Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Cell ; 74(6): 1250-1263.e6, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31054974

RESUMEN

Alternative pre-mRNA-splicing-induced post-transcriptional gene expression regulation is one of the pathways for tumors maintaining proliferation rates accompanying the malignant phenotype under stress. Here, we uncover a list of hyperacetylated proteins in the context of acutely reduced Acetyl-CoA levels under nutrient starvation. PHF5A, a component of U2 snRNPs, can be acetylated at lysine 29 in response to multiple cellular stresses, which is dependent on p300. PHF5A acetylation strengthens the interaction among U2 snRNPs and affects global pre-mRNA splicing pattern and extensive gene expression. PHF5A hyperacetylation-induced alternative splicing stabilizes KDM3A mRNA and promotes its protein expression. Pathologically, PHF5A K29 hyperacetylation and KDM3A upregulation axis are correlated with poor prognosis of colon cancer. Our findings uncover a mechanism of an anti-stress pathway through which acetylation on PHF5A promotes the cancer cells' capacity for stress resistance and consequently contributes to colon carcinogenesis.


Asunto(s)
Empalme Alternativo , Carcinogénesis/genética , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/genética , Proteínas de Unión al ARN/genética , Transactivadores/genética , Acetilcoenzima A/deficiencia , Acetilación , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Células HCT116 , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células MCF-7 , Masculino , Ratones , Ratones Desnudos , Pronóstico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Transducción de Señal , Análisis de Supervivencia , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
2.
EMBO Rep ; 24(12): e57500, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37870259

RESUMEN

SIRT2, a cytoplasmic member of the Sirtuin family, has important roles in immunity and inflammation. However, its function in regulating the response to DNA virus infection remains elusive. Here, we find that SIRT2 is a unique regulator among the Sirtuin family that negatively modulates the cGAS-STING-signaling pathway. SIRT2 is down-regulated after Herpes simplex virus-1 (HSV-1) infection, and SIRT2 deficiency markedly elevates the expression levels of type I interferon (IFN). SIRT2 inhibits the DNA binding ability and droplet formation of cGAS by interacting with and deacetylating G3BP1 at K257, K276, and K376, leading to the disassembly of the cGAS-G3BP1 complex, which is critical for cGAS activation. Administration of AGK2, a selective SIRT2 inhibitor, protects mice from HSV-1 infection and increases the expression of IFN and IFN-stimulated genes. Our study shows that SIRT2 negatively regulates cGAS activation through G3BP1 deacetylation, suggesting a potential antiviral strategy by modulating SIRT2 activity.


Asunto(s)
ADN Helicasas , Inmunidad Innata , Animales , Ratones , ADN Helicasas/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Transducción de Señal , Sirtuina 2/genética , Sirtuina 2/metabolismo
3.
Ann Hematol ; 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38847852

RESUMEN

Bone marrow stromal cells (BMSCs) can promote the growth of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Histone deacetylases (HDACs) play essential roles in the proliferation and apoptosis resistance of Ph + ALL cells. In our previous study, inhibiting histone deacetylase 1 (HDAC1) decreases the proliferation of Ph + ALL cells. However, little is known regarding how HDAC1 in BMSCs of Ph + ALL patients affects the imatinib (IM) resistance. Therefore, the present work examined the roles of HDAC1 in BMSCs. Overexpression of HDAC1 was found in BMSCs of Ph + ALL patients with IM resistance. In addition, the Ph + ALL cell line SUP-B15 was co-cultured with BMSCs after lentivirus transfection for regulating HDAC1 expression. Knockdown of HDAC1 within BMSCs elevated the IM-mediated SUP-B15 cell apoptosis, while increasing HDAC1 expression had an opposite effect. IL-6 in BMSCs, which is an important factor for the microenvironment-associated chemoresistance, showed evident up-regulation in HDAC1-upregulated BMSCs and down-regulation in HDAC1-downregulated BMSCs. While recombinant IL-6 (rIL-6) can reversed the sensitivity of SUP-B15 cells to IM induced by downregulating HDAC1 expression in BMSCs. HDAC1 showed positive regulation on IL-6 transcription and secretion. Moreover, IL-6 secretion induced by HDAC1 in BMSCs might enhance IM resistance in Ph + ALL cells. With regard to the underlying molecular mechanism, NF-κB, an important signal responsible for IL-6 transcription in BMSCs, mediated the HDAC1-regulated IL-6 expression. Collectively, this study facilitated to develop HDAC1 inhibitors based not only the corresponding direct anti-Ph + ALL activity but also the regulation of bone marrow microenvironment.

4.
Cytotherapy ; 25(7): 728-738, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36890092

RESUMEN

BACKGROUND AIMS: Recently, immune escape has been considered as a factor leading to relapse of acute myeloid leukemia (AML). In our previous study, heme oxygenase 1 (HO-1) proved to play an essential role in the proliferation and drug resistance of AML cells. In addition, recent studies by our group have shown that HO-1 is involved in immune escape in AML. Nevertheless, the specific mechanism by which HO-1 mediates immune escape in AML remains unclear. METHODS: In this study, we found that patients with AML and an overexpression of HO-1 had a high rate of recurrence. In vitro, overexpression of HO-1 attenuated the toxicity of natural killer (NK) cells to AML cells. Further study indicated that HO-1 overexpression inhibited human leukocyte antigen-C and reduced the cytotoxicity of NK cells to AML cells, leading to AML relapse. Mechanistically, HO-1 inhibited human leukocyte antigen-C expression by activating the JNK/C-Jun signaling pathway. RESULTS: In AML, HO-1 inhibits cytotoxicity of NK cells by inhibiting the expression of HLA-C, thus causing immune escape of AML cells. CONCLUSIONS: NK cell-mediated innate immunity is important for the fight against tumors, especially when acquired immunity is depleted and dysfunctional, and the HO-1/HLA-C axis can induce functional changes in NK cells in AML. Anti-HO-1 treatment can promote the antitumor effect of NK cells and may play an important role in the treatment of AML.


Asunto(s)
Hemo-Oxigenasa 1 , Leucemia Mieloide Aguda , Humanos , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Antígenos HLA-C/metabolismo , Leucemia Mieloide Aguda/terapia , Células Asesinas Naturales
5.
BMC Surg ; 23(1): 74, 2023 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-36997940

RESUMEN

BACKGROUND: This prospective randomized controlled study was designed to evaluate the effect of S-ketamine with sufentanil given intraoperatively and postoperatively on recovery of gastrointestinal (GI) function and postoperative pain in gynecological patients undergoing open abdomen surgery. METHODS: One hundred gynecological patients undergoing open abdomen surgery were randomized into an S-ketamine group (group S) or placebo group (0.9% saline; group C). Anesthesia was maintained with S-ketamine, sevoflurane, and remifentanil-propofol target-controlled infusion in group S and with sevoflurane and remifentanil-propofol target-controlled infusion in group C. All patients were connected to patient-controlled intravenous analgesia (PCIA) pump at the end of the surgery with sufentanil, ketorolac tromethamine, and tropisetron in group C and additional S-ketamine in group S. The primary outcome was the time of first postoperative flatus, and the secondary outcome was postoperative pain score of patients. Postoperative sufentanil consumption within the first postoperative 24 h and adverse events such as nausea and vomiting were recorded. RESULTS: The time of first postoperative flatus in group S was significantly shorter (mean ± SD, 50.3 ± 13.5 h) than that in group C (mean ± SD, 56.5 ± 14.3 h, p = 0.042). The patient's visual analog scale (VAS) pain score 24 h after surgery at rest was significantly lower in group S than in group C (p = 0.032). There were no differences in sufentanil consumption within the first postoperative 24 h, postoperative complications related to PCIA between the two groups. CONCLUSIONS: S-ketamine accelerated postoperative GI recovery and reduced 24 h postoperative pain in patients undergoing open gynecological surgery. TRIAL REGISTRATION: ChiCTR2200055180. Registered on 02/01/2022. It is a secondary analysis of the same trial.


Asunto(s)
Propofol , Sufentanilo , Humanos , Sufentanilo/uso terapéutico , Sufentanilo/efectos adversos , Remifentanilo/uso terapéutico , Propofol/uso terapéutico , Sevoflurano/uso terapéutico , Estudios Prospectivos , Flatulencia/inducido químicamente , Flatulencia/tratamiento farmacológico , Dolor Postoperatorio/tratamiento farmacológico
6.
J Transl Med ; 20(1): 394, 2022 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-36058936

RESUMEN

BACKGROUND: Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. Given the high relapse rate, more effective treatments are needed to improve clinical outcomes. We previously demonstrated that heme oxygenase 1 (HO1) is overexpressed in AML, while the functional roles of HO1 remain unclear. METHODS: Bioinformatics analysis and flow cytometry were conducted to assess the association between HO1 levels and immune cells or immune checkpoint/ligand molecules in AML patients. Primary natural killer (NK) cells were purified and subsequently co-cultured in vitro with transduced AML cells to determine the effects of HO1 expression on NK cell functions. AML mice models were established to investigate the effects of HO1 expression on cytotoxic effects of NK cells in vivo. The molecular mechanism was studied by flow cytometry, quantitative real-time PCR (qRT-PCR), western blotting, and immunoprecipitation. RESULTS: Bioinformatics analysis indicated a correlation between HO1 expression and the AML immune microenvironment. The present study findings indicated that HO1 specifically downregulates the expression of CD48, a ligand of the NK cell-activating receptor 2B4, thus decreasing the cytotoxic effect of NK cells. HO1 overexpression promoted tumor growth and inhibited the cytotoxic effect of NK cells in the AML mice model. Mechanistic investigations found that HO1 directly interacted with Sirt1 and increased its expression and deacetylase activity. With the overexpression of HO1, increased Sirt1 in AML cells enabled histone H3K27 deacetylation to suppress CD48 transcription and expression. Administration of Sirt1 inhibitor restored the expression of CD48. CONCLUSIONS: Collectively, HO1 promotes NK cell dysfunction in AML. Therefore, restoring NK cell function by inhibiting HO1 activity is a potential immunotherapeutic approach against AML.


Asunto(s)
Hemo-Oxigenasa 1 , Evasión Inmune , Leucemia Mieloide Aguda , Animales , Hemo-Oxigenasa 1/metabolismo , Células Asesinas Naturales , Leucemia Mieloide Aguda/metabolismo , Ligandos , Ratones , Sirtuina 1/metabolismo , Microambiente Tumoral
7.
Anticancer Drugs ; 31(2): 158-168, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31584454

RESUMEN

Although arsenic trioxide (ATO) treatment has transformed acute promyelocytic leukemia (APL) from the most fatal to the most curable hematological cancer, many high-risk APL patients who fail to achieve a complete molecular remission or relapse become resistant to ATO. Herein, we report that 7-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yloxy)-N-hydroxyheptanamide (CUDC-101) exhibits specific anticancer effects on APL and ATO-resistant APL in vitro and in vivo, while showing negligible cytotoxic effect on the noncancerous cells including normal CD34 cells and bone marrow mesenchymal stem cells from APL patients. Further mechanistic studies show that CUDC-101 triggers caspase-dependent degradation of the promyelocytic leukemia-retinoic acid receptor alpha fusion protein. As a result, APL and ATO-resistant APL cells undergo apoptosis upon CUDC-101 treatment and this apoptosis-inducing effect is even stronger than that of ATO. Finally, using a xenograft mouse model, we demonstrated that CUDC-101 significantly represses leukemia development in vivo. In conclusion, these results suggested that CUDC-101 can serve as a potential candidate drug for APL, particularly for ATO-resistant APL.


Asunto(s)
Trióxido de Arsénico/farmacología , Caspasas/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Ácidos Hidroxámicos/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Quinazolinas/farmacología , Receptor alfa de Ácido Retinoico/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor , Proliferación Celular , Femenino , Humanos , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pronóstico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Cell Physiol ; 234(4): 5252-5263, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30256411

RESUMEN

Resistance towards imatinib (IM) remains troublesome in treating many chronic myeloid leukemia (CML) patients. Heme oxygenase-1 (HO-1) is a key enzyme of antioxidative metabolism in association with cell resistance to apoptosis. Our previous studies have shown that overexpression of HO-1 resulted in resistance development to IM in CML cells, while the mechanism remains unclear. In the current study, the IM-resistant CML cells K562R indicated upregulation of some of the histone deacetylases (HDACs) compared with K562 cells. Therefore, we herein postulated HO-1 was associated with HDACs. Silencing HO-1 expression in K562R cells inhibited the expression of some HDACs, and the sensitivity to IM was increased. K562 cells transfected with HO-1 resisted IM and underwent obvious some HDACs. These findings related to the inhibitory effects of high HO-1 expression on the reactive oxygen species (ROS) signaling pathway that negatively regulated HDACs. Increased expression of HO-1 activated HDACs by inhibiting ROS production. In summary, HO-1, which is involved in the development of drug resistance in CML cells by regulating the expression of HDACs, is probably a novel target for improving CML therapy.


Asunto(s)
Antineoplásicos/farmacología , Hemo-Oxigenasa 1/metabolismo , Histona Desacetilasas/metabolismo , Mesilato de Imatinib/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Adulto , Resistencia a Antineoplásicos , Activación Enzimática , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/genética , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Adulto Joven
9.
Brain Behav Immun ; 71: 158-168, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29548993

RESUMEN

CCDC134 (coiled-coil domain containing 134), a cytokine-like molecule, was previously reported to exert antitumor effects by augmenting CD8+ T-cell mediated immunity. However, the dynamic changes in CCDC134 expression patterns in the spinal cord that may be involved in the progression of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, remains unclear. In this study, we found that CCDC134 expression was markedly increased in the spinal cord during the progression of EAE. Furthermore, we demonstrated that CCDC134 significantly reduced the severity and slowed the progression of EAE, which correlated with reduced spinal cord inflammation and demyelination. The underlying mechanism of CCDC134-induced effects involved inhibition of T helper (Th)-1 and Th17 cell differentiation and secretion of its key effector molecules IFN-γ and IL-17A via regulation of JAK/STAT signaling. These findings indicate that CCDC134 exerts potent anti-inflammatory effects through the selective modulation of pathogenic Th1 and Th17 cells by targeting critical signaling pathways. The study provides insights into the role of CCDC134 as a unique therapeutic agent for the treatment of autoimmune diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Proteínas de la Membrana/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/metabolismo , Transducción de Señal/efectos de los fármacos , Médula Espinal/metabolismo , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/fisiología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/fisiología
10.
Biochem Biophys Res Commun ; 490(2): 111-116, 2017 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-28571739

RESUMEN

CCDC134 might be an immune cytokine and plays important and complex roles in the process in vivo. It was proved to illustrate its potent antitumor effects by augmenting CD8+ T-cell-mediated immunity, but its role in the development of rheumatoid arthritis (RA) remains unclear. In this study, we demonstrated that development of adjuvant-induced arthritis and pro-inflammatory responses were more ameliorated in CCDC134-overexpressing transgenic mice than those in WT mice. The underlying mechanism of CCDC134-induced effects involved inhibition of T helper (Th) 1 and Th17 cell differentiation. These findings indicate that overexpression of CCDC134 exerts potent anti-inflammatory effects through selective modulation of pathogenic Th1 and Th17 cells, and might provide insights into the role of CCDC134 as a unique therapeutic agent for the treatment of rheumatoid arthritis.


Asunto(s)
Artritis Experimental/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Animales , Artritis Experimental/patología , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
11.
J Hazard Mater ; 467: 133707, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38335621

RESUMEN

Identifying the impact of pollutants on diseases is crucial. However, assessing the health risks posed by the interplay of multiple pollutants is challenging. This study introduces the concept of Pollutants Outcome Disease, integrating multidisciplinary knowledge and employing explainable artificial intelligence (AI) to explore the joint effects of industrial pollutants on diseases. Using lung cancer as a representative case study, an extreme gradient boosting predictive model that integrates meteorological, socio-economic, pollutants, and lung cancer statistical data is developed. The joint effects of industrial pollutants on lung cancer are identified and analyzed by employing the SHAP (Shapley Additive exPlanations) interpretable machine learning technique. Results reveal substantial spatial heterogeneity in emissions from CPG and ILC, highlighting pronounced nonlinear relationships among variables. The model yielded strong predictions (an R of 0.954, an RMSE of 4283, and an R2 of 0.911) and emphasized the impact of pollutant emission amounts on lung cancer responses. Diverse joint effects patterns were observed, varying in terms of patterns, regions (frequency), and the extent of antagonistic and synergistic effects among pollutants. The study provides a new perspective for exploring the joint effects of pollutants on diseases and demonstrates the potential of AI technology to assist scientific discovery.


Asunto(s)
Contaminantes Ambientales , Neoplasias Pulmonares , Humanos , Inteligencia Artificial , Aprendizaje Automático , Industrias , Neoplasias Pulmonares/inducido químicamente
12.
Sci Rep ; 14(1): 14012, 2024 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890346

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous hematological tumor with poor immunotherapy effect. This study was to develop a monocyte/macrophage-related prognostic risk score (MMrisk) and identify new therapeutic biomarkers for AML. We utilized differentially expressed genes (DEGs) in combination with single-cell RNA sequencing to identify monocyte/macrophage-related genes (MMGs). Eight genes were selected for the construction of a MMrisk model using univariate Cox regression analysis and LASSO regression analysis. We then validated the MMrisk on two GEO datasets. Lastly, we investigated the immunologic characteristics and advantages of immunotherapy and potential targeted drugs for MMrisk groups. Our study identified that the MMrisk is composed of eight MMGs, including HOPX, CSTB, MAP3K1, LGALS1, CFD, MXD1, CASP1 and BCL2A1. The low MMrisk group survived longer than high MMrisk group (P < 0.001). The high MMrisk group was positively correlated with B cells, plasma cells, CD4 memory cells, Mast cells, CAFs, monocytes, M2 macrophages, Endothelial, tumor mutation, and most immune checkpoints (PD1, Tim-3, CTLA4, LAG3). Furthermore, drug sensitivity analysis showed that AZD.2281, Axitinib, AUY922, ABT.888, and ATRA were effective in high-risk MM patients. Our research shows that MMrisk is a potential biomarker which is helpful to identify the molecular characteristics of AML immunology.


Asunto(s)
Leucemia Mieloide Aguda , Macrófagos , Monocitos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/mortalidad , Monocitos/inmunología , Monocitos/metabolismo , Pronóstico , Macrófagos/inmunología , Macrófagos/metabolismo , Femenino , Biomarcadores de Tumor/genética , Masculino , Persona de Mediana Edad , Inmunoterapia/métodos , Transcriptoma , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica
13.
Nat Commun ; 15(1): 5761, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38982055

RESUMEN

While protein aggregation's association with aging and age-related diseases is well-established, the specific proteins involved and whether dissolving them could alleviate aging remain unclear. Our research addresses this gap by uncovering the role of PKM2 aggregates in aging. We find that PKM2 forms aggregates in senescent cells and organs from aged mice, impairing its enzymatic activity and glycolytic flux, thereby driving cells into senescence. Through a rigorous two-step small molecule library screening, we identify two compounds, K35 and its analog K27, capable of dissolving PKM2 aggregates and alleviating senescence. Further experiments show that treatment with K35 and K27 not only alleviate aging-associated signatures but also extend the lifespan of naturally and prematurely aged mice. These findings provide compelling evidence for the involvement of PKM2 aggregates in inducing cellular senescence and aging phenotypes, and suggest that targeting these aggregates could be a promising strategy for anti-aging drug discovery.


Asunto(s)
Envejecimiento , Senescencia Celular , Proteínas de Unión a Hormona Tiroide , Animales , Envejecimiento/metabolismo , Ratones , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas Portadoras/metabolismo , Glucólisis , Hormonas Tiroideas/metabolismo , Agregado de Proteínas , Piruvato Quinasa/metabolismo , Ratones Endogámicos C57BL , Masculino
14.
Front Immunol ; 15: 1403272, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39040102

RESUMEN

Introduction: Granulocytic myeloid-derived suppressor cells (G-MDSCs) show fast recovery following allogeneic hematopoietic stem cell transplantation (allo-HSCT) constituting the major part of peripheral blood in the early phase. Although G-MDSCs mediate immune suppression through multiple mechanisms, they may also promote inflammation under specific conditions. Methods: G-MDSCs were isolated from 82 patients following allo-HSCT within 90 days after allo-HSCT, and their interactions with autologous CD3+ T-cells were examined. T-cell proliferation was assessed by flow cytometry following CFSE staining, while differentiation and interferon-γ secretion were characterized using chemokine receptor profiling and ELISpot assays, respectively. NK cell cytotoxicity was evaluated through co-culture with K562 cells. An aGVHD xenogeneic model in humanized mice was employed to study the in vivo effects of human leukocytes. Furthermore, transcriptional alterations in G-MDSCs were analyzed via RNA sequencing to investigate functional transitions. Results: G-MDSCs promoted inflammation in the early-stage, by facilitating cytokine secretion and proliferation of T cells, as well as their differentiation into pro-inflammatory T helper subsets. At day 28, patients with a higher number of G-MDSCs exhibited an increased risk of developing grades II-IV aGvHD. Besides, adoptive transfer of G-MDSCs from patients at day 28 into humanized mice exacerbated aGvHD. However, at day 90, G-MDSCs led to immunosuppression, characterized by upregulated expression of indoleamine 2,3-dioxygenase gene and interleukin-10 secretion, coupled with the inhibition of T cell proliferation. Furthermore, transcriptional analysis of G-MDSCs at day 28 and day 90 revealed that 1445 genes were differentially expressed. These genes were associated with various pathways, revealing the molecular signatures of early post-transplant differentiation in G-MDSCs. In addition, genes linked to the endoplasmic reticulum stress were upregulated in patients without aGvHD. The acquisition of immunosuppressive function by G-MDSCs may depend on the activation of CXCL2 and DERL1 genes. Conclusion: Our findings revealed the alteration in the immune characteristics of G-MDSCs within the first 90 days post-allo-HSCT. Moreover, the quantity of G-MDSCs at day 28 may serve as a predictive indicator for the development of aGvHD.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Supresoras de Origen Mieloide , Trasplante Homólogo , Humanos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Animales , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Ratones , Femenino , Masculino , Adulto , Persona de Mediana Edad , Antígenos HLA-DR/metabolismo , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/genética , Enfermedad Injerto contra Huésped/inmunología , Inflamación/inmunología , Adulto Joven , Granulocitos/inmunología , Granulocitos/metabolismo , Adolescente , Antígeno CD11b/metabolismo , Antígeno CD11b/inmunología
15.
Cell Death Differ ; 30(8): 1916-1930, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37419986

RESUMEN

Solute carrier family 25 member 51 (SLC25A51) was recently identified as the mammalian mitochondrial NAD+ transporter essential for mitochondria functions. However, the role of SLC25A51 in human disease, such as cancer, remains undefined. Here, we report that SLC25A51 is upregulated in multiple cancers, which promotes cancer cells proliferation. Loss of SLC25A51 elevates the mitochondrial proteins acetylation levels due to SIRT3 dysfunctions, leading to the impairment of P5CS enzymatic activity, which is the key enzyme in proline biogenesis, and the reduction in proline contents. Notably, we find fludarabine phosphate, an FDA-approved drug, is able to bind with and inhibit SLC25A51 functions, causing mitochondrial NAD+ decrease and proteins hyperacetylation, which could further synergize with aspirin to reinforce the anti-tumor efficacy. Our study reveals that SLC25A51 is an attractive anti-cancer target, and provides a novel drug combination of fludarabine phosphate with aspirin as a potential cancer therapy strategy.


Asunto(s)
Prolina , Sirtuina 3 , Animales , Humanos , Acetilación , Prolina/farmacología , Prolina/metabolismo , Mitocondrias/metabolismo , Sirtuina 3/metabolismo , Homeostasis , Mamíferos/metabolismo
16.
Mol Omics ; 19(4): 351-361, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-36892434

RESUMEN

Background: Individualized pain therapy conforms to the concept of precision medicine and contributes to adequate pain management after surgery. Preoperative biomarkers associated with postoperative pain may instruct anesthesiologists to improve personalized suitable analgesia. Therefore, it is essential to explore the association between preoperative proteins and postoperative acute pain using the proteomics platform. Methods: In this study, the 24 hours postoperative sufentanil consumption of 80 male patients with gastric cancer was ranked. Patients with sufentanil consumption in the lowest 12% were included in the sufentanil low consumption group, while patients with sufentanil consumption in the highest 12% were included in the sufentanil high consumption group. The secretion of serum proteins in both groups was analyzed using label-free proteomics technology. The results were validated by ELISA. Results: Proteomics identified 29 proteins that were significantly differentially expressed between groups. ELISA confirmed that secretion of TNC and IGFBP2 was down-regulated in the SLC group. The differential proteins were mainly extracellular and were involved in several terms, including calcium ion binding, laminin-1 binding, and so on. Pathway analysis showed that they were mainly enriched in focal adhesion and extracellular matrix-receptor interaction. The protein-protein interaction network analysis showed 22 proteins that interacted with other proteins. F13B had the strongest correlation with sufentanil consumption and its AUC value was 0.859. Conclusions: Several differential proteins are associated with postoperative acute pain and are involved in ECM-related processes, inflammation, and blood coagulation cascades. F13B may be a novel marker for postoperative acute pain. Our results may benefit postoperative pain management.


Asunto(s)
Dolor Agudo , Neoplasias Gástricas , Humanos , Masculino , Sufentanilo , Neoplasias Gástricas/cirugía , Proteómica , Analgesia Controlada por el Paciente/métodos , Dolor Postoperatorio/diagnóstico , Dolor Postoperatorio/etiología , Dolor Postoperatorio/terapia
17.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 31(1): 8-16, 2023 Feb.
Artículo en Zh | MEDLINE | ID: mdl-36765470

RESUMEN

OBJECTIVE: To explore the expression pattern and clinical significance of Integral membrane protein 2A(ITM2A) in drug resistant patients with chronic myeloid leukemia (CML). METHODS: The expression of ITM2A in CML was evaluated by qRT-PCR, Western blot and immunocytochemistry. In order to understand the possible biological effects of ITM2A, apoptosis, cell cycle and myeloid differentiation antigen expression of CML cells were detected by flow cytometry after over-expression of ITM2A. The nuderlying molecular mechanism of its biological effect was explored. RESULTS: The expression of ITM2A in bone marrow of CML resistant patients was significantly lower than that of sensitive patients and healthy donors(P<0.05). The CML resistant strain cell K562R was successfully constructed in vitro. The expression of ITM2A in the resistant strain was significantly lower than that in the sensitive strain(P<0.05). Overexpression of ITM2A in K562R cells increased the sensitivity of K562R cells to imatinib and blocked the cell cycle in G2 phase(P<0.05), but did not affect myeloid differentiation. Mechanistically, up-regulation of ITM2A reduced phosphorylation in ERK signaling (P<0.05). CONCLUSION: The expression of ITM2A was low in patients with drug resistance of CML, and the low expression of ITM2A may be the key factor of imatinib resistance in CML.


Asunto(s)
Antineoplásicos , Leucemia Mielógena Crónica BCR-ABL Positiva , Humanos , Antineoplásicos/farmacología , Apoptosis , Resistencia a Antineoplásicos , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Transducción de Señal
18.
Cell Death Dis ; 14(2): 98, 2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36759599

RESUMEN

During interphase, the newly duplicated pairs of centrosomes are held together by a centrosome linker, and the centrosome separation needs the disruption of this linker to induce the duplicated centrosomes separating into two distinct microtubule organization centers. The mechanism of regulating centrosome separation is however poorly understood. Here, we demonstrated that the phosphorylation of PHF5A at Y36 by the TrkA-ERK1/2-ABL1 cascade plays a critical role in regulating centrosome separation. PHF5A, a well-characterized spliceosome component, is enriched in the centrosome. The pY36-PHF5A promotes the interaction between CEP250 and Nek2A in a spliceosomal-independent manner, which leads to premature centrosome separation. Furthermore, the unmatured centrosome remodels the microtubule and subsequently regulates cell proliferation and migration. Importantly, we found that the phosphorylation cascade of TrkA-ERK1/2-ABL1-PHF5A is hyper-regulated in medulloblastoma. The inhibition of this cascade can induce senescence and restrict the proliferation of medulloblastoma. Our findings on this phosphorylation cascade in regulating centrosome separation could provide a series of potential targets for restricting the progress of medulloblastoma.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Humanos , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinasas Relacionadas con NIMA/metabolismo , Sistema de Señalización de MAP Quinasas , Meduloblastoma/metabolismo , Células HeLa , Centrosoma/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Microtúbulos/metabolismo , Neoplasias Cerebelosas/metabolismo , Autoantígenos/metabolismo , Transactivadores/metabolismo
19.
Front Immunol ; 14: 1133111, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37234153

RESUMEN

Modulation of surface T cell antigen receptor (TCR) expression is crucial for proper T cell development and maintenance of mature T cell function at steady state and upon stimulation. We previously determined that CCDC134 (coiled-coil domain containing 134), a cytokine-like molecule that served as a potential member of the γc cytokine family, contributes to antitumor responses by augmenting CD8+ T cell-mediated immunity. Here we show that T cell-specific deletion of Ccdc134 decreased peripheral mature CD4+ and CD8+ T cells, which resulted in impaired T cell homeostasis. Moreover, Ccdc134-deficient T cells exhibited an attenuated response to TCR stimulation in vitro, showing lower activation and proliferative capacity. This was further reflected in vivo, rendering mice refractory to T cell-mediated inflammatory and antitumor responses. More importantly, CCDC134 is associated with TCR signaling components, including CD3ϵ, and attenuated TCR signaling in Ccdc134-deficient T cells via altered CD3ϵ ubiquitination and degradation. Taken together, these findings suggest a role for CCDC134 as a positive regulator of TCR-proximal signaling and provide insight into the cell-intrinsic functional consequences of Ccdc134 deficiency in the attenuation of T cell-mediated inflammatory and antitumor responses.


Asunto(s)
Linfocitos T CD8-positivos , Transducción de Señal , Ratones , Animales , Receptores de Antígenos de Linfocitos T/metabolismo , Activación de Linfocitos , Citocinas/metabolismo
20.
Nat Sci Sleep ; 15: 903-913, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37954026

RESUMEN

Purpose: This study aims to evaluate the effect of S-ketamine on slow wave sleep (SWS) and the related changes in serum protein in gynecological patients after open abdomen surgery. Methods: This was a randomized controlled trial. One hundred gynecological patients undergoing open abdomen surgery were randomized into an S-ketamine group (group S) or placebo group (0.9% saline; group C). During operation, patients in group S received adjuvant S-ketamine infusion (0.2 mg·kg-1·h-1) while those in group C received 0.9% saline. All patients were connected to patient-controlled intravenous analgesia (PCIA) pump in the end of the surgery and the patients in group S with an additional S-ketamine in PCIA pump. Polysomnogram (PSG) was monitored during the next night after surgery with PCIA pump. Blood samples were collected for proteomic analysis at 6:00 AM after PSG monitoring. The primary outcome was the percentage of SWS (also known as stage 3 non-rapid eye movement sleep, stage N3) on the next night after surgery, and the secondary outcome was subjective sleep quality, pain scores, and the changes in serum proteomics. Results: Complete polysomnogram recordings were obtained from 64 study participants (31 in group C and 33 in group S). The administration of S-ketamine infusion resulted in a significant increase in the percentage of SWS/N3 compared to the control group (group C, median (IQR [range]), 8.9 (6.3, 12.5); group S, median (IQR [range]), 15.6 (12.4, 18.8), P<0.001). However, subjective evaluations of sleep quality revealed no significant variances between the two groups. The protein affected by S-ketamine was primarily associated with posttranslational modification, protein turnover, carbohydrate transport, and metabolism. Conclusion: In patients undergoing open gynecological surgery, S-ketamine enhanced the percentage of objective sleep of SWS during the next night after surgery. Additionally, there were differences observed in serum protein levels between the two groups. Trial Registration: ChiCTR2200055180. Registered on 02/01/2022.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA