Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Mol Cell Biochem ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38700746

ABSTRACT

Acute myeloid leukemia (AML) is a rare and heterogeneous disease. Over the past few decades, patient prognosis has improved with continuous improvements in treatment, but outcomes for some patients with primary drug resistance or relapse after treatment remain poor. Additional therapies to improve outcomes for these patients are urgently needed. FYB1 expression differs substantially between AML tissues and normal tissues. High FYB1 expression is correlated with poorer overall survival (OS), indicating that FYB1 may regulate AML progression. Therefore, understanding the effect of FYB1 on AML could improve the success rate of therapeutic approaches and prognosis for patients with AML. In this study, through analysis of large databases and both in vivo and in vitro experiments, we assessed the expression and role of FYB1 in AML and the relationship of FYB with patient prognosis. Downstream targets of the FYB1 gene were analyzed by RNA-seq. Database mining and in vitro experiments were used to further clarify the effect of the downstream target gelsolin-like actin-capping protein (CAPG) on AML cells and its relationship with patient prognosis. FYB1 expression was significantly higher in AML tissue and corresponded with a poor prognosis. FYB1 knockdown inhibited AML cell proliferation, promoted cell apoptosis, reduced cell adhesion capability and significantly reduced the tumor formation rate in mice. In addition, FYB1 knockdown induced a notable decrease in CAPG expression. The suppression of CAPG significantly inhibited cell proliferation and increased cell apoptosis. The conclusions of this study underscore the pivotal role of the FYB1/CAPG axis in promoting AML. We propose that the FYB1/CAPG axis could serve as a new thread in the development of therapeutic strategies for AML.

2.
Cancer Lett ; 591: 216882, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38636893

ABSTRACT

Super enhancers (SEs) are genomic regions comprising multiple closely spaced enhancers, typically occupied by a high density of cell-type-specific master transcription factors (TFs) and frequently enriched in key oncogenes in various tumors, including neuroblastoma (NB), one of the most prevalent malignant solid tumors in children originating from the neural crest. Cyclin-dependent kinase 5 regulatory subunit-associated protein 3 (CDK5RAP3) is a newly identified super-enhancer-driven gene regulated by master TFs in NB; however, its function in NB remains unclear. Through an integrated study of publicly available datasets and microarrays, we observed a significantly elevated CDK5RAP3 expression level in NB, associated with poor patient prognosis. Further research demonstrated that CDK5RAP3 promotes the growth of NB cells, both in vitro and in vivo. Mechanistically, defective CDK5RAP3 interfered with the UFMylation system, thereby triggering endoplasmic reticulum (ER) phagy. Additionally, we provide evidence that CDK5RAP3 maintains the stability of MEIS2, a master TF in NB, and in turn, contributes to the high expression of CDK5RAP3. Overall, our findings shed light on the molecular mechanisms by which CDK5RAP3 promotes tumor progression and suggest that its inhibition may represent a novel therapeutic strategy for NB.


Subject(s)
Cell Cycle Proteins , Gene Expression Regulation, Neoplastic , Neuroblastoma , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/metabolism , Animals , Cell Line, Tumor , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Endoplasmic Reticulum/metabolism , Enhancer Elements, Genetic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Proliferation , Mice, Nude , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Prognosis
3.
Cell Death Discov ; 10(1): 186, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649363

ABSTRACT

Neuroblastoma (NB) is a common childhood tumor with a high incidence worldwide. The regulatory role of RNA N6-methyladenosine (m6A) in gene expression has attracted significant attention, and the impact of methyltransferase-like 14 (METTL14) on tumor progression has been extensively studied in various types of cancer. However, the specific influence of METTL14 on NB remains unexplored. Using data from the Target database, our study revealed significant upregulation of METTL14 expression in high-risk NB patients, with strong correlation with poor prognosis. Furthermore, we identified ETS1 and YY1 as upstream regulators that control the expression of METTL14. In vitro experiments involving the knockdown of METTL14 in NB cells demonstrated significant inhibition of cell proliferation, migration, and invasion. In addition, suppressing METTL14 inhibited NB tumorigenesis in nude mouse models. Through MeRIP-seq and RNA-seq analyses, we further discovered that YWHAH is a downstream target gene of METTL14. Mechanistically, we observed that methylated YWHAH transcripts, particularly those in the 5' UTR, were specifically recognized by the m6A "reader" protein YTHDF1, leading to the degradation of YWHAH mRNA. Moreover, the downregulation of YWHAH expression activated the PI3K/AKT signaling pathway, promoting NB cell activity. Overall, our study provides valuable insights into the oncogenic effects of METTL14 in NB cells, highlighting its role in inhibiting YWHAH expression through an m6A-YTHDF1-dependent mechanism. These findings also suggest the potential utility of a biomarker panel for prognostic prediction in NB patients.

4.
BMC Cancer ; 24(1): 220, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38365636

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is a relatively prevalent primary tumor of the central nervous system in children, characterized by its high malignancy and mortality rates, along with the intricate challenges of achieving complete surgical resection. Recently, an increasing number of studies have focused on the crucial role of super-enhancers (SEs) in the occurrence and development of GBM. This study embarks on the task of evaluating the effectiveness of MZ1, an inhibitor of BRD4 meticulously designed to specifically target SEs, within the intricate framework of GBM. METHODS: The clinical data of GBM patients was sourced from the Chinese Glioma Genome Atlas (CGGA) and the Gene Expression Profiling Interactive Analysis 2 (GEPIA2), and the gene expression data of tumor cell lines was derived from the Cancer Cell Line Encyclopedia (CCLE). The impact of MZ1 on GBM was assessed through CCK-8, colony formation assays, EdU incorporation analysis, flow cytometry, and xenograft mouse models. The underlying mechanism was investigated through RNA-seq and ChIP-seq analyses. RESULTS: In this investigation, we made a noteworthy observation that MZ1 exhibited a substantial reduction in the proliferation of GBM cells by effectively degrading BRD4. Additionally, MZ1 displayed a notable capability in inducing significant cell cycle arrest and apoptosis in GBM cells. These findings were in line with our in vitro outcomes. Notably, MZ1 administration resulted in a remarkable decrease in tumor size within the xenograft model with diminished toxicity. Furthermore, on a mechanistic level, the administration of MZ1 resulted in a significant suppression of pivotal genes closely associated with cell cycle regulation and epithelial-mesenchymal transition (EMT). Interestingly, our analysis of RNA-seq and ChIP-seq data unveiled the discovery of a novel prospective oncogene, SDC1, which assumed a pivotal role in the tumorigenesis and progression of GBM. CONCLUSION: In summary, our findings revealed that MZ1 effectively disrupted the aberrant transcriptional regulation of oncogenes in GBM by degradation of BRD4. This positions MZ1 as a promising candidate in the realm of therapeutic options for GBM treatment.


Subject(s)
Brain Neoplasms , Bromodomain Containing Proteins , Glioblastoma , Animals , Child , Humans , Mice , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Bromodomain Containing Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Prospective Studies , Syndecan-1/antagonists & inhibitors , Transcription Factors/genetics
5.
Carcinogenesis ; 45(6): 424-435, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38302114

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is a highly aggressive hematologic malignancy originating from T progenitor cells. It accounts for 15% of childhood and 25% of adult ALL cases. GNE-987 is a novel chimeric molecule developed using proteolysis-targeting chimeras (PROTAC) technology for targeted therapy. It consists of a potent inhibitor of the bromodomain and extraterminal (BET) protein, as well as the E3 ubiquitin ligase Von Hippel-Lindau (VHL), which enables the effective induction of proteasomal degradation of BRD4. Although GNE-987 has shown persistent inhibition of cell proliferation and apoptosis, its specific antitumor activity in T-ALL remains unclear. In this study, we aimed to investigate the molecular mechanisms underlying the antitumor effect of GNE-987 in T-ALL. To achieve this, we employed technologies including RNA sequencing (RNA-seq), chromatin immunoprecipitation sequencing (ChIP-seq) and CUT&Tag. The degradation of BET proteins, specifically BRD4, by GNE-987 has a profound impact on T-ALL cell. In in vivo experiments, sh-BRD4 lentivirus reduced T-ALL cell proliferation and invasion, extending the survival time of mice. The RNA-seq and CUT&Tag analyses provided further insights into the mechanism of action of GNE-987 in T-ALL. These analyses revealed that GNE-987 possesses the ability to suppress the expression of various genes associated with super-enhancers (SEs), including lymphoblastic leukemia 1 (LCK). By targeting these SE-associated genes, GNE-987 effectively inhibits the progression of T-ALL. Importantly, SE-related oncogenes like LCK were identified as critical targets of GNE-987. Based on these findings, GNE-987 holds promise as a potential novel candidate drug for the treatment of T-ALL.


Subject(s)
Apoptosis , Cell Proliferation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Transcription Factors , Xenograft Model Antitumor Assays , Humans , Animals , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Cell Proliferation/drug effects , Apoptosis/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Enhancer Elements, Genetic , Bromodomain Containing Proteins
6.
Cancer Cell Int ; 24(1): 81, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38383388

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is a malignancy of the hematopoietic system, and childhood AML accounts for about 20% of pediatric leukemia. ANP32B, an important nuclear protein associated with proliferation, has been found to regulate hematopoiesis and CML leukemogenesis by inhibiting p53 activity. However, recent study suggests that ANP32B exerts a suppressive effect on B-cell acute lymphoblastic leukemia (ALL) in mice by activating PU.1. Nevertheless, the precise underlying mechanism of ANP32B in AML remains elusive. RESULTS: Super enhancer related gene ANP32B was significantly upregulated in AML patients. The expression of ANP32B exhibited a negative correlation with overall survival. Knocking down ANP32B suppressed the proliferation of AML cell lines MV4-11 and Kasumi-1, along with downregulation of C-MYC expression. Additionally, it led to a significant decrease in H3K27ac levels in AML cell lines. In vivo experiments further demonstrated that ANP32B knockdown effectively inhibited tumor growth. CONCLUSIONS: ANP32B plays a significant role in promoting tumor proliferation in AML. The downregulation of ANP32B induces cell cycle arrest and promotes apoptosis in AML cell lines. Mechanistic analysis suggests that ANP32B may epigenetically regulate the expression of MYC through histone H3K27 acetylation. ANP32B could serve as a prognostic biomarker and potential therapeutic target for AML patients.

7.
Medicine (Baltimore) ; 102(31): e34170, 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37543834

ABSTRACT

BACKGROUND: Ranibizumab addition may benefit to improve the efficacy in patients with diabetic retinopathy than only photocoagulation, and this meta-analysis aims to explore the impact of ranibizumab addition on efficacy for diabetic retinopathy. METHODS: PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases were systematically searched, and we included randomized controlled trials assessing the effect of ranibizumab addition on patients with diabetic retinopathy for this meta-analysis. RESULTS: Six randomized controlled trials were finally included in the meta-analysis. Overall, compared with control intervention for diabetic retinopathy, ranibizumab addition showed significantly increased number of neovascularization area reduction (OR = 4.20; 95% CI = 1.47-12.02; P = .007) and reduced fluorescein leakage (MD = -2.53; 95% CI = -3.31 to -1.75; P < .00001), but showed no obvious impact on neovascularization area (MD = -1.80; 95% CI = -3.68 to 0.08; P = .06), photocoagulation retreatment (OR = 1.03; 95% CI = 0.47-2.27; P = .94) or adverse events (OR = 1.45; 95% CI = 0.49-4.29; P = .50). CONCLUSIONS: Ranibizumab combined with photocoagulation is effective to improve efficacy for diabetic retinopathy.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Humans , Ranibizumab/therapeutic use , Diabetic Retinopathy/complications , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/surgery , Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Vascular Endothelial Growth Factor A/therapeutic use , Laser Coagulation , Intravitreal Injections , Diabetes Mellitus/drug therapy
8.
Jpn J Infect Dis ; 76(6): 343-350, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37518069

ABSTRACT

Mycobacterium tuberculosis (M.tb) infection causes the communicable disease tuberculosis (TB), a major disease and one of the leading causes of death worldwide. The protein encoded by the region of deletion (RD) in M.tb mediates the pathogenic properties of M.tb by inducing an inflammatory response or disrupting host cell metabolism. We cloned and purified the Rv2653 protein from RD13 to explore its regulatory effects on host macrophages. We found that Rv2653 promoted glycolysis and upregulated the expression of key glycolytic enzymes, namely, hexokinase 2 (HK2) and lactate dehydrogenase-A (LDHA) in human leukemia monocytic (THP1) cells. Furthermore, the induction of glycolysis by Rv2653 contributes to the activation of the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome. Rv2653 activated the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, and the mTORC1 inhibitor NR1 blocked Rv2653-induced HK2, LDHA, and NLRP3 expression. siRNA interfering with HK2 or LDHA significantly inhibited the activation of NLRP3 inflammasome by Rv2653, blocked Rv2653-triggered inflammatory factors interleukin (IL)-1ß, IL-6, tumor necrosis factor (TNF)-α, reactive oxygen species (ROS), and nitric oxide (NO), and promoted the survival of Bacillus Calmette-Guerin (BCG) in THP1 cells. Overall, Rv2653 promoted glycolysis by activating the mTORC1 signaling pathway, activating the NLRP3 inflammasome, and releasing inflammatory factors, ultimately inhibiting the intracellular survival of BCG in THP1 cells. Therefore, we revealed that anti-M.tb immune mechanisms induced by Rv2653 contribute to the development of new anti-TB strategies.


Subject(s)
Inflammasomes , Mycobacterium tuberculosis , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mycobacterium tuberculosis/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Inflammation , Glycolysis
9.
Diabetes Metab Res Rev ; 39(4): e3620, 2023 05.
Article in English | MEDLINE | ID: mdl-36738300

ABSTRACT

AIMS: It is acknowledged that aberrant liver immunity contributes to the development of type 2 diabetes mellitus (T2DM). Mucosal-associated invariant T (MAIT) cells, an innate-like T-cell subset, are enriched in the human liver. Nevertheless, the characterisation and potential role of hepatic MAIT cells in T2DM remain unclear. MATERIALS AND METHODS: Fourteen newly diagnosed T2DM subjects and 15 controls received liver biopsy. The frequency and cytokine production of MAIT cells were analysed by flow cytometry. The expression of genes involved in glucose metabolism was determined in HepG2 cells co-cultured with hepatic MAIT cells. RESULTS: Compared with controls, hepatic MAIT cell frequency was significantly increased in T2DM patients (24.66% vs. 14.61%, p = 0.001). There were more MAIT cells producing interferon-γ (IFN-γ, 60.49% vs. 33.33%, p = 0.021) and tumour necrosis factor-α (TNF-α, 46.84% vs. 5.91%, p = 0.021) in T2DM than in controls, whereas their production of interleukin 17 (IL-17) was comparable (15.25% vs. 4.55%, p = 0.054). Notably, an IFN-γ+ TNF-α+ IL-17+/- producing MAIT cell subset was focussed, which showed an elevated proportion in T2DM (42.66% vs. 5.85%, p = 0.021) and positively correlated with plasma glucose levels. A co-culture experiment further indicated that hepatic MAIT cells from T2DM upregulated the gene expression of pyruvate carboxylase, a key molecule involved in gluconeogenesis, in HepG2 cells, and this response was blocked with neutralising antibodies against IFN-γ and TNF-α. CONCLUSIONS: Our data implicate an increased Th1-like MAIT cell subset in the liver of newly diagnosed T2DM subjects, which induces hyperglycaemia by promoting hepatic gluconeogenesis. It provides novel insights into the immune regulation of metabolic homoeostasis. CLINICAL TRIAL REGISTRATION NUMBER: NCT03296605 (registered at www. CLINICALTRIALS: gov on 12 October 2018).


Subject(s)
Diabetes Mellitus, Type 2 , Mucosal-Associated Invariant T Cells , Humans , Mucosal-Associated Invariant T Cells/physiology , Interleukin-17 , Tumor Necrosis Factor-alpha , Gluconeogenesis , Liver
10.
Nat Metab ; 5(2): 314-330, 2023 02.
Article in English | MEDLINE | ID: mdl-36717749

ABSTRACT

The accumulation of acidic metabolic waste products within the tumor microenvironment inhibits effector functions of tumor-infiltrating lymphocytes (TILs). However, it remains unclear how an acidic environment affects T cell metabolism and differentiation. Here we show that prolonged exposure to acid reprograms T cell intracellular metabolism and mitochondrial fitness and preserves T cell stemness. Mechanistically, elevated extracellular acidosis impairs methionine uptake and metabolism via downregulation of SLC7A5, therefore altering H3K27me3 deposition at the promoters of key T cell stemness genes. These changes promote the maintenance of a 'stem-like memory' state and improve long-term in vivo persistence and anti-tumor efficacy in mice. Our findings not only reveal an unexpected capacity of extracellular acidosis to maintain the stem-like properties of T cells, but also advance our understanding of how methionine metabolism affects T cell stemness.


Subject(s)
Acidosis , Neoplasms , Animals , Mice , Neoplasms/metabolism , Cell Differentiation , Tumor Microenvironment , Acidosis/metabolism , Carbon
11.
Diabetes Metab ; 49(2): 101397, 2023 03.
Article in English | MEDLINE | ID: mdl-36283619

ABSTRACT

AIM: The association of bone turnover with the incidence and progression of nonalcoholic fatty liver disease (NAFLD) is unclear. We aimed to evaluate serum levels of bone turnover markers in relation to NAFLD and nonalcoholic hepatic steatohepatitis (NASH). METHODS: Two cohorts were involved in our study. For the first cohort, 370 participants without NAFLD were retrospectively recruited and followed up for incident NAFLD according to ultrasound. For the second cohort, 562 subjects who underwent liver biopsy were included and grouped into non-NAFLD, non-NASH or NASH according to the NASH Clinical Research Network system. The bone turnover markers osteocalcin, C-terminal telopeptide (CTX) and N-terminal propeptide of type-1 procollagen (P1NP) were measured. RESULTS: Baseline osteocalcin was significantly lower in subjects who developed NAFLD (13.93 [11.03;16.39] versus 18.24 [15.45;22.47] ng/ml, P < 0.001), with a median of 26.4 months of follow-up. Low levels of osteocalcin, but not CTX or P1NP, was an independent predictor of incident NAFLD (OR 0.755 [95%CI 0.668; 0.855] P < 0.001). Moreover, the osteocalcin level was negatively associated with the degree of liver steatosis. Furthermore, subjects with NASH had significantly lower osteocalcin than non-NASH and non-NAFLD group (13.28 [10.49;16.59] versus 14.91 [12.45;18.09] versus 18.21 [15.04;22.05] ng/ml, all P < 0.001). A low osteocalcin level was an independent risk factor for NASH (OR for highest versus lowest quartile: 0.282 [0.147;0.543] P < 0.001). CONCLUSION: Low level of osteocalcin, but not CTX or P1NP, was associated with NAFLD and NASH, indicating its potential role as an important endocrine regulator of hepatic energy metabolism.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Liver/metabolism , Non-alcoholic Fatty Liver Disease/epidemiology , Osteocalcin , Retrospective Studies , Risk Factors
12.
BMJ Open ; 12(12): e066243, 2022 12 29.
Article in English | MEDLINE | ID: mdl-36581406

ABSTRACT

OBJECTIVE: Evidence suggests that both serum uric acid (SUA) and high-density lipoprotein cholesterol (HDL-C) are risk factors for chronic kidney disease (CKD). The SUA-to-HDL-C ratio (UHR) has recently attracted attention as a new biomarker to evaluate the role between inflammatory and anti-inflammatory substances. Thus, we explored the association between UHR and CKD in a large Chinese population. DESIGN: A cross-sectional study. SETTING: Annual health check-up population in Nanjing. PARTICIPANTS: 19 458 individuals who underwent an annual health check-up in 2019 were included in our study. MAIN OUTCOME MEASURE: CKD was diagnosed according to an estimated glomerular filtration rate (eGFR) <60 mL/min/1.73 m2. RESULTS: Correlation analysis showed that UHR was negatively associated with eGFR after adjusting for confounding factors (r=-0.34). In addition, participants in the highest quartile of UHR had a higher risk of CKD than those in the lowest quartiles (OR=9.28, p<0.001). CONCLUSION: We found that high UHR values were positively associated with CKD risk in health check-up population. An increased UHR may be a useful measure by which to assess CKD risk in the preclinical stage.


Subject(s)
Renal Insufficiency, Chronic , Uric Acid , Humans , Cross-Sectional Studies , Glomerular Filtration Rate , Renal Insufficiency, Chronic/diagnosis , Risk Factors , Cholesterol, HDL
13.
Methods Mol Biol ; 2530: 109-123, 2022.
Article in English | MEDLINE | ID: mdl-35761045

ABSTRACT

Thiolated/selenolated amino acids are irreplaceable despite their rare abundance in proteins. They play critical roles in regulating the conformation and function of proteins and peptide design as well as bioconjugation. Furthermore, ß-thiolated/selenolated amino acids are important motifs in native chemical ligation-dechalcogenation strategy for protein synthesis. However, a universal method to access enantiopure ß-thiolated/selenolated amino acids has not been reported. Herein, we developed a practical strategy for the preparation of a variety of enantiopure ß-thiolated/selenolated amino acids via photoredox-catalyzed Giese reaction.


Subject(s)
Amino Acids , Light , Amino Acids/chemistry , Catalysis , Peptides/chemistry , Proteins
14.
Sci Signal ; 15(723): eabk3083, 2022 03.
Article in English | MEDLINE | ID: mdl-35230873

ABSTRACT

CARD11 acts as a gatekeeper for adaptive immune responses after T cell or B cell antigen receptor (TCR/BCR) ligation on lymphocytes. PKCθ/ß-catalyzed phosphorylation of CARD11 promotes the assembly of the CARD11-BCL10-MALT1 (CBM) complex and lymphocyte activation. Here, we demonstrated that PKCθ/ß-dependent CARD11 phosphorylation also suppressed CARD11 functions in T or B cells. Through mass spectrometry-based proteomics analysis, we identified multiple constitutive and inducible CARD11 phosphorylation sites in T cells. We demonstrated that a single TCR- or BCR-inducible phosphorylation on Ser893 in the carboxyl terminus of CARD11 prevented the activation of the transcription factor NF-κB, the kinase JNK, and the protease MALT1. Moreover, CARD11 Ser893 phosphorylation sensitized BCR-addicted lymphoma cells to toxicity induced by Bruton's tyrosine kinase (BTK) inhibitors. Phosphorylation of Ser893 in CARD11 by PKCθ controlled the strength of CARD11 scaffolding by impairing the formation of the CBM complex. Thus, PKCθ simultaneously catalyzes both stimulatory and inhibitory CARD11 phosphorylation events, which shape the strength of CARD11 signaling in lymphocytes.


Subject(s)
CARD Signaling Adaptor Proteins , Serine , B-Cell CLL-Lymphoma 10 Protein/genetics , B-Cell CLL-Lymphoma 10 Protein/metabolism , B-Lymphocytes/metabolism , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation
15.
Trends Cancer ; 8(3): 159-160, 2022 03.
Article in English | MEDLINE | ID: mdl-35115254

ABSTRACT

Different subsets of tumor-infiltrating dendritic cells (DCs) influence immune response and tolerance in cancer settings. Duong et al. discovered that conventional DC2 subset (cDC2s) can differentiate into stimulatory interferon-stimulated gene (ISG)+ DCs by type I interferon (IFN-I) produced in regressor tumors and acquire and present tumor-derived peptide-MHC (pMHC) class I complexes to increase the protective antitumor CD8+ T cell response.


Subject(s)
CD8-Positive T-Lymphocytes , Dendritic Cells , Bandages , Humans , Immune Tolerance
16.
Cell Mol Life Sci ; 79(2): 112, 2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35099607

ABSTRACT

T cell activation initiates protective adaptive immunity, but counterbalancing mechanisms are critical to prevent overshooting responses and to maintain immune homeostasis. The CARD11-BCL10-MALT1 (CBM) complex bridges T cell receptor engagement to NF-κB signaling and MALT1 protease activation. Here, we show that ABIN-1 is modulating the suppressive function of A20 in T cells. Using quantitative mass spectrometry, we identified ABIN-1 as an interactor of the CBM signalosome in activated T cells. A20 and ABIN-1 counteract inducible activation of human primary CD4 and Jurkat T cells. While A20 overexpression is able to silence CBM complex-triggered NF-κB and MALT1 protease activation independent of ABIN-1, the negative regulatory function of ABIN-1 depends on A20. The suppressive function of A20 in T cells relies on ubiquitin binding through the C-terminal zinc finger (ZnF)4/7 motifs, but does not involve the deubiquitinating activity of the OTU domain. Our mechanistic studies reveal that the A20/ABIN-1 module is recruited to the CBM complex via A20 ZnF4/7 and that proteasomal degradation of A20 and ABIN-1 releases the CBM complex from the negative impact of both regulators. Ubiquitin binding to A20 ZnF4/7 promotes destructive K48-polyubiquitination to itself and to ABIN-1. Further, after prolonged T cell stimulation, ABIN-1 antagonizes MALT1-catalyzed cleavage of re-synthesized A20 and thereby diminishes sustained CBM complex signaling. Taken together, interdependent post-translational mechanisms are tightly controlling expression and activity of the A20/ABIN-1 silencing module and the cooperative action of both negative regulators is critical to balance CBM complex signaling and T cell activation.


Subject(s)
DNA-Binding Proteins/physiology , T-Lymphocytes/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/physiology , B-Cell CLL-Lymphoma 10 Protein/metabolism , CARD Signaling Adaptor Proteins/metabolism , Cells, Cultured , Guanylate Cyclase/metabolism , HEK293 Cells , Humans , Jurkat Cells , Lymphocyte Activation/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Multiprotein Complexes/metabolism , NF-kappa B/metabolism , Protein Binding , RNA Interference/immunology , Signal Transduction/physiology , T-Lymphocytes/immunology
17.
Medicine (Baltimore) ; 100(38): e27224, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34559115

ABSTRACT

BACKGROUNG: Tumor microenvironment (TME) has gradually emerged as an important research topic in the fight against cancer. The immune system is a major contributing factor in TME, and investigations have revealed that tumors are partially infiltrated with numerous immune cell subsets. METHOD: We obtained transcriptome RNA-seq data from the the Cancer Genome Atlas databases for 521 patients with colon adenocarcinoma (COAD). ESTIMATE algorithms are then used to estimate the fraction of stromal and immune cells in COAD samples. RESULT: A total of 1109 stromal-immune score-related differentially expressed genes were identified and used to generate a high-confidence protein-protein interaction network and univariate COX regression analysis. C-X-C motif chemokine 10 (CXCL10) was identified as the core gene by intersection analysis of data from protein-protein interaction network and univariate COX regression analysis. Then, for CXCL10, we performed gene set enrichment analysis, survival analysis and clinical analysis, and we used CIBERSORT algorithms to estimate the proportion of tumor-infiltrating immune cells in COAD samples. CONCLUSION: We discovered that CXCL10 levels could be effective for predicting the prognosis of COAD patients as well as a clue that the status of TME is transitioning from immunological to metabolic activity, which provided additional information for COAD therapies.


Subject(s)
Chemokine CXCL10/analysis , Chemokine CXCL10/pharmacology , Colonic Neoplasms/complications , Tumor Microenvironment , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Chemokine CXCL10/blood , Colonic Neoplasms/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis
18.
Article in English | MEDLINE | ID: mdl-33014113

ABSTRACT

BACKGROUND: Oxaliplatin (L-OHP) resistance is a major obstacle to the effective treatment of colorectal cancer. The resistance mechanism(s) of colorectal tumors to L-OHP may be related to the regulation of ERCC1 by cancer-expressed miRNAs, but no in-depth studies on the miRNAs that affect drug resistance have been performed. Curcumin (Cur) can reverse the drug resistance of cancer cells, but its effects on ERCC1 expression and miRNA profiles in colorectal cancer have not been studied. METHODS: To study the regulation effect of curcumin on ERCC1 expression and its effects on miRNAs, the L-OHP-resistant colorectal cancer cell line HCT116/L-OHP was established. MTT assays were used to evaluate cell proliferation. Flow cytometry was used to investigate apoptotic induction. Western blot and RT-PCR analysis were used to evaluate the expression of drug-associated ERCC1, Bcl-2, GST-π, MRP, P-gp, and survivin. RESULTS: HCT116//L-OHP cell lines were successfully established. The combination of L-OHP and curcumin could reduce L-OHP resistance in vitro. In addition, combination therapy inhibited the expression of ERCC1, Bcl-2, GST-π, MRP, P-gp, and survivin at the mRNA and protein level. Curcumin was found to inhibit ERCC1 through its ability to modulate miR-409-3p. CONCLUSION: Curcumin can overcome L-OHP resistance in colorectal cancer cells through its effects on miR-409-3p mediated ERCC1 expression.

19.
J Am Chem Soc ; 142(33): 14201-14209, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32787248

ABSTRACT

Sulfur and selenium occupy a distinguished position in biology owing to their redox activities, high nucleophilicity, and acyl transfer capabilities. Thiolated/selenolated amino acids, including cysteine, selenocysteine, and their derivatives, play critical roles in regulating the conformation and function of proteins and serve as an important motif for peptide design and bioconjugation. Unfortunately, a general and concise method to attain enantiopure ß-thiolated/selenolated amino acids remains an unsolved problem. Herein, we present a photoredox-catalyzed asymmetric method for the preparation of enantiopure ß-thiolated/selenolated amino acids using a simple chiral auxiliary, which controls the diastereoselectivity of the key alkylation step and acts as an orthogonal protecting group in the subsequent peptide synthesis. Our protocol can be used to prepare a wide range of ß-thiolated/selenolated amino acids on a gram scale, which would otherwise be difficult to obtain using conventional methods. The effect of our chemistry was further highlighted and validated through the preparation of a series of peptidyl thiol/selenol analogues, including cytochrome c oxidase subunit protein 7C and oxytocin.


Subject(s)
Amino Acids/chemical synthesis , Selenium/chemistry , Sulfhydryl Compounds/chemistry , Amino Acids/chemistry , Catalysis , Molecular Conformation , Oxidation-Reduction , Photochemical Processes
20.
Eur J Pharmacol ; 885: 173495, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32861661

ABSTRACT

GDF11 has been reported to play a critical role in rejuvenating hypertrophy heart, skeletal muscle, and blood vessel regeneration in aged mice. Whether GDF11 can regulate autophagy in cardiomyocytes remains largely unknown. Thus, the purpose of the present study was to investigate the effects of GDF11 on cardiomyocyte autophagy induced by hypoxia, in addition to the underlying mechanisms. By using the MTT assay, Flow cytometry assay, LIVE/DEAD® Viability/Cytotoxicity Kit Stains and TUNEL assay, we found that exogenous GDF11 decreased apoptosis caused by prolonged hypoxia in cardiomyocytes. The expression of GDF11 was decreased obviously both in the cardiac tissue of myocardial infarction mice and the hypoxia treated cardiomyocytes. Protein levels of cleaved caspase-3, p-AMPK, SQSTM1, LC3B-I/II and GDF11 were detected by western blot. Autophagosomes and autolysosomes were identified by confocal laser microscopy after transfecting with the mRFP-eGFP-LC3 plasmids. Antibody against GDF11 (anti-GDF11) was used to inhibit the function of GDF11. At the molecular level, exogenous GDF11 increased AMPK function and enhanced autophagy activity. Anti-GDF11 inhibited autophagy and aggravated hypoxia-induced apoptosis in cardiomyocytes. Thus, GDF11 might be a potential target for myocardial infarction therapy.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Bone Morphogenetic Proteins/genetics , Cell Hypoxia/drug effects , Growth Differentiation Factors/genetics , Myocytes, Cardiac/drug effects , Animals , Antibodies, Blocking/pharmacology , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/drug effects , Electrocardiography/drug effects , Growth Differentiation Factors/antagonists & inhibitors , Growth Differentiation Factors/drug effects , Lysosomes/drug effects , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Phagosomes/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...