Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Cancer Res Clin Oncol ; 147(6): 1685-1697, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33738544

RESUMO

BACKGROUND/AIMS: A proliferation-inducing ligand (APRIL, also known as TNFSF13, CD256) is a member of the tumor necrosis factor (TNF) superfamily and involved in a diverse set of diseases. In this work, we explored the potential associations and underlying mechanism in patients suffered from gastric cancer between the expression of APRIL and H. pylori infection. METHODS: We analyzed APRIL expression levels in 200 GC tissue samples by immunohistochemistry staining. H. pylori infection was detected by modified Giemsa staining. The biological effects of APRIL on human GC cells in vitro and in vivo were tested by CCK-8 assay, colony formation, flow cytometry detection, transwell migration assay, matrigel invasion assay, and tumor xenograft assay in animals. RESULTS: APRIL reactivity was positively correlated with H. pylori infection in vitro and vivo. It turned out that the decrease of miR-145 expression was dose-dependent and time-dependent on H. pylori infection and in consistent with APRIL expression. MiR-145 significantly attenuated the effect of H. pylori infection on APRIL gene expression in SGC7901 and BGC823 cell lines. Furthermore, APRIL overexpression promoted the proliferation, migration, invasion, and transfer of GC cells and decreased apoptosis, while APRIL knockdown suppressed these effects. We confirmed that APRIL activated the canonical NF-κB pathway through phosphorylation of AKT. CONCLUSION: The expression of APRIL, which promoted the proliferation, migration, invasion, viability, and metastasis of GC cells, was upregulated in human H. pylori-infected GC through miR-145. Besides, APRIL-induced gastric tumorigenicity via activating NF-κB pathway. These results may provide a framework for the deeper analysis of APRIL in GC risk and prognosis.


Assuntos
Adenocarcinoma/genética , Helicobacter pylori/fisiologia , Neoplasias Gástricas/genética , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Adenocarcinoma/patologia , Adenocarcinoma/virologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transformação Celular Viral/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Infecções por Helicobacter/complicações , Infecções por Helicobacter/genética , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Gástricas/patologia , Neoplasias Gástricas/virologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
2.
Arterioscler Thromb Vasc Biol ; 39(9): 1705-1714, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31315439

RESUMO

Atherosclerosis is a lipid-driven chronic inflammatory disease that is modulated by many immune cell subsets, including B cells. Therefore, targeting the inflammatory component of cardiovascular disease represents a promising therapeutic strategy. In the past years, immunotherapy has revolutionized the treatment of autoimmunity and cancer. Many of these clinically used strategies target B cells. Given the multifaceted role of B cells in atherogenesis, it is conceivable that B-cell-directed therapies can modulate disease development. Here, we review clinically available B-cell-targeted therapies and the possible benefits or detrimental effects on cardiovascular disease.


Assuntos
Linfócitos B/efeitos dos fármacos , Doenças Cardiovasculares/tratamento farmacológico , Aneurisma Aórtico/tratamento farmacológico , Aterosclerose/tratamento farmacológico , Receptor do Fator Ativador de Células B/antagonistas & inibidores , Receptor do Fator Ativador de Células B/fisiologia , Linfócitos B/fisiologia , Doenças Cardiovasculares/imunologia , Humanos , Imunização , Depleção Linfocítica , Receptores de Antígenos de Linfócitos B/antagonistas & inibidores , Receptores de Antígenos de Linfócitos B/fisiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
3.
Tumour Biol ; 37(9): 11573-11588, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27317256

RESUMO

Syndecans (SDC, SYND) comprise a group of four structurally related type 1 transmembrane heparan sulfate proteoglycans (HSPGs) that play important roles in tumorigenic processes. SDCs exert signaling via their protein cores and their conserved transmembrane and cytoplasmic domains or by forming complexes with growth factors (GFs). In classical Hodgkin's lymphoma (cHL), a lymphoid neoplasm of predominantly B cell origin, SDC1 and SDC4 are the active SDCs, and a number of GF (vascular endothelial growth factor, fibroblast growth factor, etc.) signaling pathways have been studied. However, despite extensive pre-clinical and clinical research on SDC-mediated GF signaling in many cancer types, there is very limited data for this interaction in cHL. Thus, this review highlights the relevant literature focusing on the potential interactions of SDCs and GFs in cHL pathogenesis. Also discussed are the pre-clinical and clinical studies targeting signaling through these pathways.


Assuntos
Doença de Hodgkin/etiologia , Transdução de Sinais/fisiologia , Sindecana-1/fisiologia , Animais , Humanos , Lisofosfolipídeos/fisiologia , Neovascularização Fisiológica , Receptor IGF Tipo 1/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Esfingosina/análogos & derivados , Esfingosina/fisiologia , Sindecana-1/análise , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
4.
J Am Soc Nephrol ; 27(11): 3430-3439, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27068226

RESUMO

TNF superfamily member 13 (TNFSF13) has been identified as a susceptibility gene for IgA nephropathy in recent genetic studies. However, the role of TNFSF13 in the progression of IgA nephropathy remains unresolved. We evaluated two genetic polymorphisms (rs11552708 and rs3803800) and plasma levels of TNFSF13 in 637 patients with IgA nephropathy, and determined the risk of ESRD according to theses variable. Neither of the examined genetic polymorphisms associated with a clinical outcome of IgA nephropathy. However, high plasma levels of TNFSF13 increased the risk of ESRD. To explore the causal relationship and underlying mechanism, we treated B cells from patients (n=21) with or without recombinant human TNFSF13 (rhTNFSF13) and measured the expression of IgA and galactose-deficient IgA (GdIgA) using ELISA and flow cytometry. Treatment with rhTNFSF13 significantly increased the total IgA level among B cells, and TNFSF13 receptor blockade abrogated this increase. Furthermore, the absolute levels of GdIgA increased with rhTNFSF13 treatment, but the total IgA-normalized levels did not change. Both RNA sequencing and quantitative PCR results showed that rhTNFSF13 did not alter the expression of glycosyltransferase enzymes. These results suggest that high plasma TNFSF13 levels associate with a worse prognosis of IgA nephropathy through the relative increase in GdIgA levels.


Assuntos
Glomerulonefrite por IGA/complicações , Falência Renal Crônica/etiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Adulto , Linfócitos B/fisiologia , Progressão da Doença , Feminino , Humanos , Masculino , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/sangue
5.
Blood ; 127(25): 3225-36, 2016 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-27127303

RESUMO

Here we show that overexpression or activation of B-cell maturation antigen (BCMA) by its ligand, a proliferation-inducing ligand (APRIL), promotes human multiple myeloma (MM) progression in vivo. BCMA downregulation strongly decreases viability and MM colony formation; conversely, BCMA overexpression augments MM cell growth and survival via induction of protein kinase B (AKT), MAPK, and nuclear factor (NF)-κB signaling cascades. Importantly, BCMA promotes in vivo growth of xenografted MM cells harboring p53 mutation in mice. BCMA-overexpressing tumors exhibit significantly increased CD31/microvessel density and vascular endothelial growth factor compared with paired control tumors. These tumors also express increased transcripts crucial for osteoclast activation, adhesion, and angiogenesis/metastasis, as well as genes mediating immune inhibition including programmed death ligand 1, transforming growth factor ß, and interleukin 10. These target genes are consistently induced by paracrine APRIL binding to BCMA on MM cells, which is blocked by an antagonistic anti-APRIL monoclonal antibody hAPRIL01A (01A). 01A is cytotoxic against MM cells even in the presence of protective bone marrow (BM) myeloid cells including osteoclasts, macrophages, and plasmacytoid dendritic cells. 01A further decreases APRIL-induced adhesion and migration of MM cells via blockade of canonical and noncanonical NF-κB pathways. Moreover, 01A prevents in vivo MM cell growth within implanted human bone chips in SCID mice. Finally, the effect of 01A on MM cell viability is enhanced by lenalidomide and bortezomib. Taken together, these data delineate new molecular mechanisms of in vivo MM growth and immunosuppression critically dependent on BCMA and APRIL in the BM microenvironment, further supporting targeting this prominent pathway in MM.


Assuntos
Antígeno de Maturação de Linfócitos B/fisiologia , Medula Óssea/fisiologia , Proliferação de Células/genética , Microambiente Celular , Tolerância Imunológica/genética , Mieloma Múltiplo/patologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Antígeno de Maturação de Linfócitos B/genética , Medula Óssea/patologia , Linhagem Celular Tumoral , Microambiente Celular/genética , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , Mieloma Múltiplo/genética , Osteoclastos/patologia , Osteoclastos/fisiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
6.
Am J Transplant ; 15(2): 346-57, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25496308

RESUMO

Donor-reactive memory T cells undermine organ transplant survival and are poorly controlled by immunosuppression or costimulatory blockade. Memory CD4 T cells provide CD40-independent help for the generation of donor-reactive effector CD8 T cells and alloantibodies (alloAbs) that rapidly mediate allograft rejection. The goal of this study was to investigate the role of B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) in alloresponses driven by memory CD4 T cells. The short-term neutralization of BAFF alone or BAFF plus APRIL synergized with anti-CD154 mAb to prolong heart allograft survival in recipients containing donor-reactive memory CD4 T cells. The prolongation was associated with reduction in antidonor alloAb responses and with inhibited reactivation and helper functions of memory CD4 T cells. Additional depletion of CD8 T cells did not enhance the prolonged allograft survival suggesting that donor-reactive alloAbs mediate late graft rejection in these recipients. This is the first report that targeting the BAFF cytokine network inhibits both humoral and cellular immune responses induced by memory CD4 T cells. Our results suggest that reagents neutralizing BAFF and APRIL may be used to enhance the efficacy of CD40/CD154 costimulatory blockade and improve allograft survival in T cell-sensitized recipients.


Assuntos
Fator Ativador de Células B/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/fisiologia , Antígenos CD40/fisiologia , Transplante de Coração , Memória Imunológica/fisiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Antígenos CD40/imunologia , Ligante de CD40/imunologia , Ligante de CD40/fisiologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Imunidade Celular/fisiologia , Imunidade Humoral/fisiologia , Isoanticorpos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Animais
7.
Scand J Rheumatol ; 43(6): 462-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24898359

RESUMO

OBJECTIVES: To determine the differences in a proliferation-inducing ligand (APRIL) between seropositive and seronegative rheumatoid arthritis (RA) patients and further investigate the possible pathogenesis of the two subtypes of RA. METHOD: Concentrations of APRIL in sera (18 seropositive RA patients, 16 seronegative RA patients, and 10 healthy controls) and synovial fluid (SF) (eight seropositive RA patients, two seronegative RA patients, and 10 controls) were detected by enzyme-linked immunosorbent assay (ELISA). Infiltration of plasma cells, macrophages, and APRIL-positive cells in the synovium [14 seropositives, eight seronegatives, and 10 osteoarthritis (OA) controls] was detected by immunohistochemistry. Correlations between serum C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), 28-joint Disease Activity Score (DAS28), and sera/SF levels of APRIL and synovial cell infiltration were analysed. RESULTS: The mean serum APRIL level of seropositive RA patients was significantly higher than that of seronegative patients (26.1 ± 31.2 vs. 8.0 ± 10.2 ng/mL, p = 0.03). The level of APRIL in the SF of seropositive RA patients was comparable to that of seronegative patients [47.9 ± 54.4 vs. 32.82 (6.52-59.12) ng/mL, p > 0.05]. The SF APRIL level of RA patients was higher than that of patients with other inflammatory arthritis. Dramatically increased infiltration of APRIL-positive cells in the RA synovium was observed compared with the OA group (seropositive RA vs. OA, p < 0.001; seronegative RA vs. OA, p = 0.001). The infiltration of both plasma cells and macrophages was more in seropositive RA than in OA (p = 0.013 and p = 0.003, respectively). CONCLUSIONS: The serum APRIL levels of seropositive RA patients are significantly higher than those of seronegative RA patients. APRIL may participate in the formation of seropositive RA.


Assuntos
Artrite Reumatoide/etiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Adulto , Idoso , Feminino , Humanos , Macrófagos/fisiologia , Masculino , Pessoa de Meia-Idade , Plasmócitos/fisiologia , Líquido Sinovial/química , Membrana Sinovial/citologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/análise , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/sangue
8.
Nat Rev Rheumatol ; 10(6): 365-73, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24614588

RESUMO

Systemic lupus erythematosus (SLE) is characterized by multisystem immune-mediated injury in the setting of autoimmunity to nuclear antigens. The clinical heterogeneity of SLE, the absence of universally agreed clinical trial end points, and the paucity of validated therapeutic targets have, historically, contributed to a lack of novel treatments for SLE. However, in 2011, a therapeutic monoclonal antibody that neutralizes the cytokine TNF ligand superfamily member 13B (also known as B-cell-activating factor of the TNF family [BAFF]), belimumab, became the first targeted therapy for SLE to have efficacy in a randomized clinical trial. Because of its specificity, the efficacy of belimumab provides an opportunity to increase understanding of SLE pathophysiology. Although belimumab depletes B cells, this effect is not as powerful as that of other B-cell-directed therapies that have not been proven efficacious in randomized clinical trials. In this article, therefore, we review results suggesting that neutralizing BAFF can have effects on the immune system other than depletion of B cells. We also identify aspects of the BAFF system for which data in relation to SLE are still missing, and we suggest studies to investigate the pathogenesis of SLE and ways to refine anti-BAFF therapies. The role of a related cytokine, TNF ligand superfamily member 13 (also known as a proliferation-inducing ligand [APRIL]) in SLE is much less well understood, and hence this review focuses on BAFF.


Assuntos
Fator Ativador de Células B/fisiologia , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/fisiopatologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Autoimunidade , Humanos , Imunossupressores/uso terapêutico
9.
J Immunol ; 191(4): 1935-47, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23858028

RESUMO

Induction of broad and potent neutralizing Abs at the mucosal portals of entry remains a primary goal for most vaccines against mucosally acquired viral infections. Selection of appropriate adjuvants capable of promoting both systemic and mucosal responses will be crucial for the development of effective immunization strategies. In this study, we investigated whether plasmid codelivery of cytokines APRIL, CCL19, or CCL28 can enhance Ag-induced immune responses to HIV-1 gp140. Our results demonstrated that pCCL19 and pCCL28, but not pAPRIL, significantly enhanced Ag-specific systemic and mucosal Ab responses. gp140-specific Abs in serum enhanced by pCCL19 or pCCL28 were broadly distributed across all four IgG subclasses, of which IgG1 was predominant. The enhanced systemic and mucosal Abs showed increased neutralizing activity against both homologous and heterologous HIV-1, and potency correlated with gp140-specific serum IgG and vaginal IgA levels. Measurement of gp140-specific cytokines produced by splenocytes demonstrated that pCCL19 and pCCL28 augmented balanced Th1/Th2 responses. pCCL19 and pCCL28 also increased IgA(+) cells in colorectal mucosal tissue. pCCL19 codelivery resulted in an increase of CCR7(+) CD11c(+) cells in mesenteric lymph nodes and both CCR7(+) CD11c(+) cells and CCR7(+) CD3e(+) cells in spleen, whereas pCCL28 codelivery resulted in an augment of CCR10(+) CD19(+) cells in both spleen and mesenteric lymph nodes. Together, our data indicate that pCCL19 and pCCL28 can enhance HIV-1 envelope-specific systemic and mucosal Ab responses, as well as T cell responses. Such enhancements appear to be associated with mobilization of responsive immunocytes into secondary lymphoid organs and mucosal tissues through interactions with corresponding receptors.


Assuntos
Subpopulações de Linfócitos B/imunologia , Quimiocina CCL19/fisiologia , Quimiocinas CC/fisiologia , Anticorpos Anti-HIV/biossíntese , Tecido Linfoide/imunologia , Subpopulações de Linfócitos T/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Vacinas contra a AIDS/imunologia , Animais , Quimiocina CCL19/genética , Quimiocinas CC/genética , Quimiotaxia , Feminino , Vetores Genéticos/administração & dosagem , Células HEK293 , Anticorpos Anti-HIV/imunologia , Células HeLa , Humanos , Isotipos de Imunoglobulinas/biossíntese , Isotipos de Imunoglobulinas/imunologia , Imunofenotipagem , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Mucosa/imunologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Organismos Livres de Patógenos Específicos , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Vacinação , Vacinas de DNA/imunologia , Vagina/imunologia
10.
Blood ; 122(5): 726-33, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23775715

RESUMO

The characterization of immunoglobulin heavy chain (IGH) translocations provides information on the diagnosis and guides therapeutic decisions in mature B-cell malignancies while enhancing our understanding of normal and malignant B-cell biology. However, existing methodologies for the detection of IGH translocations are labor intensive, often require viable cells, and are biased toward known IGH fusions. To overcome these limitations, we developed a capture sequencing strategy for the identification of IGH rearrangements at nucleotide level resolution and tested its capabilities as a diagnostic and discovery tool in 78 primary diffuse large B-cell lymphomas (DLBCLs). We readily identified IGH-BCL2, IGH-BCL6, IGH-MYC, and IGH-CCND1 fusions and discovered IRF8, EBF1, and TNFSF13 (APRIL) as novel IGH partners in these tumors. IRF8 and TNFSF13 expression was significantly higher in lymphomas with IGH rearrangements targeting these loci. Modeling the deregulation of IRF8 and EBF1 in vitro defined a lymphomagenic profile characterized by up-regulation of AID and/or BCL6, down-regulation of PRMD1, and resistance to apoptosis. Using a capture sequencing strategy, we discovered the B-cell relevant genes IRF8, EBF1, and TNFSF13 as novel targets for IGH deregulation. This methodology is poised to change how IGH translocations are identified in clinical settings while remaining a powerful tool to uncover the pathogenesis of B-cell malignancies.


Assuntos
Cadeias Pesadas de Imunoglobulinas/genética , Fatores Reguladores de Interferon/genética , Linfoma de Células B/genética , Proteínas de Fusão Oncogênica/genética , Transativadores/genética , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Sequência de Bases , Linhagem Celular Tumoral , Biblioteca Gênica , Rearranjo Gênico do Linfócito B/genética , Células HEK293 , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Fatores Reguladores de Interferon/fisiologia , Dados de Sequência Molecular , Transativadores/fisiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Estudos de Validação como Assunto
11.
Cytokine Growth Factor Rev ; 24(3): 203-15, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23684423

RESUMO

The BAFF system plays a key role in the development of autoimmunity, especially in systemic lupus erythematosus (SLE). This often leads to the assumption that BAFF is mostly a B cell factor with a specific role in autoimmunity. Focus on BAFF and autoimmunity, driven by pharmaceutical successes with the recent approval of a novel targeted therapy Belimumab, has relegated other potential roles of BAFF to the background. Far from being SLE-specific, the BAFF system has a much broader relevance in infection, cancer and allergy. In this review, we provide the latest views on additional roles of the BAFF system in health and diseases, as well as an update on BAFF and autoimmunity, with particular focus on current clinical trials.


Assuntos
Fator Ativador de Células B/fisiologia , Linfócitos B/imunologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Asma/fisiopatologia , Autoimunidade/imunologia , Antígeno de Maturação de Linfócitos B/fisiologia , Infecções Bacterianas/fisiopatologia , Ensaios Clínicos como Assunto , Doença Enxerto-Hospedeiro/fisiopatologia , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Doenças Parasitárias/fisiopatologia , Proteínas Recombinantes de Fusão/uso terapêutico , Proteína Transmembrana Ativadora e Interagente do CAML/fisiologia , Imunologia de Transplantes/fisiologia , Viroses/fisiopatologia
12.
Clin Immunol ; 148(3): 322-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23269199

RESUMO

The B lymphocyte stimulator (BLyS)-A PRoliferation-Inducing Ligand (APRIL) signaling pathway has an important role in the selection, maturation and survival of B cells and plays a significant role in the pathogenesis of systemic lupus erythematosus (SLE). The inhibition of BLyS, a survival factor for transitional and mature B cells, has recently proven to be successful in large phase III clinical trials that led to the approval of an anti-BLyS monoclonal antibody (belimumab) for the treatment of SLE. Yet, there is currently a need to both understand better the mechanisms of action of belimumab in SLE and better define the subsets of patients that are more likely to respond to the drug.


Assuntos
Fator Ativador de Células B/antagonistas & inibidores , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Fator Ativador de Células B/fisiologia , Ensaios Clínicos Fase III como Assunto , Humanos , Lúpus Eritematoso Sistêmico/etiologia , Lúpus Eritematoso Sistêmico/imunologia , Receptores do Fator de Necrose Tumoral/fisiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
13.
J Neurotrauma ; 30(6): 434-40, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23088438

RESUMO

Autoimmunity is thought to contribute to poor neurological outcomes after spinal cord injury (SCI). There are few mechanism-based therapies, however, designed to reduce tissue damage and neurotoxicity after SCI because the molecular and cellular bases for SCI-induced autoimmunity are not completely understood. Recent groundbreaking studies in rodents indicate that B cells are responsible for SCI-induced autoimmunity. This novel paradigm, if confirmed in humans, could aid in the design of neuroprotective immunotherapies. The aim of this study was to investigate the molecular signaling pathways and mechanisms by which autoimmunity is induced after SCI, with the goal of identifying potential targets in therapies designed to reduce tissue damage and inflammation in the chronic phase of SCI. To that end, we performed an exploratory microarray analysis of peripheral blood mononuclear cells to identify differentially expressed genes in chronic SCI. We identified a gene network associated with lymphoid tissue structure and development that was composed of 29 distinct molecules and five protein complexes, including two cytokines, a proliferation-inducing ligand (APRIL) and B-cell-activating factor (BAFF), and one receptor, B-cell maturation antigen (BMCA) involved in B cell development, proliferation, activation, and survival. Real-time polymerase chain reaction analysis from ribonucleic acid samples confirmed upregulation of these three genes in SCI. To our knowledge, this is the first report that peripheral blood mononuclear cells produce increased levels of BAFF and APRIL in chronic SCI. This finding provides evidence of systemic regulation of SCI-autoimmunity via APRIL and BAFF mediated activation of B cells through BMCA and points toward these molecules as potential targets of therapies designed to reduce neuroinflammation after SCI.


Assuntos
Autoimunidade/fisiologia , Fator Ativador de Células B/fisiologia , Antígeno de Maturação de Linfócitos B/fisiologia , Traumatismos da Medula Espinal/imunologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Adulto , Idoso , Linfócitos B/imunologia , Proliferação de Células , Humanos , Ligantes , Masculino , Pessoa de Meia-Idade , Transdução de Sinais/fisiologia , Traumatismos da Medula Espinal/patologia
14.
Curr Rheumatol Rep ; 14(4): 295-302, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22535567

RESUMO

BAFF (B-cell-activating factor) is a critical survival factor for transitional and mature B cells and is a promising therapeutic target for systemic lupus erythematosus (SLE). In 2010-2011, two phase 3 clinical trials showed that the addition of the anti-BAFF antibody belimumab to standard-of-care therapy in patients with moderately active SLE results in a better outcome at 52 weeks than standard-of-care therapy alone. Belimumab has been US Food and Drug Administration approved for the treatment of SLE, and other drugs that target BAFF are now in various stages of clinical testing. This review describes the function of BAFF and its homolog APRIL (a proliferation-inducing ligand) and addresses the rationale for the treatment of SLE with BAFF/APRIL inhibitors.


Assuntos
Fator Ativador de Células B/antagonistas & inibidores , Fatores Imunológicos/uso terapêutico , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Fator Ativador de Células B/fisiologia , Subpopulações de Linfócitos B/efeitos dos fármacos , Subpopulações de Linfócitos B/imunologia , Humanos , Fatores Imunológicos/farmacologia , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Terapia de Alvo Molecular/métodos , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
15.
PLoS One ; 7(2): e31837, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22355399

RESUMO

SLE pathogenesis is complex, but it is now widely accepted that autoantibodies play a key role in the process by forming excessive immune complexes; their deposits within tissues leading to inflammation and functional damages. A proliferation inducing ligand (APRIL) is a member of the tumor necrosis factor (TNF) superfamily mediating antibody-producing plasma cell (PC)-survival that may be involved in the duration of pathogenic autoantibodies in lupus. We found significant increases of APRIL at the mRNA and protein levels in bone marrow but not spleen cells from NZB/W lupus mice, as compared to control mice. Selective antibody-mediated APRIL blockade delays disease development in this model by preventing proteinuria, kidney lesions, and mortality. Notably, this was achieved by decreasing anti-DNA and anti-chromatin autoantibody levels, without any perturbation of B- and T-cell homeostasis. Thus, anti-APRIL treatment may constitute an alternative therapy in SLE highly specific to PCs compared to other B-cell targeting therapies tested in this disease, and likely to be associated with less adverse effects than any anti-inflammatory and immunosuppressant agents previously used.


Assuntos
Anticorpos Monoclonais/farmacologia , Lúpus Eritematoso Sistêmico/prevenção & controle , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Anticorpos Antinucleares/sangue , Autoanticorpos/sangue , Western Blotting , Feminino , Citometria de Fluxo , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Baço/imunologia , Baço/metabolismo , Baço/patologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
16.
Dev Comp Immunol ; 37(1): 202-6, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22197036

RESUMO

Here we describe the identification of a Danio rerio homologue of a proliferation-inducing ligand (APRIL) of the TNF family (designated zAPRIL). Sequence analysis showed that the open reading frame of zAPRIL consists of 600 bases encoding a protein of 199-amino acids. Recombinant soluble APRIL (zsAPRIL) was constructed consisting of fluorescence-enhanced green fluorescent protein (EGFP) and cloned into a pET28a vector. SDS-PAGE and western blotting analysis indicated a high-level expression of soluble EGFP/zsAPRIL protein in Escherichia coli BL21 (DE3). Observation by confocal microscopy demonstrated that EGFP/zsAPRIL could successfully bind to the surface receptors of zebrafish lymphocytes. In vitro survival analysis revealed that purified EGFP/zsAPRIL was able to promote the survival of zebrafish lymphocytes in a dose-dependent manner. The biological role of APRIL does not seem to be restricted to proliferation induction. Zebrafish may could served as a model organism for further study of APRIL.


Assuntos
Proteínas Recombinantes de Fusão/genética , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Proteínas de Peixe-Zebra/genética , Animais , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Clonagem Molecular , Linfócitos/fisiologia , Dados de Sequência Molecular , Filogenia , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Baço/citologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia
17.
Reumatol Clin ; 6S3: S20-4, 2011 Mar.
Artigo em Espanhol | MEDLINE | ID: mdl-21794767

RESUMO

Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by synovitis and progressive destruction of the joint cartilage and underlying bone, together with diverse extra-articular manifestations. Cytokines act as soluble effector mediators of the inflammatory process. Therapeutic neutralization with monoclonal antibodies against the pro-inflammatory cytokines TNF-alpha and interleukin 1 (IL-1) has shown a clear efficacy on inflammation and clinical manifestations of RA, although a percentage of patients do not respond. This review covers new relevant cytokines in the RA physiopathology and potential biomarkers of inflammation. The current challenge is to develop biomarkers that enable an earlier diagnosis, as well as prognostic markers and new therapeutic candidates. Combined administration of several of these cytokines could eventually address a personalized treatment approach for each patient.


Assuntos
Artrite Reumatoide/fisiopatologia , Interleucinas/fisiologia , Adipocinas/fisiologia , Antirreumáticos/farmacologia , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Fator Ativador de Células B/fisiologia , Linfócitos B/imunologia , Linfócitos B/patologia , Citocina TWEAK , Citocinas/antagonistas & inibidores , Citocinas/fisiologia , Previsões , Humanos , Inflamação , Interleucina-1/antagonistas & inibidores , Interleucinas/antagonistas & inibidores , Medicina de Precisão , Receptores de Citocinas/antagonistas & inibidores , Receptores de Citocinas/fisiologia , Membrana Sinovial/metabolismo , Inibidores do Fator de Necrose Tumoral , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fatores de Necrose Tumoral/fisiologia
18.
Clin Calcium ; 21(3): 364-71, 2011 Mar.
Artigo em Japonês | MEDLINE | ID: mdl-21358057

RESUMO

NF-κB activation is frequently found in a variety of tumors, especially in those that are resistant to chemotherapy. The malignant phenotypes of tumor cells that are required for invasion and metastasis could be established by the output of NF-κB signaling including interactions with white blood cells, anti-apoptosis and angiogenesis. Consequently, by utilizing the similar mechanisms for immune cells to migrate through the body, tumor cells migrate via blood stream, survive against selective pressures in other organs they stop by, achieve re-growth as metastasis, or make a clinical relapse by traveling back to the primary site.


Assuntos
Inflamação , NF-kappa B/fisiologia , Neoplasias , Proteína de Ligação a CREB/fisiologia , Quimiotaxia de Leucócito/fisiologia , Humanos , Hipóxia , Ligantes , Neoplasias/imunologia , Neoplasias/patologia , Neovascularização Patológica , Espécies Reativas de Oxigênio , Transdução de Sinais , Receptor 4 Toll-Like/fisiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/fisiologia
19.
Immunity ; 34(2): 247-57, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21333555

RESUMO

Although both conventional dendritic cells (cDCs) and plasmacytoid dendritic cells (pDCs) are present in the gut-associated lymphoid tissues (GALT), the roles of pDCs in the gut remain largely unknown. Here we show a critical role for pDCs in T cell-independent (TI) IgA production by B cells in the GALT. When pDCs of the mesenteric lymph nodes (MLNs) and Peyer's patches (PPs) (which are representative GALT) were cultured with naive B cells to induce TI IgA class switch recombination (CSR), IgA production was substantially higher than in cocultures of these cells with cDCs. IgA production was dependent on APRIL and BAFF production by pDCs. Importantly, pDC expression of APRIL and BAFF was dependent on stromal cell-derived type I IFN signaling under steady-state conditions. Our findings provide insight into the molecular basis of pDC conditioning to induce mucosal TI IgA production, which may lead to improvements in vaccination strategies and treatment for mucosal-related disorders.


Assuntos
Células Dendríticas/imunologia , Imunidade nas Mucosas/imunologia , Imunoglobulina A Secretora/biossíntese , Switching de Imunoglobulina , Transferência Adotiva , Animais , Fator Ativador de Células B/fisiologia , Técnicas de Cocultura , Citocinas/fisiologia , Vida Livre de Germes , Imunoglobulina A Secretora/genética , Interferon Tipo I/fisiologia , Linfonodos/imunologia , Mesentério/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nódulos Linfáticos Agregados/imunologia , Receptor Cross-Talk , Receptores de Antígenos de Linfócitos T alfa-beta/deficiência , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/deficiência , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Células Estromais/imunologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
20.
Neoplasma ; 58(1): 45-50, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21067265

RESUMO

It was demonstrated that TNF superfamily proteins may affect significantly the time of leukemic cells' survival in the course of B-cell chronic lymphocytic leukemia (B-CLL). The aim of our study was to evaluate the expression and release of BAFF (B-cell activating factor), APRIL (a proliferation-inducing ligand) and TRAIL (TNF-related apoptosis inducing ligand) molecules belonging to the cytokines of the superfamily of the tumor necrosis factor (TNF) by neutrophils (PMNs) and, for comparison, B cells isolated from the blood of patients with B-CLL vs. their concentration in the blood serum. 40 patients suffering from B-CLL and a control group of 15 healthy subjects were included in the study. Cytoplasmic fractions of PMNs and B cells were analyzed with the use of western blotting for the presence of TRAIL, BAFF and APRIL. Soluble TRAIL, BAFF and APRIL in the culture supernatants and the serum were assessed using ELISA kits. PMNs and B cells of patients with B-CLL before treatment demonstrated the statistically significantly higher expression of APRIL and BAFF proteins when compared with the control group of healthy subjects. In contrast, the expression of TRAIL protein in both types of cells of patients was statistically significantly lower than its expression in the control cells. In the supernatants of PMN and B lymphocytes of patients the decreased concentrations of sBAFF, unchanged of APRIL and increased of sTRAIL molecules were demonstrated. The results of studies carried out in patients with B-CLL before treatment indicate that the relations demonstrated between APRIL, BAFF and TRAIL molecules, released by neutrophils and B cells and relations between their concentrations in the serum can significantly influence the development of B-CLL.


Assuntos
Fator Ativador de Células B/fisiologia , Linfócitos B/fisiologia , Leucemia Linfocítica Crônica de Células B/patologia , Neutrófilos/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Adulto , Idoso , Fator Ativador de Células B/sangue , Ensaio de Imunoadsorção Enzimática , Humanos , Leucemia Linfocítica Crônica de Células B/imunologia , Pessoa de Meia-Idade , Ligante Indutor de Apoptose Relacionado a TNF/sangue , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...