Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Infect Dis ; 229(3): 855-865, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-37603461

RESUMEN

BACKGROUND: Calcitonin gene-related peptide (CGRP), an immunomodulatory neuropeptide, is important for regulating pain transmission, vasodilation, and the inflammatory response. However, the molecular mechanisms of the CGRP-mediated immune response remain unknown. METHODS: The effects of CGRP on bacterial meningitis (BM) and its underlying mechanisms were investigated in BM mice in vivo and macrophages in vitro. RESULTS: Peripheral injection of CGRP attenuated cytokine storms and protected mice from fatal pneumococcal meningitis, marked by increased bacterial clearance, improved neuroethology, and reduced mortality. When the underlying mechanisms were investigated, we found that CGRP induces proteasome-dependent degradation of major histocompatibility complex class II (MHC-II) in macrophages and then inhibits CD4+ T-cell activation. MARCH1 was identified as an E3 ligase that can be induced by CGRP engagement and promote K48-linked ubiquitination and degradation of MHC-II in macrophages. These results provide new insights into neuropeptide CGRP-mediated immune regulation mechanisms. CONCLUSIONS: We conclude that targeting the nervous system and manipulating neuroimmune communication is a promising strategy for treating intracranial infections like BM.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina , Meningitis Bacterianas , Ratones , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Antígenos de Histocompatibilidad Clase II , Ubiquitinación , Complejo Mayor de Histocompatibilidad , Homeostasis , Ubiquitina-Proteína Ligasas/metabolismo
2.
PLoS Pathog ; 17(9): e1009901, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34506605

RESUMEN

Neddylation, an important type of post-translational modification, has been implicated in innate and adapted immunity. But the role of neddylation in innate immune response against RNA viruses remains elusive. Here we report that neddylation promotes RNA virus-induced type I IFN production, especially IFN-α. More importantly, myeloid deficiency of UBA3 or NEDD8 renders mice less resistant to RNA virus infection. Neddylation is essential for RNA virus-triggered activation of Ifna gene promoters. Further exploration has revealed that mammalian IRF7undergoes neddylation, which is enhanced after RNA virus infection. Even though neddylation blockade does not hinder RNA virus-triggered IRF7 expression, IRF7 mutant defective in neddylation exhibits reduced ability to activate Ifna gene promoters. Neddylation blockade impedes RNA virus-induced IRF7 nuclear translocation without hindering its phosphorylation and dimerization with IRF3. By contrast, IRF7 mutant defective in neddylation shows enhanced dimerization with IRF5, an Ifna repressor when interacting with IRF7. In conclusion, our data demonstrate that myeloid neddylation contributes to host anti-viral innate immunity through targeting IRF7 and promoting its transcriptional activity.


Asunto(s)
Inmunidad Innata/inmunología , Factor 7 Regulador del Interferón/inmunología , Células Mieloides/inmunología , Infecciones por Virus ARN/inmunología , Virus ARN/inmunología , Animales , Factor 7 Regulador del Interferón/biosíntesis , Ratones , Células Mieloides/metabolismo , Proteína NEDD8/deficiencia , Procesamiento Proteico-Postraduccional , Ubiquitinas/deficiencia
3.
Immunol Invest ; 52(1): 1-19, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35997714

RESUMEN

The mechanisms by which retinoic acid-inducible gene I (RIG-I), a critical RNA virus sensor, is regulated in many biological and pathological processes remain to be determined. Here, we demonstrate that T cell immunoglobulin and mucin protein-3 (Tim-3), an immune checkpoint inhibitor, mediates infection tolerance by suppressing RIG-I-type I interferon pathway. Overexpression or blockade of Tim-3 affects type I interferon expression, virus replication, and tissue damage in mice following H1N1 infection. Tim-3 signaling decreases RIG-I transcription via STAT1 in macrophages and promotes the proteasomal dependent degradation of RIG-I by enhancing K-48-linked ubiquitination via the E3 ligase RNF-122. Silencing RIG-I reversed Tim-3 blockage-mediated upregulation of type I interferon in macrophages. We thus identified a new mechanism through which Tim-3 mediates the immune evasion of H1N1, which may have clinical implications for the treatment of viral diseases.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Interferón Tipo I , Ratones , Animales , Receptor 2 Celular del Virus de la Hepatitis A/genética , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Macrófagos , Interferón Tipo I/genética , Ubiquitina-Proteína Ligasas/genética
4.
Eur J Immunol ; 51(1): 103-114, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32652569

RESUMEN

Deficiency of Itch, an E3 ubiquitin ligase, usually induced severe systemic and progressive autoimmune disease. The Itch function is well studied in T cells but not in B cells. We hypothesize that B-cell-specific Itch deficiency promoted antigen-induced B-cell activation and antibody-expressing plasma cell (PC) production. We found that unlike Itch KO, Itch cKO (CD19cre Itchf/f ) mice did not demonstrated a significant increase in the sizes of spleens and LNs, antibody level, and base mutation of antibody gene. However, in line with the fact that Itch expression decreased in GC B cells, PCs, and plasmablast (PB)-like SP 2/0 cells, Itch deficiency promoted B-cell activation and antibody production induced by antigens including lipopolysaccharide (LPS) and sheep red blood cells (SRBCs). Mechanistically, we found that Itch deficiency promotes antigen-induced cytokine production because Itch controls the proteins (e.g., eIF3a, eIF3c, eIF3h) with translation initiation factor activity. Altogether, our data suggest that Itch deficiency promotes antigen-driven B-cell response. This may provide hints for Itch-targeted treatment of patients with autoimmune disease.


Asunto(s)
Linfocitos B/enzimología , Linfocitos B/inmunología , Ubiquitina-Proteína Ligasas/deficiencia , Animales , Formación de Anticuerpos , Antígenos/inmunología , Citocinas/biosíntesis , Eritrocitos/inmunología , Factores Eucarióticos de Iniciación/metabolismo , Humanos , Lipopolisacáridos/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Inmunológicos , Ovinos , Ubiquitina-Proteína Ligasas/genética
5.
J Med Virol ; 94(7): 3233-3239, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35322423

RESUMEN

Although individuals with coronavirus disease 2019 (COVID-19) are known to be at increased risk for other conditions resulting from pathogenic changes (including metaplastic or anaplastic) in the lungs and other organs and organ systems, it is still unknown whether COVID-19 affects childhood intelligence. The present two-sample Mendelian randomization study aims to identify the genetic causal link between COVID-19 and childhood intelligence. Four COVID-19 genetic instrumental variants (IVs) were chosen from the largest genome-wide association studies (GWAS) for COVID-19 (hospitalized vs. population) (6406 cases and 902 088 controls of European ancestry). The largest childhood intelligence GWAS (n = 12 441 individuals of European ancestry) was used to evaluate the effect of the identified COVID-19-associated genetic IVs on childhood intelligence. We found that as the genetic susceptibility to COVID-19 increased, childhood intelligence followed a decreasing trend, according to mr_egger (ß = -0.156; p = 0.601; odds ratio [OR] = 0.856; 95% confidence interval [CI]: 0.522-1.405), simple mode (ß = -0.126; p = 0.240; OR = 0.882; 95% CI: 0.745-1.044), and weighted mode (ß = -0.121; p = 0.226; OR = 0.886; 95% CI: 0.758-1.036) analyses. This trend was further demonstrated by the weighted median (ß = -0.134; p = 0.031; OR = 0.875; 95% CI: 0.774-0.988) and the inverse variance weighted (ß = -0.152; p = 0.004; OR = 0.859; 95% CI: 0.776-0.952). Our analysis suggests a causal link between genetically increased COVID-19 and decreased childhood intelligence. Thus, COVID-19 may be a risk factor for declines in childhood intelligence.


Asunto(s)
COVID-19 , COVID-19/epidemiología , COVID-19/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Inteligencia , Análisis de la Aleatorización Mendeliana , Polimorfismo de Nucleótido Simple
6.
Scand J Immunol ; 93(2): e12981, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33031600

RESUMEN

T cell immunoglobulin and mucin domain-3 (Tim-3), an immune checkpoint molecule, plays critical roles in maintaining innate immune homeostasis; however, the mechanisms underlying these roles remain to be determined. Here, we determined that Tim-3 controls glycolysis in macrophages and thus contributes to phenotype shifting. Tim-3 signal blockade significantly increases lactate production by macrophages, but does not influence cell proliferation or apoptosis. Tim-3 attenuates glucose uptake by inhibiting hexokinase 2 (HK2) expression in macrophages. Tim-3-mediated inhibition of macrophage glycolysis and the expression of proinflammatory cytokines, tumour necrosis factor (TNF)-α and interleukin (IL)-1ß are reversed by HK2 silencing. Finally, we demonstrated that Tim-3 inhibits HK2 expression via the STAT1 pathway. We have thus discovered a new way by which Tim-3 modulates macrophage function.


Asunto(s)
Glucólisis/inmunología , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Hexoquinasa/inmunología , Macrófagos/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Animales , Apoptosis/inmunología , Línea Celular , Proliferación Celular/fisiología , Citocinas/inmunología , Células HEK293 , Humanos , Inmunidad Innata/inmunología , Inflamación/inmunología , Interleucina-1beta/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/inmunología
7.
J Infect Dis ; 221(5): 830-840, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-31586389

RESUMEN

BACKGROUND: T-cell immunoglobulin and mucin protein 3 (Tim-3) is an immune checkpoint inhibitor that has therapeutic implications for many tumors and infectious diseases. However, the mechanisms by which Tim-3 promotes immune evasion remain unclear. METHODS: In this study, we demonstrated that Tim-3 inhibits the expression of major histocompatibility complex class I (MHC-I) in macrophages at both the messenger ribonucleic acid and protein levels by inhibiting the STAT1-NLRC5 signaling pathway. RESULTS: As a result, MHC-I-restricted antigen presentation by macrophages was inhibited by Tim-3 both in vitro and in a Listeria monocytogenes infection model in vivo. Systemic overexpression of Tim-3 or specific knockout of Tim-3 in macrophages significantly attenuated or enhanced CD8+ T-cell activation and infection damage in L monocytogenes-infected mice, respectively. CONCLUSIONS: Thus, we identified a new mechanism by which Tim-3 promotes L monocytogenes immune evasion. Further studies on this pathway might shed new light on the physio-pathological roles of Tim-3 and suggest new approaches for intervention.


Asunto(s)
Antígenos HLA-A/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Evasión Inmune/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Macrófagos/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Células HEK293 , Receptor 2 Celular del Virus de la Hepatitis A/genética , Humanos , Listeriosis/microbiología , Activación de Linfocitos/genética , Ratones , Ratones Transgénicos , Células RAW 264.7 , Transfección
8.
Am J Respir Cell Mol Biol ; 61(6): 727-736, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31162951

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive lung disorder and lacks effective treatments because of unclear mechanisms. Aberrant function of alveolar macrophages is directly linked to pulmonary fibrosis. Here, we show TIM-3 (T-cell immunoglobulin domain and mucin domain-3), a key regulator of macrophage function, aggravates pulmonary fibrosis. TIM-3 mRNA of patients with IPF was analyzed based on the Gene Expression Omnibus and Array Express databases. Lung pathology and profibrotic molecules were assessed in a bleomycin (BLM)-induced pulmonary fibrosis model using wild-type (WT) and TIM-3 transgenic (TIM-3-TG) mice. Macrophage cells, RAW264.7, were then applied to investigate the effect of macrophage TIM-3 under BLM exposure in vitro. Macrophage depletion and adoptive-transfer experiments were finally performed to examine lung morphology and profibrotic molecules. TIM-3 expression was increased both in patients with IPF and in our BLM-induced mouse model. TIM-3-TG mice developed more serious pathological changes in lung tissue and higher expressions of TGF-ß1 (transforming growth factor-ß1) and IL-10 than WT mice. After BLM treatment, TGF-ß1 and IL-10 expression was significantly decreased in RAW264.7 cells after TIM-3 knock-out, whereas it was increased in TIM-3-TG peritoneal macrophages. The scores of pulmonary fibrosis in WT and TIM-3-TG mice were significantly reduced, and there was no difference between them after macrophage depletion. Furthermore, WT mice receiving adoptive macrophages from TIM-3-TG mice also had more serious lung fibrosis and increased expression of TGF-ß1 and IL-10 than those receiving macrophages from WT mice. Our findings revealed that overexpressed TIM-3 in alveolar macrophages aggravated pulmonary fibrosis.


Asunto(s)
Receptor 2 Celular del Virus de la Hepatitis A/sangre , Receptor 2 Celular del Virus de la Hepatitis A/fisiología , Fibrosis Pulmonar Idiopática/patología , Macrófagos Alveolares/metabolismo , Traslado Adoptivo , Animales , Bleomicina/toxicidad , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Receptor 2 Celular del Virus de la Hepatitis A/deficiencia , Receptor 2 Celular del Virus de la Hepatitis A/genética , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Interleucina-10/biosíntesis , Pulmón/patología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/trasplante , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células RAW 264.7 , ARN Mensajero/biosíntesis , Factor de Crecimiento Transformador beta1/biosíntesis
9.
Scand J Immunol ; 89(2): e12738, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30506563

RESUMEN

T cell immunoglobulin and mucin domain protein 3 (Tim-3) is an immune checkpoint inhibitor in T cells and innate immune cells. The deregulated upregulation of Tim-3 is related to immune exhaustion in tumour and viral infection. To overcome Tim-3-mediated immune tolerance, we developed a novel monoclonal antibody against human Tim-3 (L3G) and investigated its roles in inhibiting Tim-3 signalling and overcoming immune tolerance in T cells and monocytes/macrophages. The administration of L3G to cultured peripheral blood mononuclear cells (PBMCs) significantly increased the production of IFN-γ and IL-2 and the expression of type I interferon. The administration of L3G also increased the production of IFN-γ, IL-8 and type I interferon in U937 cells and primary monocytes. We investigated the mechanisms by which L3G enhances pro-inflammatory cytokine expression, and our data show that L3G enhances STAT1 phosphorylation in both monocytes/macrophages and T cells. Finally, in an H1N1 infection model of PBMCs and U937 cells, L3G decreased the viral load and enhanced the expression of interferon. Thus, we developed a functional antibody with therapeutic potential against Tim-3-mediated infection tolerance.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Macrófagos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Linfocitos T/inmunología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Macrófagos/virología , Ratones , Ratones Endogámicos BALB C , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Células U937 , Carga Viral
10.
BMC Cancer ; 19(1): 700, 2019 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-31311517

RESUMEN

BACKGROUND: Multiple myeloma (MM), characterized by cancerous proliferation of plasmablasts (PB) and plasma cells (PC), remains incurable in many patients. Differentially expressed molecules between MM PCs and healthy PCs have been explored in order to identify novel targets for treating MM. In the present study, we searched for novel MM therapeutic targets by comparing mRNA expression patterns between the Mus musculus myeloma plasmablast-like SP 2/0 cell line and LPS-induced PB/PC. METHODS: Gene expression profiles of LPS-induced PB/PC and SP 2/0 cells were determined using RNA-sequencing. A predicted gene (Gm40600) was found to be expressed at a low level in SP 2/0 cells. To study the role of Gm40600 in malignant PC, Gm40600 cDNA was cloned into a lentiviral vector (LV201) containing a puromycin selectable marker that was then transfected into SP 2/0 cells. Stable Gm40600-expressing SP 2/0 cells were selected using puromycin. The effect of Gm40600 on SP 2/0 cell proliferation, cell cycle/apoptosis, and tumor progression was assessed by cell counting kit-8 (CCK8), flow cytometry (FACS), and the SP 2/0 isograft mouse model, respectively. The effect of Gm40600 on mRNA and protein expression was evaluated by RNA-sequencing and western blotting, respectively. RESULTS: We found that SP 2/0 cells expressed lower level of Gm40600 mRNA as compared to LPS-induced PB/PC. Overexpression of Gm40600 significantly suppressed SP 2/0 cell proliferation and isograft tumor progression in an isograft mouse model by promoting apoptosis. In addition, Gm40600 overexpression suppressed transcription of the gene encoding Bcl2. Gm40600 overexpression also reduced the expression of PC-associated transcription factors Blimp1 and Xbp1, which promote transcription of the gene that encodes Bcl2. CONCLUSIONS: Gm40600 reduced SP 2/0 cell proliferation and isograft tumor growth and progression by suppressing Blimp1 and Xbp1-mediated Bcl2 transcription to induce apoptosis. Thus, regulation of a human homolog of Gm40600, or associated factors, may be a potential therapeutic approach for treating MM.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Proteína 1 de Unión a la X-Box/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Isoinjertos , Ratones , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/genética , Proteína 1 de Unión a la X-Box/metabolismo
11.
Immunology ; 153(1): 71-83, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28799242

RESUMEN

The Nod-like receptor protein 3 (NLRP3) inflammasome plays roles in host defence against invading pathogens and in the development of autoimmune damage. Strict regulation of these responses is important to avoid detrimental effects. Here, we demonstrate that T cell Ig mucin-3 (Tim-3), an immune checkpoint inhibitor, inhibits NLRP3 inflammasome activation by damping basal and lipopolysaccharide-induced nuclear factor-κB-mediated up-regulation of NLRP3 and interleukin-1ß during the priming step and basal and ATP/lipopolysaccharide-induced ATP production, K+ efflux, and reactive oxygen species production during the activation step. Residues Y256/Y263 in the C-terminal region of Tim-3 are required for these inhibitory effects on the NLRP3 inflammasome. In mice with alum-induced peritonitis, blockade of Tim-3 exacerbates peritonitis by overcoming the inhibitory effect of Tim-3 on NLRP3 inflammasome activation, while transgenic expression of Tim-3 attenuates inflammation by inhibiting NLRP3 inflammasome activation. Our results show that Tim-3 is a critical negative regulator of NLRP3 inflammasome and provides a potential target for intervention of diseases with uncontrolled inflammasome activation.


Asunto(s)
Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Inflamasomas/inmunología , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Peritonitis/inmunología , Peritonitis/metabolismo , Adenosina Trifosfato/biosíntesis , Adulto , Animales , Estudios de Casos y Controles , Caspasa 1 , Línea Celular , Modelos Animales de Enfermedad , Femenino , Receptor 2 Celular del Virus de la Hepatitis A/química , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Peritonitis/patología , Potasio/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Adulto Joven
12.
Cell Immunol ; 328: 9-17, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29499909

RESUMEN

IL-1α in vitro promotes immunoglobulin secretion by inducing proliferation of mature B cells, whereas IL-1α deficiency has no effect on in vivo antibody production. However, the reason IL-1α deficiency does not reduce in vivo antibody production is still unclear. In this study, we found that similar as in vivo data, IL-1α deficiency did not affect antibody production in in vitro LPS-stimulated B cells. Surprisingly, LPS-stimulated IL-1α-/- B cells reduced a key antibody production-related transcription factor X-box binding protein 1 (Xbp-1) expression. Furthermore, we found that IL-1α deficiency up-regulated mTOR expression, which bypassed Xbp-1 for immunoglobulin secretion. Finally, we showed that Xbp-1 suppressed mTOR expression, whereas mTOR suppressed the activation of Xbp-1 promoter via JunB. Together, these data suggest that IL-1a deficiency reduced Xbp-1 and up-regulated mTOR. This may explain why IL-1α deficiency has no effect on antibody production.


Asunto(s)
Linfocitos B/inmunología , Serina-Treonina Quinasas TOR/fisiología , Proteína 1 de Unión a la X-Box/metabolismo , Animales , Formación de Anticuerpos , Linfocitos B/metabolismo , Linfocitos B/fisiología , Diferenciación Celular/inmunología , Proteínas de Unión al ADN/genética , Femenino , Regulación de la Expresión Génica/inmunología , Interleucina-1alfa/inmunología , Interleucina-1alfa/metabolismo , Interleucina-1alfa/fisiología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Células Plasmáticas/inmunología , Transporte de Proteínas , Serina-Treonina Quinasas TOR/inmunología , Factores de Transcripción/genética , Activación Transcripcional , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/inmunología
13.
Cancer Cell Int ; 18: 138, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30220882

RESUMEN

BACKGROUND: Both multiple myeloma (MM) and systemic lupus erythematosus (SLE) are associated with abnormal production of plasma cells, although their pathological mechanism of each disease is different. The main characteristic of both diseases is uncontrolled differentiation of B cells into plasmablast/plasma cells. Despite continuous research on prognostic factors and the introduction of new agents for MM and SLE, treatments still do not exist for controlling plasmablast/plasma cells. Thus, it is necessary to identify novel therapeutic targets of plasmablast/plasma cells. Because of its plasmablast-like characteristics, the mus musculus myeloma SP 2/0 cell line was used in this study to test the effect of a novel therapeutic agent (BC094916 overexpression) on plasmablast/plasma cells. METHODS: We first determined gene expression profiles of plasma cells using Affymetrix microarrays and RNA-sequencing. The effect of BC094916 on SP 2/0 cell proliferation, cell cycle, and apoptosis was determined by CCK8 and fluorescence-activated cell sorting. The SP 2/0 xenograft mouse model was used to assess the impact of BC094916 on tumor progression. The luciferase reporter system was used to evaluate the effect of BC094916 on Creb1 and Bcl2 transcription. RESULTS: We found that BC094916 mRNA was decreased in plasma cells. The mouse myeloma cell line SP 2/0 expressed low levels of BC094916 mRNA, whereas BC094916 overexpression suppressed SP 2/0 cell proliferation by inducing apoptosis. BC094916 overexpression suppressed tumor progression in the SP 2/0 xenograft mouse model. We also found that BC094916 mediate apoptosis by suppressing transcription of the Creb1 and Bcl2 genes, which promote the transcription of eukaryotic translation initiation and elongation factor genes. CONCLUSIONS: BC094916 overexpression suppressed Creb1 and Bcl2 transcription to induce cell apoptosis, which suppressed SP 2/0 proliferation and xenograft tumor progression. Thus, BC094916 overexpression may be a potential therapeutic agent for treatment of MM and autoimmune diseases such as SLE.

14.
J Cell Mol Med ; 21(12): 3658-3669, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28707394

RESUMEN

As the first line of defence, marginal zone (MZ) B cells play principal roles in clearing blood-borne pathogens during infection and are over-primed in autoimmune diseases. However, the basic mechanisms underlying MZ B-cell development are still unclear. We found here that CD19 deficiency blocked the differentiation of marginal zone precursors (MZP) to MZ B cells, whereas CD19 expression in CD19-deficient MZP rescues MZ B-cell generation. Furthermore, CD19 regulates Notch2 cleavage by up-regulating ADAM28 expression in MZP. Finally, we found that CD19 suppressed Foxo1 expression to promote ADAM28 expression in MZP. These results suggest that CD19 controls the differentiation of MZP to MZ B cells by regulating ADAM28-mediated Notch2 cleavage. Thus, we demonstrated the basic mechanisms underlying the differentiation of MZP to MZ B cells.


Asunto(s)
Proteínas ADAM/genética , Antígenos CD19/genética , Linfocitos B/inmunología , Tejido Linfoide/inmunología , Receptor Notch2/genética , Proteínas ADAM/inmunología , Animales , Antígenos CD19/inmunología , Linfocitos B/citología , Diferenciación Celular/inmunología , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/inmunología , Regulación de la Expresión Génica , Tejido Linfoide/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteolisis , Receptor Notch2/inmunología , Transducción de Señal
15.
Immunology ; 150(4): 478-488, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27995618

RESUMEN

Interleukin-10-positive (IL-10+ ) regulatory B (Breg) cells play an important role in restraining excessive inflammatory responses by secreting IL-10. However, it is still unclear what key transcription factors determine Breg cell differentiation. Hence, we explore what transcription factor plays a key role in the expression of IL-10, a pivotal cytokine in Breg cells. We used two types of web-based prediction software to predict transcription factors binding the IL-10 promoter and found that IL-10 promoter had many binding sites for Foxd3. Chromatin immunoprecipitation PCR assay demonstrated that Foxd3 directly binds the predicted binding sites around the start codon upstream by -1400 bp. Further, we found that Foxd3 suppressed the activation of IL-10 promoter by using an IL-10 promoter report system. Finally, knocking out Foxd3 effectively promotes Breg cell production by up-regulating IL-10 expression. Conversely, up-regulated Foxd3 expression was negatively associated with IL-10+ Breg cells in lupus-prone MRL/lpr mice. Hence, our data suggest that Foxd3 suppresses the production of IL-10+ Breg cells by directly binding the IL-10 promoter. This study demonstrates the mechanism for Breg cell production and its application to the treatment of autoimmune diseases by regulating Foxd3 expression.


Asunto(s)
Linfocitos B Reguladores/inmunología , Factores de Transcripción Forkhead/metabolismo , Interleucina-10/metabolismo , Lupus Eritematoso Sistémico/inmunología , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Biología Computacional , Modelos Animales de Enfermedad , Femenino , Factores de Transcripción Forkhead/genética , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Represoras/genética
16.
Eur J Immunol ; 46(6): 1343-50, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27019190

RESUMEN

Interleukin-12 family cytokines have emerged as critical regulators of immunity with some members (IL-12, IL-23) associated with disease pathogenesis while others (IL-27, IL-35) mitigate autoimmune diseases. Each IL-12 family member is comprised of an α and a ß chain, and chain-sharing is a key feature. Although four bona fide members have thus far been described, promiscuous chain-pairing between alpha (IL-23p19, IL-27p28, IL-12/IL-35p35) and beta (IL-12/IL-23p40, IL-27/IL-35Ebi3) subunits, predicts six possible heterodimeric IL-12 family cytokines. Here, we describe a new IL-12 member composed of IL-23p19 and Ebi3 heterodimer (IL-39) that is secreted by LPS-stimulated B cells and GL7(+) activated B cells of lupus-like mice. We further show that IL-39 mediates inflammatory responses through activation of STAT1/STAT3 in lupus-like mice. Taken together, our results show that IL-39 might contribute to immunopathogenic mechanisms of systemic lupus erythematosus, and could be used as a possible target for its treatment.


Asunto(s)
Subunidad p19 de la Interleucina-23/metabolismo , Lupus Eritematoso Sistémico/etiología , Lupus Eritematoso Sistémico/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Receptores de Citocinas/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Inmunofenotipificación , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Subunidad p19 de la Interleucina-23/química , Subunidad p19 de la Interleucina-23/genética , Lupus Eritematoso Sistémico/patología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos MRL lpr , Antígenos de Histocompatibilidad Menor/química , Antígenos de Histocompatibilidad Menor/genética , Multimerización de Proteína , Receptores de Citocinas/química , Receptores de Citocinas/genética , Receptores de Interleucina/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
17.
J Infect Dis ; 213(1): 71-9, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26401029

RESUMEN

Rickettsia heilongjiangensis is the pathogen of Far eastern spotted fever, and T-cell immunoglobulin and mucin domain protein 3 (Tim-3) is expressed in human vascular endothelial cells, the major target cells of rickettsiae. In the present study, we investigated the effects of altered Tim-3 expression in vivo in mice and in vitro in human endothelial cells, on day 3 after R. heilongjiangensis infection. Compared with corresponding controls, rickettsial burdens both in vivo and in vitro were significantly higher with blocked Tim-3 signaling or silenced Tim-3 and significantly lower with overexpressed Tim-3. Additionally, the expression of inducible nitric oxide synthase and interferon γ in endothelial cells with blocked Tim-3 signaling or silenced Tim-3 was significantly lower, while the expression of inducible nitric oxide synthase, interferon γ, and tumor necrosis factor α in transgenic mice with Tim-3 overexpression was significantly higher. These results reveal that enhanced Tim-3 expression facilitates intracellular rickettsial killing in a nitric oxide-dependent manner in endothelial cells during the early phase of rickettsial infection.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas de la Membrana/biosíntesis , Infecciones por Rickettsia/metabolismo , Rickettsia/inmunología , Animales , Línea Celular , Chlorocebus aethiops , Células Endoteliales/inmunología , Células Endoteliales/microbiología , Receptor 2 Celular del Virus de la Hepatitis A , Interacciones Huésped-Patógeno/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Infecciones por Rickettsia/inmunología , Infecciones por Rickettsia/microbiología , Células Vero
18.
J Immunol ; 192(9): 4192-201, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24670802

RESUMEN

Clinical trials have shown that BAFF inhibitors do not reduce memory B cell levels but can reduce the number of mature B cells. It remains uncertain whether BAFF affects memory-maintaining cytokines such as IL-15. We found that BAFF suppressed IL-15 expression in B cells from lupus-like or experimental allergic encephalomyelitis mice. When BAFF was blocked with atacicept-IgG, IL-15 expression was upregulated in lupus-like or experimental allergic encephalomyelitis mice. Finally, we showed that BAFF suppressed IL-15 expression in transitional 2 B cells by reducing Foxo1 expression and inducing Foxo1 phosphorylation. This study suggests that BAFF suppresses IL-15 expression in autoimmune diseases, and this opens up the possible opportunity for the clinical application of BAFF- and IL-15-specific therapeutic agents.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Factor Activador de Células B/inmunología , Linfocitos B/inmunología , Interleucina-15/biosíntesis , Animales , Enfermedades Autoinmunes/metabolismo , Factor Activador de Células B/metabolismo , Linfocitos B/metabolismo , Western Blotting , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Interleucina-15/inmunología , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Esclerosis Múltiple/sangre , Esclerosis Múltiple/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Clin Immunol ; 160(2): 142-54, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26071318

RESUMEN

Recently B-cell activating factor (BAFF) was identified by our group and others as a novel therapeutic target for the treatment of autoimmune diseases. To expand upon this, we utilized microarrays to screen for molecules upregulated in B cells from BAFF-inhibited mice with lupus-like disease and identified metabotropic glutamate receptor 3 (Grm3). In addition to confirming the expression of this receptor in B cells, a synthetic agonist of Grm3 was found to downregulate B cells and ameliorate autoimmune symptoms in mice. Conversely, a Grm3 antagonist increased B-cell numbers and further aggravated disease. Thus, these results suggest that activation of Grm3 ameliorates lupus-like disease in mice by reducing B cell numbers. Not only do the findings presented in this study increase our understanding of the inhibitory signals initiated on the surface of B cells, but they also identify a novel potential target for the treatment of autoimmune diseases.


Asunto(s)
Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Esclerosis Múltiple Crónica Progresiva/inmunología , ARN Mensajero/metabolismo , Receptores de Glutamato Metabotrópico/inmunología , Animales , Linfocitos B/metabolismo , Proliferación Celular , Perfilación de la Expresión Génica , Humanos , Riñón/patología , Lupus Eritematoso Sistémico/genética , Ratones , Esclerosis Múltiple Crónica Progresiva/genética , Receptores de Glutamato Metabotrópico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Clin Immunol ; 160(2): 328-35, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26208474

RESUMEN

Tim-3 is involved in the physiopathology of inflammatory bowel disease (IBD), but the underlying mechanism is unknown. Here, we demonstrated that, in mouse with DSS colitis, Tim-3 inhibited the polarization of pathogenic pro-inflammatory M1 macrophages, while Tim-3 downregulation or blockade resulted in an increased M1 response. Adoptive transfer of Tim-3-silenced macrophages worsened DSS colitis and enhanced inflammation, while Tim-3 overexpression attenuated DSS colitis by decreasing the M1 macrophage response. Co-culture of Tim-3-overexpressing macrophages with intestinal lymphocytes decreased the pro-inflammatory response. Tim-3 shaped intestinal macrophage polarization may be TLR-4 dependent since Tim-3 blockade failed to exacerbate colitis or increase M1 macrophage response in the TLR-4 KO model. Finally, Tim-3 signaling inhibited phosphorylation of IRF3, a TLR-4 downstream transcriptional factor regulating macrophage polarization. A better understanding of this pathway may shed new light on colitis pathogenesis and result in a new therapeutic strategy.


Asunto(s)
Colitis/inmunología , Colon/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Macrófagos/inmunología , Receptores Virales/inmunología , Traslado Adoptivo , Animales , Técnicas de Cocultivo , Colitis/inducido químicamente , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Receptor 2 Celular del Virus de la Hepatitis A , Homeostasis , Factor 3 Regulador del Interferón/metabolismo , Linfocitos/inmunología , Ratones , Ratones Noqueados , Fosforilación , Transducción de Señal , Receptor Toll-Like 4/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA