUSION: Our results show that inhalation of aerosolized allergen generates long-lived, allergen-specific IgG(1)-secreting, IgA-secreting, and IgE-secreting plasma cells that survive cytostatic treatment.


Assuntos
Alérgenos/imunologia , Asma/imunologia , Imunoglobulina E/sangue , Plasmócitos/imunologia , Animais , Asma/metabolismo , Medula Óssea/imunologia , Medula Óssea/metabolismo , Ciclofosfamida/farmacologia , Modelos Animais de Doenças , Feminino , Imunidade nas Mucosas , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunossupressores/farmacologia , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Plasmócitos/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo
10.
J Immunol ; 182(4): 2113-23, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19201864

RESUMO

IL-6 and APRIL influence the growth, differentiation, and survival of normal and neoplastic Ab-forming cells (AFC). In this study, we identify two subsets of myeloid cells that associate with the AFC and are the main producers of these factors during a T-dependent Ab response to alum-precipitated protein in mouse lymph nodes. First CD11c(+)CD8alpha(-) dendritic cells located in the perivascular area of the T zone provide about half of the IL-6 mRNA produced in the node together with significant amounts of APRIL mRNA. The number of these cells increases during the response, at least in part due to local proliferation. The second subset comprises Gr1(+)CD11b(+)F4/80(+) monocyte/macrophages. These colonize the medullary cords during the response and are the other main IL-6 mRNA producers and the greatest source of APRIL mRNA. This medullary cord monocyte/macrophage subset results in local increase of APRIL mRNA that mirrors the polarity of CXCL12 expression in the node. The distribution of these myeloid cell subsets correlates with a gradient of AFC maturation assessed by progressive loss of Ki67 as AFC pass from the B cell follicle along the perivascular areas to the medullary cords.


Assuntos
Células Dendríticas/citologia , Interleucina-6/imunologia , Leucócitos Mononucleares/citologia , Linfonodos/citologia , Plasmócitos/citologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Transferência Adotiva , Animais , Diferenciação Celular/imunologia , Proliferação de Células , Células Dendríticas/imunologia , Citometria de Fluxo , Leucócitos Mononucleares/imunologia , Linfonodos/imunologia , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microdissecção , Microscopia Confocal , Células Mieloides/citologia , Células Mieloides/imunologia , Plasmócitos/imunologia , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Nat Med ; 14(7): 748-55, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18542049

RESUMO

Autoantibody-mediated diseases like myasthenia gravis, autoimmune hemolytic anemia and systemic lupus erythematosus represent a therapeutic challenge. In particular, long-lived plasma cells producing autoantibodies resist current therapeutic and experimental approaches. Recently, we showed that the sensitivity of myeloma cells toward proteasome inhibitors directly correlates with their immunoglobulin synthesis rates. Therefore, we hypothesized that normal plasma cells are also hypersensitive to proteasome inhibition owing to their extremely high amount of protein biosynthesis. Here we show that the proteasome inhibitor bortezomib, which is approved for the treatment of multiple myeloma, eliminates both short- and long-lived plasma cells by activation of the terminal unfolded protein response. Treatment with bortezomib depleted plasma cells producing antibodies to double-stranded DNA, eliminated autoantibody production, ameliorated glomerulonephritis and prolonged survival of two mouse strains with lupus-like disease, NZB/W F1 and MRL/lpr mice. Hence, the elimination of autoreactive plasma cells by proteasome inhibitors might represent a new treatment strategy for antibody-mediated diseases.


Assuntos
Ácidos Borônicos/farmacologia , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Nefrite/prevenção & controle , Plasmócitos/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Inibidores de Proteassoma , Pirazinas/farmacologia , Animais , Bortezomib , Lúpus Eritematoso Sistêmico/enzimologia , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos MRL lpr , Camundongos Endogâmicos NZB , Modelos Imunológicos , Nefrite/imunologia , Ovalbumina , Fatores de Tempo
12.
Nat Immunol ; 8(4): 419-29, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17322888

RESUMO

The survival of long-lived plasma cells, which produce most serum immunoglobulin, is central to humoral immunity. We found here that the inhibitory Fc receptor FcgammaRIIb was expressed on plasma cells and controlled their persistence in the bone marrow. Crosslinking FcgammaRIIb induced apoptosis of plasma cells, which we propose contributes to the control of their homeostasis and suggests a method for therapeutic deletion. Plasma cells from mice prone to systemic lupus erythematosus did not express FcgammaRIIb and were protected from apoptosis. Human plasmablasts expressed FcgammaRIIb and were killed by crosslinking, as were FcgammaRIIb-expressing myeloma cells. Our results suggest that FcgammaRIIb controls bone marrow plasma cell persistence and that defects in it may contribute to autoantibody production.


Assuntos
Apoptose/imunologia , Células da Medula Óssea/imunologia , Imunoglobulina G/imunologia , Plasmócitos/imunologia , Receptores de IgG/imunologia , Animais , Formação de Anticorpos/imunologia , Células da Medula Óssea/citologia , Humanos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Camundongos Endogâmicos NOD , Camundongos Endogâmicos NZB , Camundongos Knockout , Camundongos Transgênicos , Mieloma Múltiplo/imunologia , Plasmócitos/citologia , RNA/química , RNA/genética , Receptores de IgG/biossíntese , Receptores de IgG/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica/imunologia
13.
Mol Immunol ; 44(7): 1606-12, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17067679

RESUMO

The differentiation of B lymphocytes in the bone marrow is guided by the surrounding microenvironment determined by cytokines, adhesion molecules, and the extracellular matrix. These microenvironmental factors are mainly provided by stromal cells. In this paper, we report the identification of a VCAM-1-positive stromal cell population by flow cytometry. This population showed the expression of cell surface markers known to be present on stromal cells (CD10, CD13, CD90, CD105) and had a fibroblastoid phenotype in vitro. Single cell RT-PCR analysis of its cytokine expression pattern revealed transcripts for haematopoietic cytokines important for either the early B lymphopoiesis like flt3L or the survival of long-lived plasma cells like BAFF or both processes like SDF-1. Whereas SDF-1 transcripts were detectable in all VCAM-1-positive cells, flt3L and BAFF were only expressed by some cells suggesting the putative existence of different subpopulations with distinct functional properties. In summary, the VCAM-1-positive cell population seems to be a candidate stromal cell population supporting either developing B cells and/or long-lived plasma cells in human bone marrow.


Assuntos
Linfócitos B/imunologia , Células da Medula Óssea/imunologia , Citocinas/genética , Células-Tronco Hematopoéticas/citologia , Plasmócitos/citologia , Molécula 1 de Adesão de Célula Vascular/análise , Fator Ativador de Células B/genética , Linfócitos B/citologia , Células da Medula Óssea/citologia , Linhagem da Célula , Quimiocina CXCL12 , Quimiocinas CXC/genética , Humanos , Proteínas de Membrana/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Células Estromais/citologia , Células Estromais/imunologia , Transcrição Gênica
14.
Blood ; 106(10): 3609-17, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16081689

RESUMO

Mutation of p53 is a rare event in multiple myeloma, but it is unknown if p53 signaling is functional in myeloma cells, and if targeted nongenotoxic activation of the p53 pathway is sufficient to kill tumor cells. Here, we demonstrate that treatment of primary tumor samples with a small-molecule inhibitor of the p53-murine double minute 2 (MDM2) interaction increases the level of p53 and induces p53 targets and apoptotic cell death. Significantly, given the importance of the bone marrow microenvironment for the support and drug resistance of myeloma cells, tumor cells undergo effective apoptosis also in the presence of stromal cells, which themselves appear to tolerate exposure to nutlin-3. The in vitro toxicity of nutlin-3 was similar to that of the genotoxic drug melphalan. Because nutlin-mediated p53 activation is not dependent on DNA damage, MDM2 antagonists may help to avoid or reduce the severe genotoxic side effects of chemotherapeutic agents currently used to treat multiple myeloma. Therefore, MDM2 antagonists may offer a new treatment option for this disease.


Assuntos
Apoptose/efeitos dos fármacos , Imidazóis/farmacologia , Mieloma Múltiplo/metabolismo , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Animais , Antineoplásicos Alquilantes/efeitos adversos , Antineoplásicos Alquilantes/farmacologia , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Humanos , Imidazóis/uso terapêutico , Melfalan/efeitos adversos , Melfalan/farmacologia , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mutagênicos/farmacologia , Piperazinas/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Células Estromais/metabolismo , Proteína Supressora de Tumor p53/genética
15.
Blood ; 105(10): 3965-71, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15687242

RESUMO

C-X-C motif chemokine receptor 3 (CXCR3) and CXCR4 expressed on immunoglobulin G (IgG)-plasma-cell precursors formed in memory immune responses are crucial modulators of the homing of these cells. Here, we studied the regulation of the expression of these chemokine receptors during the differentiation of human memory B cells into plasma cells. We show that CXCR3 is absent on CD27- naive B cells but is expressed on a fraction of memory B cells, preferentially on those coexpressing IgG1. On differentiation into plasma-cell precursors, CXCR3+ memory B cells maintain the expression of this chemokine receptor. CXCR3- memory B cells up-regulate CXCR3 and migrate toward concentration gradients of its ligands only when costimulated with interferon gamma (IFN-gamma), but not interleukin 4 (IL-4), IL-1beta, IL-6, IFN-alpha, IFN-beta, or tumor necrosis factor alpha (TNF-alpha). In contrast, the differentiation of CXCR4- B cells into plasma cells is generally accompanied by the induction of CXCR4 expression. These results show that lack of CXCR4 expression on plasma-cell precursors is not a limiting factor for plasma-cell homing and that the expression of CXCR3 on memory B cells and plasma-cell precursors is induced by IFN-gamma, provided in human T helper type 1 (Th1)-biased immune responses. Once induced in memory B cells, CXCR3 expression remains part of the individual cellular memory.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Diferenciação Celular , Regulação da Expressão Gênica , Memória Imunológica , Receptores CXCR4/metabolismo , Receptores de Quimiocinas/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Movimento Celular , Células Cultivadas , Quimiocina CXCL9 , Quimiocinas CXC/imunologia , Quimiocinas CXC/metabolismo , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Memória Imunológica/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interferon gama/imunologia , Interferon gama/farmacologia , Receptores CXCR3 , Receptores CXCR4/imunologia , Receptores de Quimiocinas/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Regulação para Cima/efeitos dos fármacos
16.
J Leukoc Biol ; 75(6): 1022-8, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15020647

RESUMO

Despite the important role immunoglobulin G (IgG)-secreting plasma cells play in memory immune responses, the differentiation and homeostasis of these cells are not completely understood. Here, we studied the differentiation of human IgG-secreting cells ex vivo and in vitro, identifying these cells by the cellular affinity matrix technology. Several subpopulations of IgG-secreting cells were identified among the cells isolated from tonsils and bone marrow, particularly differing in the expression levels of CD9, CD19, and CD38. CD38 low IgG-secreting cells were present exclusively in the tonsils. A major fraction of these cells appeared to be early plasma cell precursors, as upon activation of B cells in vitro, IgG secretion preceded up-regulation of CD38, and on tonsillar sections, IgG-containing, CD38 low cells with a plasmacytoid phenotype were found in follicles, where plasma cell differentiation starts. A unitary phenotype of migratory peripheral blood IgG-secreting cells suggests that all bone marrow plasma cell populations share a common precursor cell. These data are compatible with a multistep model for plasma cell differentiation and imply that a common CD38 low IgG-secreting precursor gives rise to a diverse plasma cell compartment.


Assuntos
ADP-Ribosil Ciclase/metabolismo , Antígenos CD/metabolismo , Medula Óssea/imunologia , Imunoglobulina G/metabolismo , Tonsila Palatina/imunologia , Plasmócitos/imunologia , ADP-Ribosil Ciclase 1 , Antígenos CD19/metabolismo , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Diferenciação Celular , Movimento Celular , Humanos , Técnicas In Vitro , Glicoproteínas de Membrana/metabolismo , Tonsila Palatina/citologia , Fenótipo , Plasmócitos/metabolismo , Tetraspanina 29
17.
J Immunol ; 171(4): 1684-90, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12902466

RESUMO

Recent results suggest that plasma cell longevity is not an intrinsic capacity, but depends on yet unknown factors produced in their environment. In this study, we show that the cytokines IL-5, IL-6, TNF-alpha, and stromal cell-derived factor-1alpha as well as signaling via CD44 support the survival of isolated bone marrow plasma cells. The cytokines IL-7 and stem cell factor, crucially important for early B cell development, do not mediate plasma cell survival, indicating that plasma cells and early B cells have different survival requirements. As shown in IL-6-deficient mice, IL-6 is required for a normal induction, but not for the maintenance of plasma cell responses in vivo, indicating that the effects of individual survival factors are redundant. Optimal survival of isolated plasma cells requires stimulation by a combination of factors acting synergistically. These results strongly support the concept that plasma cell survival depends on niches in which a combination of specific signals, including IL-5, IL-6, stromal cell-derived factor-1alpha, TNF-alpha, and ligands for CD44, provides an environment required to mediate plasma cell longevity.


Assuntos
Citocinas/fisiologia , Plasmócitos/citologia , Plasmócitos/fisiologia , Transdução de Sinais/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Formação de Anticorpos/genética , Apoptose/genética , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Adesão Celular/fisiologia , Separação Celular , Sobrevivência Celular/fisiologia , Células Cultivadas , Sinergismo Farmacológico , Regulação da Expressão Gênica/fisiologia , Interleucina-5/deficiência , Interleucina-5/genética , Interleucina-6/deficiência , Interleucina-6/genética , Interleucina-6/imunologia , Interleucina-6/fisiologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmócitos/imunologia , Plasmócitos/metabolismo
18.
Curr Opin Immunol ; 14(4): 517-21, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12088688

RESUMO

A selected fraction of plasmablasts enters the compartment of nondividing, long-lived plasma cells to maintain humoral antibody memory. In accord with a current model for lymphocyte homeostasis, the lifetime of long-lived plasma cells is probably regulated by competition for a limited number of survival niches present in splenic red pulp, bone marrow and inflamed tissue. Plasma cells secreting autoantibodies specific for some, but not all, self-antigens are probably 'allowed' to enter the compartment of long-lived plasma cells and provide antibody-mediated 'autoimmune memory' that is resistant to conventional therapies.


Assuntos
Formação de Anticorpos , Memória Imunológica , Plasmócitos/imunologia , Animais , Autoanticorpos/imunologia , Doenças Autoimunes/imunologia , Medula Óssea/imunologia , Movimento Celular , Sobrevivência Celular , Homeostase , Inflamação/imunologia , Camundongos , Modelos Imunológicos , Baço/imunologia
19.
Eur J Immunol ; 32(4): 923-7, 2002 04.
Artigo em Inglês | MEDLINE | ID: mdl-11920557

RESUMO

Antigen-specific serum antibodies are protective for long periods of time 1, 2. These serum antibodies, the "humoral memory", are secreted by plasma cells derived from activated, antigen-specific B lymphocytes. Given their crucial role in immunity, surprisingly little is known about the biology of plasma cells. One of the fundamental questions is whether persisting protective serum antibody responses are maintained by long-lived plasma cells, or by short-lived plasma cells generated continuously from activated memory B cells. Here, we review some recent experiments suggesting that plasma cells have the capacity to live for unlimited time if rescued by specific factors provided in a limited number of survival niches in the body.


Assuntos
Formação de Anticorpos , Plasmócitos/citologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Células da Medula Óssea/citologia , Senescência Celular , Humanos , Memória Imunológica , Camundongos , Plasmócitos/imunologia , Baço/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA