Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
FASEB J ; 38(2): e23443, 2024 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-38265281

RESUMEN

Immune-mediated acute hepatic injury is characterized by the destruction of a large number of hepatocytes and severe liver function damage. Interleukin-28A (IL-28A), a member of the IL-10 family, is notable for its antiviral properties. However, despite advances in our understanding of IL-28A, its role in immune-mediated acute injury remains unclear. The present study investigated the role of IL-28A in concanavalin A (Con A)-induced acute immune liver injury. After Con A injection in mice, IL-28A level significantly increased. IL-28A deficiency was found to protect mice from acute liver injury, prolong survival time, and reduce serum aspartate aminotransferase and alanine aminotransferase levels. In contrast, recombinant IL-28A aggravated liver injury in mice. The proportion of activated M1 macrophages was significantly lower in the IL-28A-deficiency group than in the wild-type mouse group. In adoptive transfer experiments, M1 macrophages from WT could exacerbate mice acute liver injury symptoms in the IL-28A deficiency group. Furthermore, the expression of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), IL-12, IL-6, and IL-1ß, by M1 macrophages decreased significantly in the IL-28A-deficiency group. Western blotting demonstrated that IL-28A deficiency could limit M1 macrophage polarization by modulating the nuclear factor (NF)-κB, mitogen-activated protein kinase (MAPK), and interferon regulatory factor (IRF) signaling pathways. In summary, IL-28A deletion plays an important protective role in the Con A-induced acute liver injury model and IL-28A deficiency inhibits the activation of M1 macrophages by inhibiting the NF-κB, MAPK, and IRF signaling pathways. These results provide a potential new target for the treatment of immune-related hepatic injury.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Citocinas , Interferón lambda , Interleucinas , Animales , Ratones , Concanavalina A , Factores Reguladores del Interferón , Hígado , Macrófagos , Proteínas Quinasas Activadas por Mitógenos , Interferón lambda/genética , Interleucinas/genética
2.
Cell Mol Life Sci ; 81(1): 110, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38429401

RESUMEN

Toll-like receptors (TLRs), especially TLR7, play an important role in systemic lupus erythematosus (SLE) pathogenesis. However, the regulatory mechanism underlying the abnormal activation of TLR pathways in patients with SLE has not been elucidated. Notably, accumulating evidence indicates that myeloid-derived suppressor cells (MDSCs) are important regulators of inflammation and autoimmune diseases. Compared with healthy control subjects, patients with SLE have a greater proportion of MDSCs among peripheral blood mononuclear cells (PBMCs); however, the effect of MDSCs on TLR7 pathway activation has not been determined. In the present study, lupus MDSCs significantly promoted TLR7 pathway activation in macrophages and dendritic cells (DCs), exacerbating the imiquimod-induced lupus model. RNA-sequencing analysis revealed significant overexpression of S100 calcium-binding protein A8 (S100A8) and S100A9 in MDSCs from diseased MRL/lpr mice. In vitro and in vivo studies demonstrated that S100A8/9 effectively promoted TLR7 pathway activation and that S100A8/9 deficiency reversed the promoting effect of MDSCs on TLR7 pathway activation in lupus. Mechanistically, MDSC-derived S100A8/9 upregulated interferon gamma (IFN-γ) secretion by macrophages and IFN-γ subsequently promoted TLR7 pathway activation in an autocrine manner. Taken together, these findings suggest that lupus MDSCs promote TLR7 pathway activation and lupus pathogenesis through the S100A8/9-IFN-γ axis. Our study identified an important target for SLE therapy.


Asunto(s)
Calgranulina A , Calgranulina B , Lupus Eritematoso Sistémico , Células Supresoras de Origen Mieloide , Animales , Ratones , Células Dendríticas/metabolismo , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , Macrófagos/metabolismo , Ratones Endogámicos MRL lpr , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo
3.
Br J Cancer ; 130(9): 1542-1551, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38461171

RESUMEN

BACKGROUND: Tumor cells continue to evolve the metastatic potential in response to signals provided by the external microenvironment during metastasis. Platelets closely interact with tumor cells during hematogenous metastasis and facilitate tumor development. However, the molecular mechanisms underlying this process are not fully understood. METHODS: RNA-sequencing was performed to screen differentially expressed genes mediated by platelets. The effects of platelet and CD39 on tumor metastasis were determined by experimental metastasis models with WT, NCG and CD39-/- mice. RESULTS: RNA-sequencing results showed that platelets significantly up-regulated CD39 expression in tumor cells. CD39 is a novel immune checkpoint molecule and a key driver of immunosuppression. Our data provided evidence that the expression of CD39 was enhanced by platelets in a platelet-tumor cell contact dependent manner. Although the role of CD39 expressed by immune cells is well established, the effect of CD39 expressed by tumor cells on tumor cell behavior, anti-tumor immunity and tumor metastasis is unclear. We found that CD39 promoted tumor cell invasion, but had no effect on proliferation and migration. Notably, we showed that the ability of platelets to prime tumor cells for metastasis depends on CD39 in the experimental tumor metastasis model. CD39 silencing resulted in fewer experimental metastasis formation, and this anti-metastasis effect was significantly reduced in platelet-depleted mice. Furthermore, overexpression of CD39 in tumor cells promoted metastasis. In order to eliminate the effect of CD39 expressed in cells other than tumor cells, we detected tumor metastasis in CD39-/- mice and obtained similar results. Moreover, overexpression of CD39 in tumor cells inhibited antitumor immunity. Finally, the data from human samples also supported our findings. CONCLUSIONS: Our study shows that direct contact with platelets induces CD39 expression in tumor cells, leading to immune suppression and promotion of metastasis.


Asunto(s)
Antígenos CD , Apirasa , Plaquetas , Metástasis de la Neoplasia , Animales , Apirasa/genética , Apirasa/metabolismo , Plaquetas/metabolismo , Plaquetas/patología , Ratones , Antígenos CD/genética , Antígenos CD/metabolismo , Humanos , Línea Celular Tumoral , Femenino , Ratones Noqueados , Movimiento Celular , Microambiente Tumoral/inmunología , Regulación Neoplásica de la Expresión Génica
4.
FASEB J ; 37(7): e22967, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37269155

RESUMEN

Neutrophils participate in the pathogenesis of ulcerative colitis (UC) through regulating the intestinal homeostasis. Several inflammatory diseases are reported to be regulated by proline-rich tyrosine kinase 2B (PTK2B). However, the role of PTK2B in regulating the function of neutrophils and the pathogenesis of UC remains unknown. In this study, the mRNA and protein levels of PTK2B in the colonic tissues from UC patients were measured by using quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, and immunohistochemistry. TAE226, a PTK2B inhibitor, was used to inhibit the activity of PTK2B in neutrophils, and then, the pro-inflammatory factors were analyzed by using qRT-PCR and ELISA. To determine the role of PTK2B in intestinal inflammation, a dextran sulfate sodium (DSS)-induced colitis model was established in PTK2B gene knockout (PTK2B KO) and wild-type (WT) mice. We found that compared with healthy donor controls, the expression level of PTK2B was significantly elevated in inflamed mucosa from UC patients. In addition, expression of PTK2B was positively correlated with the severity of disease. Pharmacological inhibition of PTK2B could markedly reduce the generation of reactive oxygen species (ROS), myeloperoxidase (MPO), and antimicrobial peptides (S100a8 and S100a9) in neutrophils. The vitro study showed that tumor necrosis factor (TNF)-α is involved in promoting the expression of PTK2B in neutrophils. As expected, UC patients treated with infliximab, an anti-TNF-α agent, showed significantly reduced level of PTK2B in neutrophils, as well as in the intestinal mucosa. Of note, compared with DSS-treated WT mice, DSS-treated PTK2B KO mice showed more severe colitis symptoms. Mechanistically, PTK2B could enhance neutrophil migration by regulating CXCR2 and GRK2 expression via the p38 MAPK pathway. Additionally, mice treated with TAE226 exhibited the same effects. In conclusion, PTK2B is involved in the pathogenesis of UC by promoting the migration of neutrophils and inhibiting mucosal inflammation, highlighting PTK2B as a new potential therapeutic target to treat UC.


Asunto(s)
Colitis Ulcerosa , Quinasa 2 de Adhesión Focal , Animales , Ratones , Colitis Ulcerosa/metabolismo , Colon/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Quinasa 2 de Adhesión Focal/genética , Quinasa 2 de Adhesión Focal/metabolismo , Inmunidad , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Neutrófilos/metabolismo , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Factor de Necrosis Tumoral alfa/metabolismo , Humanos
5.
Biochem Cell Biol ; 100(4): 309-324, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35544948

RESUMEN

Liver fibrosis is a very common health problem and currently lacks effective treatments. Cullin RING E3 ligases (CRLs) regulate the turnover of ∼20% of mammalian cell proteins. Neddylation, the process by which NEDD8 is covalently attached to cullin proteins through sequential enzymatic reactions, is critical for the activation of CRLs and was recently found to be elevated in liver fibrosis. NEDD8-activating enzyme E1-specific inhibition led to the reduced liver damage characterized by decreased apoptosis, inflammation, and fibrosis. However, the relevance of a co-E3 ligase, DCN1, in liver fibrosis remains unclear. Here, a novel and potent DCN1-UBC12 interaction inhibitor HZX-960 was discovered with an IC50 value of 9.37 nmol/L, which could inhibit the neddylation of cullin3. Importantly, we identified that HZX-960 treatment could attenuate transforming growth factor ß-induced liver fibrotic responses by reducing the deposition of collagen I and α-smooth muscle actin, and upregulating cellular NF-E2-related factor 2, hemeoxygenase 1, and NADPH quinone oxidoreductase-1 levels in two hepatic stellate cell lines. Additionally, DCN1 was shown to be unregulated in CCl4-induced mice liver tissue, and liver fibrotic signaling in mice was reduced by HZX-960. Therefore, our data demonstrated that HZX-960 possessed anti-liver fibrosis ability and that DCN1 may be a potential therapeutic target for liver fibrosis treatment.


Asunto(s)
Inhibidores Enzimáticos , Cirrosis Hepática , Enzimas Ubiquitina-Conjugadoras , Ubiquitina-Proteína Ligasas , Animales , Proteínas Cullin/metabolismo , Inhibidores Enzimáticos/farmacología , Cirrosis Hepática/tratamiento farmacológico , Ratones , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitinación
6.
Int J Mol Sci ; 23(17)2022 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-36076977

RESUMEN

Polycomp group (PcG) proteins are members of highly conserved multiprotein complexes, recognized as gene transcriptional repressors during development and shown to play a role in various physiological and pathological processes. PcG proteins consist of two Polycomb repressive complexes (PRCs) with different enzymatic activities: Polycomb repressive complexes 1 (PRC1), a ubiquitin ligase, and Polycomb repressive complexes 2 (PRC2), a histone methyltransferase. Traditionally, PRCs have been described to be associated with transcriptional repression of homeotic genes, as well as gene transcription activating effects. Particularly in cancer, PRCs have been found to misregulate gene expression, not only depending on the function of the whole PRCs, but also through their separate subunits. In this review, we focused especially on the recent findings in the transcriptional regulation of PRCs, the oncogenic and tumor-suppressive roles of PcG proteins, and the research progress of inhibitors targeting PRCs.


Asunto(s)
Proteínas de Drosophila , Neoplasias , Humanos , Neoplasias/genética , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 2/genética , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo
7.
Bioorg Chem ; 105: 104424, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33161253

RESUMEN

In this paper, based on molecular hybridization, a series of [1,2,3]triazolo[4,5-d]pyrimidine derivatives containing hydrazine was synthesized and their antiproliferative activities against 5 cancer cell lines (MGC-803, PC3, PC9, EC9706 and SMMC-7721) were evaluated. We found that most of them exhibited obvious growth inhibition effects on these tested cancer cells, especially compound 34 on PC3 cells (IC50 = 26.25 ± 0.28 nM). Meanwhile, compound 34 displayed best selectivity on PC3, compared with the other cancer cell lines, as well as excellent selectivity towards normal cell lines (Het-1A, L02 and GES-1). Further investigations demonstrated that 34 could significantly inhibit PC3 cells' colony formation, increase cellular ROS content, suppress EGFR expression and induce apoptosis. Our findings indicate that 34 may serve as a novel lead compound for the discovery of more triazolopyrimidine derivatives with improved anticancer potency and selectivity.


Asunto(s)
Antineoplásicos/farmacología , Hidrazonas/farmacología , Pirimidinas/farmacología , Triazoles/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Hidrazonas/química , Estructura Molecular , Pirimidinas/síntesis química , Pirimidinas/química , Relación Estructura-Actividad , Triazoles/síntesis química , Triazoles/química , Células Tumorales Cultivadas
8.
Int J Mol Sci ; 21(17)2020 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-32887217

RESUMEN

Invasive breast cancer is highly regulated by tumor-derived cytokines in tumor microenvironment. The development of drugs that specifically target cytokines are promising in breast cancer treatment. In this study, we reported that arctigenin, a bioactive compound from Arctium lappa L., could decrease tumor-promoting cytokines GM-CSF, MMP-3, MMP-9 and TSLP in breast cancer cells. Arctigenin not only inhibited the proliferation, but also the invasion and stemness of breast cancer cells via decreasing GM-CSF and TSLP. Mechanistically, arctigenin decreased the promoter activities of GM-CSF and TSLP via reducing the nuclear translocation of NF-κB p65 which is crucial for the transcription of GM-CSF and TSLP. Furthermore, arctigenin-induced depletion of GM-CSF and TSLP inhibited STAT3 phosphorylation and ß-catenin signaling resulting in decreased proliferation, invasion and stemness of breast cancer cells in vitro and in vivo. Our findings provide new insights into the mechanism by which tumor-promoting cytokines regulate breast cancer progression and suggest that arctigenin is a promising candidate for cytokine-targeted breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Citocinas/metabolismo , Furanos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Lignanos/farmacología , Factor de Transcripción STAT3/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Citocinas/genética , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factor de Transcripción STAT3/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
10.
J Cell Mol Med ; 23(7): 4738-4745, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31062436

RESUMEN

Long non-coding RNA MIR503 host gene (MIR503HG) is located on chromosome Xq26.3, and has been found to be deregulated in many types of human malignancy and function as tumour suppressor or promoter based on cancer types. The role of MIR503HG in breast cancer was still unknown. In our study, we found MIR503HG expression was significantly decreased in triple-negative breast cancer tissues and cell lines. Furthermore, we observed low MIR503HG expression was correlated with late clinical stage, lymph node metastasis and distant metastasis. In the survival analysis, we observed that triple-negative breast cancer patients with low MIR503HG expression had a statistically significant worse prognosis compared with those with high MIR503HG expression, and low MIR503HG expression was a poor independent prognostic factor for overall survival in triple-negative breast cancer patients. The study in vitro suggested MIR503HG inhibits cell migration and invasion via miR-103/OLFM4 axis in triple negative breast cancer. In conclusion, MIR503HG functions as a tumour suppressive long non-coding RNA in triple negative breast cancer.


Asunto(s)
Movimiento Celular/genética , Factor Estimulante de Colonias de Granulocitos/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/genética , Secuencia de Bases , Línea Celular Tumoral , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Persona de Mediana Edad , Análisis Multivariante , Invasividad Neoplásica , Pronóstico , Modelos de Riesgos Proporcionales , ARN Largo no Codificante/genética
11.
Immunology ; 157(3): 257-267, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31120548

RESUMEN

Asthma is a chronic inflammatory disease that involves a variety of cytokines and cells. Interleukin-16 (IL-16) is highly expressed during allergic airway inflammation and is involved in its development. However, its specific mechanism of action remains unclear. In the present study, we used an animal model of ovalbumin (OVA)-induced allergic asthma with mice harboring an IL-16 gene deletion to investigate the role of this cytokine in asthma, in addition to its underlying mechanism. Increased IL-16 expression was observed during OVA-induced asthma in C57BL/6J mice. However, when OVA was used to induce asthma in IL-16-/- mice, a diminished inflammatory reaction, decreased bronchoalveolar lavage fluid (BALF) eosinophil numbers, and the suppression of OVA-specific IgE levels in the serum and BALF were observed. The results also demonstrated decreased levels of T helper type 2 (Th2) and Th17 cytokines upon OVA-induced asthma in IL-16-/- mice. Hence, we confirmed that IL-16 enhances the lung allergic inflammatory response and suggest a mechanism possibly associated with the up-regulation of IgE and the promotion of Th2 and Th17 cytokine production. This work explored the mechanism underlying the regulation of IL-16 in asthma and provides a new target for the clinical treatment of asthma.


Asunto(s)
Asma/metabolismo , Hiperreactividad Bronquial/metabolismo , Interleucina-16/metabolismo , Pulmón/metabolismo , Ovalbúmina , Células Th17/metabolismo , Células Th2/metabolismo , Animales , Asma/inmunología , Asma/fisiopatología , Asma/prevención & control , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/fisiopatología , Hiperreactividad Bronquial/prevención & control , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Broncoconstricción , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Interleucina-16/deficiencia , Interleucina-16/genética , Pulmón/inmunología , Pulmón/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Bazo/inmunología , Bazo/metabolismo , Células Th17/inmunología , Células Th2/inmunología
12.
J Autoimmun ; 102: 50-64, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31080014

RESUMEN

Accumulating evidence suggests granulocyte macrophage-colony stimulating factor (GM-CSF) can function as an inflammatory mediator, but whether GM-CSF-producing CD4+ T cells (TH-GM-CSF) are a distinct T helper cell subset is lacking. Herein we demonstrate that interleukin (IL)-1ß exclusively drives differentiation of naïve CD4+ T cells into TH-GM-CSF cells via inducing ubiquitination of IL-1 receptor-associated kinase 1 (IRAK1) and subsequent activation of the transcription factor NF-kappaB (NF-κB), independent of RAR-related orphan receptor gamma (RORγt) required for TH17 differentiation. In vivo, TH-GM-CSF cells are present in murine Citrobacter Rodentium infections and mediate colitis following adoptive transfer of CD4+ T cells into Rag1-/- mice via GM-CSF-induced macrophage activation. The TH-GM-CSF cell phenotype is stable and distinct from the TH17 genetic program, but IL-1ß can convert pre-formed TH17 cells into TH-GM-CSF cells, thereby accounting for previously reported associations between IL-17 and GM-CSF. Together, our results newly identify IL-1ß/NF-κB-dependent TH-GM-CSF cells as a unique T helper cell subset and highlight the importance of CD4+ T cell-derived GM-CSF induced macrophage activation as a previously undescribed T cell effector mechanism.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-1beta/inmunología , Activación de Macrófagos/inmunología , Células Th17/citología , Células Th17/inmunología , Animales , Diferenciación Celular/inmunología , Citrobacter rodentium/inmunología , Colitis/inmunología , Inflamación/inmunología , Inflamación/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Células Th17/patología , Ubiquitinación
13.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2796-2807, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28802852

RESUMEN

Autophagy extensively participates in immune responses and inflammatory diseases. Myeloid-derived suppressor cells (MDSCs) are derived from CD11b+Gr1+ cells under pathological conditions and play an immunosuppressive role in the pathogenesis of cancer and inflammatory diseases. However, the role of autophagy in regulating the accumulation and activity of MDSCs remains unknown. In the present study, we evaluated the effects and mechanisms of autophagy on regulating accumulation and activity of MDSCs. We first found that granulocytic MDSCs (G-MDSCs), but not monocytic MDSCs (M-MDSCs), were accumulated in mice challenged by lipopolysaccharide (LPS) and showed an elevated autophagy activity. Pharmacological inhibition of autophagy significantly enhanced accumulation of G-MDSCs in vivo and in vitro. Notably, inhibition of autophagy enhanced the immunosuppressive activity of G-MDSCs on M1 macrophage polarization by promoting reactive oxygen species (ROS) production. Inhibition of autophagy promotes the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in G-MDSCs, which is required for the accumulation and activity of MDSCs. In addition, in vivo pharmacological inhibition of autophagy significantly attenuated the condition of mice challenged by LPS. Thus, we conclude that inhibition of autophagy contributes to accumulation and immunosuppressive function of G-MDSCs by promoting the activation of STAT3 signaling, suggesting that autophagy may play a critical role in regulating accumulation and activity of MDSCs. Our study provides new insights into understanding the mechanisms of autophagy in regulating immune responses and pathogenesis of inflammatory diseases.


Asunto(s)
Autofagia/inmunología , Granulocitos/inmunología , Células Supresoras de Origen Mieloide/inmunología , Factor de Transcripción STAT3/inmunología , Choque Séptico/inmunología , Transducción de Señal/inmunología , Animales , Autofagia/efectos de los fármacos , Granulocitos/patología , Lipopolisacáridos/toxicidad , Ratones , Células Supresoras de Origen Mieloide/patología , Choque Séptico/inducido químicamente , Choque Séptico/patología , Transducción de Señal/efectos de los fármacos
14.
Biochim Biophys Acta ; 1852(8): 1585-98, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25912736

RESUMEN

The activation of IFN-α signaling in B cells contributes to the pathogenesis of systemic lupus erythematosus (SLE). Many studies suggest that estrogens are closely related to the gender difference in the prevalence of SLE. However, the underlying mechanism of the interaction between estrogens and the activation of IFN-α signaling in SLE B cells remains incompletely understood. In the present study, we first found that healthy female mice showed an up-regulated type I IFN-induced gene signature in B cells compared with age-matched male mice, and an in vivo study revealed that the gender difference was related to 17ß-estradiol. Moreover, we found that 17ß-estradiol could enhance the activation of IFN-α signaling in an ERα-dependent manner by down-regulating the expression of three microRNAs, including let-7e-5p, miR-98-5p and miR-145a-5p. These microRNAs could target the 3'UTR of the IKKε-encoding gene IKBKE directly and regulate the expression of IKKε, which can promote the activation of IFN-α signaling. In addition, compared with age-matched male mice, female mice showed a higher level of IKKε and lower levels of let-7e-5p, miR-98-5p and miR-145a-5p in B cells. Moreover, peripheral blood mononuclear cells from women showed a higher level of IKKε and lower levels of let-7e-5p, miR-98-5p and miR-145a-5p compared with those from age-matched men. These data suggest that 17ß-estradiol amplifies the activation of IFN-α signaling in B cells via IKKε by down-regulating the expression of let-7e-5p, miR-98-5p and miR-145a-5p. Our findings may provide a new perspective for understanding the mechanism underlying the gender difference in the prevalence of SLE.


Asunto(s)
Linfocitos B/efectos de los fármacos , Estradiol/farmacología , Quinasa I-kappa B/genética , Interferón-alfa/metabolismo , MicroARNs/genética , Animales , Linfocitos B/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Quinasa I-kappa B/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
15.
Eur J Immunol ; 45(8): 2377-88, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25959715

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the overexpression of IFN-α. IFN-α induces autophagy via the JAK1-STAT1 signaling pathway, contributing to the pathogenesis of SLE. Recent studies reported that B cells from patients with SLE and NZB/W F1 mice had enhanced autophagy activity; however, the mechanism still remains unknown. Here, we show that the protein tyrosine phosphatase STS-1 (suppressor of T-cell receptor signaling 1) was significantly overexpressed in B cells from patients with SLE and MRL/lpr mice. Notably, STS-1 promoted IFN-α-induced autophagy in B cells by enhancing the JAK1-STAT1 signaling activation. STS-1 inhibited the phosphorylation of the E3 ubiquitin protein ligase c-cbl, and subsequently promoted IFN-α-induced phosphorylation of tyrosine kinase 2, leading to JAK1-STAT1 signaling activation. Furthermore, STAT1 and JAK1 inhibitors blocked the IFN-α-induced autophagy promoted by STS-1, indicating that STS-1 promotes IFN-α-induced autophagy via the JAK1-STAT1 signaling. Our results demonstrate the importance of STS-1 in regulating IFN-α-induced autophagy in B cells, and this could be used as a therapeutic approach to treat SLE.


Asunto(s)
Autofagia/inmunología , Linfocitos B/inmunología , Interferón-alfa/inmunología , Janus Quinasa 1/inmunología , Proteínas Tirosina Fosfatasas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Factor de Transcripción STAT1/inmunología , Transducción de Señal/inmunología , Animales , Femenino , Humanos , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/terapia , Masculino , Ratones , Proteínas Proto-Oncogénicas c-cbl/inmunología , TYK2 Quinasa/inmunología
16.
Eur J Immunol ; 45(7): 1934-45, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25929465

RESUMEN

B cells present lipid antigens to CD1d-restricted invariant natural killer T (iNKT) cells to maintain autoimmune tolerance, and this process is disrupted in systemic lupus erythematosus (SLE). Inflammation may inhibit CD1d expression to exacerbate the pathology of lupus. However, how inflammation regulates CD1d expression on B cells is unclear in SLE. In the present study, we showed that the surface expression of CD1d on B cells from SLE mice was decreased and that stimulation of inflammatory responses through TLR9 decreased the membrane and total CD1d levels of CD1d on B cells. Moreover, inflammation-related microRNA-155 (miR-155) negatively correlated with the expression of CD1d in B cells. miR-155 directly targeted the 3'-untranslated region (3'-UTR) of CD1d upon TLR9 activation in both humans and mice. The inhibitory effects of miR-155 on CD1d expression in B cells impaired their antigen-presenting capacity to iNKT cells. In addition, Ets-1, a susceptibility gene of SLE, also directly regulated the expression of the CD1d gene at the transcriptional level. These findings provide new insight into the mechanism underlying decreased CD1d expression on B cells in SLE, suggesting that inhibition of inflammation may increase CD1d expression in B cells to ameliorate SLE via modulating iNKT cells.


Asunto(s)
Antígenos CD1d/biosíntesis , Linfocitos B/inmunología , Regulación de la Expresión Génica/inmunología , Lupus Eritematoso Sistémico/inmunología , MicroARNs/inmunología , Proteína Proto-Oncogénica c-ets-1/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos CD1d/inmunología , Western Blotting , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inflamación/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Células T Asesinas Naturales/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Toll-Like 9/inmunología , Transfección
17.
Acta Biochim Biophys Sin (Shanghai) ; 47(8): 620-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26071573

RESUMEN

Estrogens are strongly implicated in gender differences in immune responses by influencing the development and activation of immune cells. Recent studies have shown that myeloid-derived suppressor cells (MDSCs), derived from CD11b(+)Gr-1(+) myeloid cells under pathological conditions, play vital roles in modulating immune responses. However, it is still unknown the effects of estrogens on MDSCs. In the present study, we investigated the effects and mechanisms of estrogens on regulating the accumulation of MDSCs. It was found that, compared with male patients with systemic lupus erythematosus (SLE), female patients with SLE showed a higher frequency of MDSCs in peripheral blood mononuclear cells and a higher level of tumor necrosis factor α (TNF-α) in serum. Notably, estradiol level in the serum of female patients with SLE was positively correlated with the frequency of MDSCs. Moreover, 17ß-estradiol could promote TNF-α-induced accumulation of MDSCs in vivo by increasing the fundamental frequency of CD11b(+)Gr-1(+) cells. Furthermore, 17ß-estradiol promoted the secretion of TNF-α in vivo, which contributed to the increase of the frequency of CD11b(+)Gr-1(+) cells. In addition, it was also found that female mice showed a higher frequency of CD11b(+)Gr-1(+) cells and a higher TNF-α level in blood than the age-matched male mice. These data indicate that 17ß-estradiol contributes to the accumulation of MDSCs in blood by promoting TNF-α secretion, which increases the fundamental frequency of CD11b(+)Gr-1(+) cells. Our findings provide a new insight into the mechanism of gender difference in the prevalence of inflammation and autoimmune diseases.


Asunto(s)
Estradiol/metabolismo , Lupus Eritematoso Sistémico/sangre , Células Mieloides/metabolismo , Factor de Necrosis Tumoral alfa/sangre , Animales , Femenino , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/fisiopatología , Masculino , Ratones , Células Mieloides/patología , Factores Sexuales , Factores Supresores Inmunológicos/sangre , Factor de Necrosis Tumoral alfa/metabolismo
18.
Cell Death Dis ; 15(1): 88, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-38272906

RESUMEN

Fatty acid metabolism, particularly fatty acid synthesis, is a very important cellular physiological process in which nutrients are used for energy storage and biofilm synthesis. As a key enzyme in the fatty acid metabolism, fatty acid synthase (FASN) is receiving increasing attention. Although previous studies on FASN have mainly focused on various malignancies, many studies have recently reported that FASN regulates the survival, differentiation, and function of various immune cells, and subsequently participates in the occurrence and development of immune-related diseases. However, few studies to date systematically summarized the function and molecular mechanisms of FASN in immune cell biology and related diseases. In this review, we discuss the regulatory effect of FASN on immune cells, and the progress in research on the implications of FASN in immune-related diseases. Understanding the function of FASN in immune cell biology and related diseases can offer insights into novel treatment strategies for clinical diseases.


Asunto(s)
Ácido Graso Sintasas , Lipogénesis , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Línea Celular Tumoral , Metabolismo de los Lípidos , Ácidos Grasos
19.
Cell Rep Med ; 5(5): 101510, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38614093

RESUMEN

Key gene mutations are essential for colorectal cancer (CRC) development; however, how the mutated tumor cells impact the surrounding normal cells to promote tumor progression has not been well defined. Here, we report that PIK3CA mutant tumor cells transmit oncogenic signals and result in malignant transformation of intestinal epithelial cells (IECs) via paracrine exosomal arachidonic acid (AA)-induced H3K4 trimethylation. Mechanistically, PIK3CA mutations sustain SGK3-FBW7-mediated stability of the cPLA2 protein, leading to the synthetic increase in AA, which is transported through exosome and accumulated in IECs. Transferred AA directly binds Menin and strengthens the interactions of Menin and MLL1/2 methyltransferase. Finally, the combination of VTP50469, an inhibitor of the Menin-MLL interaction, and alpelisib synergistically represses PDX tumors harboring PIK3CA mutations. Together, these findings unveil the metabolic link between PIK3CA mutant tumor cells and the IECs, highlighting AA as the potential target for the treatment of patients with CRC harboring PIK3CA mutations.


Asunto(s)
Ácido Araquidónico , Transformación Celular Neoplásica , Ensamble y Desensamble de Cromatina , Fosfatidilinositol 3-Quinasa Clase I , Mutación , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Humanos , Ácido Araquidónico/metabolismo , Animales , Mutación/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ensamble y Desensamble de Cromatina/genética , Ratones , Línea Celular Tumoral , Colon/patología , Colon/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Exosomas/metabolismo , Exosomas/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Histonas/metabolismo , Histonas/genética
20.
J Med Chem ; 66(1): 1063-1081, 2023 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-36580346

RESUMEN

Interrupting the embryonic ectoderm development (EED)-H3K27me3 interaction represents a promising strategy to allosterically inhibit polycomb repressive complex 2 (PRC2) for cancer therapy. In this work, we report the structure-based design of new triazolopyrimidine-based EED inhibitors, which structurally feature the electron-rich indole ring at the C8 position. Particularly, ZJH-16 directly binds to EED (HTRF IC50 = 2.72 nM, BLI KD = 4.4 nM) and potently inhibits the growth of KARPAS422 and Pfeiffer cells. In both cells, ZJH-16 is selectively engaged with EED and reduces H3K27 trimethylation levels. ZJH-16 inhibits the gene silencing function of PRC2 in KARPAS422 cells. ZJH-16 possesses favorable pharmacokinetic (PK) profiles with an excellent oral bioavailability (F = 94.7%). More importantly, ZJH-16 shows robust tumor regression in the KARPAS422 xenograft model after oral administration with the tumor growth inhibition reaching nearly 100%. The robust antitumor efficacy and favorable PK profiles of ZJH-16 warrant further advanced preclinical development for lymphoma treatment.


Asunto(s)
Histonas , Linfoma , Humanos , Histonas/metabolismo , Complejo Represivo Polycomb 2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA