Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.301
1.
Brain Behav ; 14(3): e3465, 2024 Mar.
Article En | MEDLINE | ID: mdl-38468469

BACKGROUND: SP gene family, consisting of SP100, SP110, SP140, and SP140L, has been implicated in the initiation and advancement of numerous malignancies. Nevertheless, their clinical significance in glioma remains incompletely understood. METHOD: Expression levels and prognostic significance of SP family members were evaluated in the TCGA and CGGA datasets. Multifactorial analysis was used to identify SP gene family members that can independently impact the prognosis of glioma patients. A SP140-based predictive risk model/nomogram was developed in TCGA dataset and validated in CGGA dataset. The model's performance was evaluated through receiver operating characteristic (ROC) curves, calibration plots, and decision curve analyses. Phenotypic associations of SP140 and TRIM22 were examined through CancerSEA and TIMER. The effect of SP140 inhibitor in glioma progress and TRIM22/PI3K/AKT signaling pathway was confirmed in U251/U87 glioma cells. RESULTS: The SP family members exhibited elevated expression in gliomas and were negatively correlated with prognosis. SP140 emerged as an independent prognostic factor, and a SP140-based nomogram/predictive risk model demonstrated high accuracy. SP140 inhibitor, GSK761, lead to the suppression of TRIM22 expression and the PI3K/AKT signaling pathway. GSK761 also restrain glioma proliferation, migration, and invasion. Furthermore, SP140 and TRIM22 coexpressed in glioma cells with high level of vascular proliferation, TRIM22 is closely associated with the immune cell infiltration. CONCLUSION: SP140-based nomogram proved to be a practical tool for predicting the survival of glioma patients. SP140 inhibitor could suppress glioma progress via TRIM22/PI3K/AKT signaling pathway.


Glioma , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Proliferation , Signal Transduction , Glioma/drug therapy , Glioma/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/pharmacology , Repressor Proteins/metabolism , Minor Histocompatibility Antigens/pharmacology , Transcription Factors , Antigens, Nuclear/metabolism , Antigens, Nuclear/pharmacology
2.
Mol Biol Rep ; 51(1): 61, 2024 Jan 03.
Article En | MEDLINE | ID: mdl-38170326

BACKGROUND: Breast adenocarcinoma cells (MCF-7) are characterized by the overexpression of apoptotic marker genes and proliferative cell nuclear antigen (PCNA), which promote cancer cell proliferation. Thymol, derived from Nigella sativa (NS), has been investigated for its potential anti-proliferative and anticancer properties, especially its ability to suppress Cyclin D1 and PCNA expression, which are crucial in the proliferation of cancer cells. METHODS: The cytotoxicity of thymol on MCF-7 cells was assessed using 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) and lactate dehydrogenase (LDH) release methods. Thymol was tested at increasing concentrations (0-1000 µM) to evaluate its impact on MCF-7 cell growth. Additionally, Cyclin D1 and PCNA gene expression in thymol-treated and vehicle control groups of MCF-7 were quantified using real-time Polymerase Chain Reaction (RT-qPCR). Protein-ligand interactions were also investigated using the CB-Dock2 server. RESULTS: Thymol significantly inhibited MCF-7 cell growth, with a 50% inhibition observed at 200 µM. The gene expression of Cyclin D1 and PCNA was down-regulated in the thymol-treated group relative to the vehicle control. The experimental results were verified through protein-ligand interaction investigations. CONCLUSIONS: Thymol, extracted from NS, demonstrated specific cytotoxic effects on MCF-7 cells by suppressing the expression of Cyclin D1 and PCNA, suggesting its potential as an effective drug for MCF-7. However, additional in vivo research is required to ascertain its efficacy and safety in medical applications.


Breast Neoplasms , Nigella sativa , Humans , Female , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , MCF-7 Cells , Breast Neoplasms/genetics , Thymol/pharmacology , Thymol/therapeutic use , Nigella sativa/metabolism , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Antigens, Nuclear/therapeutic use , Cyclin D1/genetics , Cyclin D1/metabolism , Down-Regulation , Ligands , Cell Proliferation
3.
Sci Rep ; 14(1): 1188, 2024 01 12.
Article En | MEDLINE | ID: mdl-38216643

Ku70 is a multifunctional protein with pivotal roles in DNA repair via non-homologous end-joining, V(D)J recombination, telomere maintenance, and neuronal apoptosis control. Nonetheless, its regulatory mechanisms remain elusive. Chicken Ku70 (GdKu70) cDNA has been previously cloned, and DT40 cells expressing it have significantly contributed to critical biological discoveries. GdKu70 features an additional 18 amino acids at its N-terminus compared to mammalian Ku70, the biological significance of which remains uncertain. Here, we show that the 5' flanking sequence of GdKu70 cDNA is not nearly encoded in the chicken genome. Notably, these 18 amino acids result from fusion events involving the NFE2L1 gene on chromosome 27 and the Ku70 gene on chromosome 1. Through experiments using newly cloned chicken Ku70 cDNA and specific antibodies, we demonstrated that Ku70 localizes within the cell nucleus as a heterodimer with Ku80 and promptly accumulates at DNA damage sites following injury. This suggests that the functions and spatiotemporal regulatory mechanisms of Ku70 in chickens closely resemble those in mammals. The insights and resources acquired will contribute to elucidate the various mechanisms by which Ku functions. Meanwhile, caution is advised when interpreting the previous numerous key studies that relied on GdKu70 cDNA and its expressing cells.


Antigens, Nuclear , Chickens , DNA Damage , Ku Autoantigen , Animals , Amino Acids/genetics , Antigens, Nuclear/metabolism , Chickens/genetics , Chickens/metabolism , Cloning, Molecular , DNA Damage/genetics , DNA Repair , DNA, Complementary , DNA-Binding Proteins/metabolism , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Mammals/metabolism
4.
Nucleic Acids Res ; 52(5): 2273-2289, 2024 Mar 21.
Article En | MEDLINE | ID: mdl-38118002

Albeit N1-Methyladenosine (m1A) RNA modification represents an important regulator of RNA metabolism, the role of m1A modification in carcinogenesis remains enigmatic. Herein, we found that histone lactylation enhances ALKBH3 expression and simultaneously attenuates the formation of tumor-suppressive promyelocytic leukemia protein (PML) condensates by removing the m1A methylation of SP100A, promoting the malignant transformation of cancers. First, ALKBH3 is specifically upregulated in high-risk ocular melanoma due to excessive histone lactylation levels, referring to m1A hypomethylation status. Moreover, the multiomics analysis subsequently identified that SP100A, a core component for PML bodies, serves as a downstream candidate target for ALKBH3. Therapeutically, the silencing of ALKBH3 exhibits efficient therapeutic efficacy in melanoma both in vitro and in vivo, which could be reversed by the depletion of SP100A. Mechanistically, we found that YTHDF1 is responsible for recognition of the m1A methylated SP100A transcript, which increases its RNA stability and translational efficacy. Conclusively, we initially demonstrated that m1A modification is necessary for tumor suppressor gene expression, expanding the current understandings of dynamic m1A function during tumor progression. In addition, our results indicate that lactylation-driven ALKBH3 is essential for the formation of PML nuclear condensates, which bridges our knowledge of m1A modification, metabolic reprogramming, and phase-separation events.


AlkB Homolog 3, Alpha-Ketoglutarate-Dependent Dioxygenase , Antigens, Nuclear , Autoantigens , Eye Neoplasms , Histones , Melanoma , Humans , AlkB Homolog 3, Alpha-Ketoglutarate-Dependent Dioxygenase/metabolism , Demethylation , DNA Methylation , Histones/genetics , Histones/metabolism , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , RNA/metabolism , Transcription Factors/metabolism , Antigens, Nuclear/metabolism , Autoantigens/metabolism , Eye Neoplasms/metabolism
5.
Sci Rep ; 13(1): 12162, 2023 07 27.
Article En | MEDLINE | ID: mdl-37500706

The Ku70/80 heterodimer is a key player in non-homologous end-joining DNA repair but is involved in other cellular functions like telomere regulation and maintenance, in which Ku's role is not fully characterized. It was previously reported that knockout of Ku80 in a human cell line results in lethality, but the underlying cause of Ku essentiality in human cells has yet to be fully explored. Here, we established conditional Ku70 knockout cells using CRISPR/Cas9 editing to study the essentiality of Ku70 function. While we observed loss of cell viability upon Ku depletion, we did not detect significant changes in telomere length, nor did we record lethal levels of DNA damage upon loss of Ku. Analysis of global proteome changes following Ku70 depletion revealed dysregulations of several cellular pathways including cell cycle/mitosis, RNA related processes, and translation/ribosome biogenesis. Our study suggests that the driving cause of loss of cell viability in Ku70 knockouts is not linked to the functions of Ku in DNA repair or at telomeres. Moreover, our data shows that loss of Ku affects multiple cellular processes and pathways and suggests that Ku plays critical roles in cellular processes beyond DNA repair and telomere maintenance to maintain cell viability.


Antigens, Nuclear , DNA-Binding Proteins , Humans , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , DNA Repair/genetics , DNA End-Joining Repair , DNA Damage , Telomere/genetics , Telomere/metabolism
6.
Int J Mol Sci ; 24(8)2023 Apr 11.
Article En | MEDLINE | ID: mdl-37108203

The Ku heterodimer, composed of subunits Ku70 and Ku80, is known for its essential role in repairing double-stranded DNA breaks via non-homologous end joining (NHEJ). We previously identified Ku70 S155 as a novel phosphorylation site within the von Willebrand A-like (vWA) domain of Ku70 and documented an altered DNA damage response in cells expressing a Ku70 S155D phosphomimetic mutant. Here, we conducted proximity-dependent biotin identification (BioID2) screening using wild-type Ku70, Ku70 S155D mutant, and Ku70 with a phosphoablative substitution (S155A) to identify Ku70 S155D-specific candidate proteins that may rely on this phosphorylation event. Using the BioID2 screen with multiple filtering approaches, we compared the protein interactor candidate lists for Ku70 S155D and S155A. TRIP12 was exclusive to the Ku70 S155D list, considered a high confidence interactor based on SAINTexpress analysis, and appeared in all three biological replicates of the Ku70 S155D-BioID2 mass spectrometry results. Using proximity ligation assays (PLA), we demonstrated a significantly increased association between Ku70 S155D-HA and TRIP12 compared to wild-type Ku70-HA cells. In addition, we were able to demonstrate a robust PLA signal between endogenous Ku70 and TRIP12 in the presence of double-stranded DNA breaks. Finally, co-immunoprecipitation analyses showed an enhanced interaction between TRIP12 and Ku70 upon treatment with ionizing radiation, suggesting a direct or indirect association in response to DNA damage. Altogether, these results suggest an association between Ku70 phospho-S155 and TRIP12.


DNA-Binding Proteins , Ku Autoantigen , Ubiquitin-Protein Ligases , Humans , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Biotin/metabolism , Cell Line , DNA Damage , DNA End-Joining Repair , DNA Repair , DNA-Binding Proteins/metabolism , Ku Autoantigen/metabolism , Ubiquitin-Protein Ligases/metabolism
7.
Nat Struct Mol Biol ; 30(2): 140-147, 2023 02.
Article En | MEDLINE | ID: mdl-36604499

DNA-dependent protein kinase (DNA-PK), a multicomponent complex including the DNA-PK catalytic subunit and Ku70/80 heterodimer together with DNA, is central to human DNA damage response and repair. Using a DNA-PK-selective inhibitor (M3814), we identified from one dataset two cryo-EM structures of the human DNA-PK complex in different states, the intermediate state and the active state. Here we show that activation of the kinase is regulated through conformational changes caused by the binding ligand and the string region (residues 802-846) of the DNA-PK catalytic subunit, particularly the helix-hairpin-helix motif (residues 816-836) that interacts with DNA. These observations demonstrate the regulatory role of the ligand and explain why DNA-PK is DNA dependent. Cooperation and coordination among binding partners, disordered flexible regions and mechanically flexible HEAT repeats modulate the activation of the kinase. Together with previous findings, these results provide a better molecular understanding of DNA-PK catalysis.


DNA-Activated Protein Kinase , Protein Serine-Threonine Kinases , Humans , DNA-Activated Protein Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , DNA-Binding Proteins/metabolism , Phosphorylation , Ligands , Nuclear Proteins/metabolism , Protein Kinases/metabolism , DNA/metabolism , DNA Repair , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism
8.
Cell Death Differ ; 30(2): 500-514, 2023 02.
Article En | MEDLINE | ID: mdl-36481789

The Cancer/Testis Antigen (CTA) genes comprise a group of genes whose expression under physiological conditions is restricted to the testis but is activated in many human cancers. Depending on the particular expression pattern, the CTA genes are speculated to play a role in spermatogenesis, but evidence is limited thus far. Here, we reported patients with a hemizygous nonsense mutation in cancer-testis antigen 55 (CT55) suffering from male infertility with extreme disruption in sperm production, morphology, and locomotion. Specifically, the insufficiency of sperm individualization, excessive residue of unnecessary cytoplasm, and defects in acrosome development were evident in the spermatozoa of the patients. Furthermore, mouse models with depletion of Ct55 showed accelerated infertility with age, mimicking the defects in sperm individualization, unnecessary cytoplasm removal, and meanwhile exhibiting the disrupted cumulus-oocyte complex penetration. Mechanistically, our functional experiments uncovered CT55 as a new autophagic manipulator to regulate spermatogenesis via selectively interacting with LAMP2 and GABARAP (which are key regulators in the autophagy process) and further fine-tuning their expression. Therefore, our findings revealed CT55 as a novel CTA gene involved in spermatogenesis due to its unprecedented autophagy activity.


Antigens, Nuclear , Infertility, Male , Neoplasms , Animals , Humans , Male , Mice , Infertility, Male/genetics , Infertility, Male/metabolism , Neoplasms/metabolism , Semen/metabolism , Spermatogenesis/genetics , Spermatozoa/metabolism , Testis/metabolism , Antigens, Nuclear/metabolism
9.
J Med Virol ; 95(1): e28255, 2023 01.
Article En | MEDLINE | ID: mdl-36284455

Kaposi's sarcoma (KS) is the second most common tumor in people infected with human immunodeficiency virus worldwide, but its pathogenesis is still unclear. In this study, we discovered that the expression of GATA-binding protein 3 (GATA3) was lowly expressed in KS tissues and KSHV-infected cells, while microRNA-155 (miR-155) was highly expressed in KS serum and KSHV-infected cells. miR-155 promoted the proliferation, migration and invasion of KSHV infection by targeting GATA3. Further, The KSHV-encoded protein, the Latency associated nuclear antigen (LANA), promotes the proliferation, migration and invasion of KSHV-infected cells by regulating the miR-155/GATA3 axis. Regarding the molecular mechanism, c-Jun and c-Fos interact to form a complex. LANA upregulates the expression of c-Jun and c-Fos and enhances the formation of c-Jun/c-Fos complex. The complex binds to the -95∼-100 bp site of miR-155 promoter and transcriptionally activates miR-155. All in all, LANA enhances the c-Jun/c-Fos interaction, resulting in enhanced transcriptional regulation of miR-155 by the c-Jun/c-Fos complex, thereby downregulating GATA3 and promoting the proliferation, migration and invasion of KSHV-infected cells. The discovery of LANA/c-Jun/c-Fos/miR-155/GATA3 further refines the pathogenesis of KS, potentially opening a new avenue for developing effective drugs against KS.


Herpesvirus 8, Human , MicroRNAs , Sarcoma, Kaposi , Humans , Herpesvirus 8, Human/physiology , Cell Line , Antigens, Viral/metabolism , Antigens, Nuclear/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Proliferation , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism
10.
Tissue Cell ; 79: 101954, 2022 Dec.
Article En | MEDLINE | ID: mdl-36270072

BACKGROUND: To explore the regulatory effect of serine/arginine repetitive matrix 1 (SRRM1) on hepatocellular carcinoma (HCC) and its potential pathway. METHODS: SRRM1 level in the tissue was tested by western blot and immunohistochemistry. Cells proliferation, apoptosis, migration, and invasion were tested by EdU, flow cytometry, wound healing assay, and Transwell, respectively. The potential mechanism of SRRM1 was explored through GSEA enrichment analysis and GeneMANIA protein-protein interaction (PPI) network. Co-immunoprecipitation assay was used to detect PPI. Levels of Bcl-2, Bax, Cleaved caspase 3, E-cadherin, N-cadherin, Vimentin, p-JAK2, JAK2, p-STAT3, and STAT3 were tested by Western blot. SRRM1 was highly expressed in HCC tissues, and was related to the survival and prognosis of patients. RESULTS: SRRM1 is significantly upregulated in HCC tumour tissues and correlated with progression of HCC. Overexpression of SRRM1 accelerated the proliferation, migration and invasion of HCC cells, and inhibited cell apoptosis, but low expression of SRRM1 had the opposite effect. SRRM1 positively correlated with the expression of IL6ST (GP130) and activated the JAK/STAT signaling pathways. SRRM1 affected the level of key molecules p-JAK2, JAK2, p-STAT3, and STAT3 in the JAK/STAT pathway. CONCLUSION: SRRM1 played a significant role in the proliferation, migration, invasion and apoptosis of HCC, and promoted cancer by regulating the JAK/STAT signaling pathway.


Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Janus Kinases/metabolism , Janus Kinases/pharmacology , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Signal Transduction , STAT Transcription Factors/metabolism , STAT Transcription Factors/pharmacology , Cell Proliferation/genetics , Apoptosis/genetics , Cell Movement/genetics , MicroRNAs/metabolism , RNA-Binding Proteins , Nuclear Matrix-Associated Proteins/metabolism , Nuclear Matrix-Associated Proteins/pharmacology , Antigens, Nuclear/metabolism , Antigens, Nuclear/pharmacology
12.
Cell Death Dis ; 13(9): 754, 2022 09 01.
Article En | MEDLINE | ID: mdl-36050295

Ivermectin is a widely used antiparasitic drug and shows promising anticancer activity in various cancer types. Although multiple signaling pathways modulated by ivermectin have been identified in tumor cells, few studies have focused on the exact target of ivermectin. Herein, we report the pharmacological effects and targets of ivermectin in prostate cancer. Ivermectin caused G0/G1 cell cycle arrest, induced cell apoptosis and DNA damage, and decreased androgen receptor (AR) signaling in prostate cancer cells. Further in vivo analysis showed ivermectin could suppress 22RV1 xenograft progression. Using integrated omics profiling, including RNA-seq and thermal proteome profiling, the forkhead box protein A1 (FOXA1) and non-homologous end joining (NHEJ) repair executer Ku70/Ku80 were strongly suggested as direct targets of ivermectin in prostate cancer. The interaction of ivermectin and FOXA1 reduced the chromatin accessibility of AR signaling and the G0/G1 cell cycle regulator E2F1, leading to cell proliferation inhibition. The interaction of ivermectin and Ku70/Ku80 impaired the NHEJ repair ability. Cooperating with the downregulation of homologous recombination repair ability after AR signaling inhibition, ivermectin increased intracellular DNA double-strand breaks and finally triggered cell death. Our findings demonstrate the anticancer effect of ivermectin in prostate cancer, indicating that its use may be a new therapeutic approach for prostate cancer.


Hepatocyte Nuclear Factor 3-alpha , Ivermectin , Ku Autoantigen , Prostatic Neoplasms , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Repair , Hepatocyte Nuclear Factor 3-alpha/drug effects , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Ivermectin/pharmacology , Ivermectin/therapeutic use , Ku Autoantigen/drug effects , Ku Autoantigen/metabolism , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism
13.
Stroke ; 53(10): 3192-3201, 2022 10.
Article En | MEDLINE | ID: mdl-36111544

BACKGROUND: Species-specific differences in astrocytes and their Alzheimer disease-associated pathology may influence cellular responses to other insults. Herein, human glial chimeric mice were generated to evaluate how Alzheimer disease predisposing genetic background in human astrocytes contributes to behavioral outcome and brain pathology after cortical photothrombotic ischemia. METHODS: Neonatal (P0) immunodeficient mice of both sexes were transplanted with induced pluripotent stem cell-derived astrocyte progenitors from Alzheimer disease patients carrying PSEN1 exon 9 deletion (PSEN1 ΔE9), with isogenic controls, with cells from a healthy donor, or with mouse astrocytes or vehicle. After 14 months, a photothrombotic lesion was produced with Rose Bengal in the motor cortex. Behavior was assessed before ischemia and 1 and 4 weeks after the induction of stroke, followed by tissue perfusion for histology. RESULTS: Open field, cylinder, and grid-walking tests showed a persistent locomotor and sensorimotor impairment after ischemia and female mice had larger infarct sizes; yet, these were not affected by astrocytes with PSEN1 ΔE9 background. Staining for human nuclear antigen confirmed that human cells successfully engrafted throughout the mouse brain. However, only a small number of human cells were positive for astrocytic marker GFAP (glial fibrillary acidic protein), mostly located in the corpus callosum and retaining complex human-specific morphology with longer processes compared with host counterparts. While host astrocytes formed the glial scar, human astrocytes were scattered in small numbers close to the lesion boundary. Aß (beta-amyloid) deposits were not present in PSEN1 ΔE9 astrocyte-transplanted mice. CONCLUSIONS: Transplanted human cells survived and distributed widely in the host brain but had no impact on severity of ischemic damage after cortical photothrombosis in chimeric mice. Only a small number of transplanted human astrocytes acquired GFAP-positive glial phenotype or migrated toward the ischemic lesion forming glial scar. PSEN1 ΔE9 astrocytes did not impair behavioral recovery after experimental stroke.


Alzheimer Disease , Stroke , Animals , Antigens, Nuclear/metabolism , Astrocytes/pathology , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , Humans , Ischemia/metabolism , Male , Mice , Rose Bengal/metabolism , Stroke/pathology
14.
BMC Biol ; 20(1): 182, 2022 08 19.
Article En | MEDLINE | ID: mdl-35986286

BACKGROUND: SP140 is a bromodomain-containing protein expressed predominantly in immune cells. Genetic polymorphisms and epigenetic modifications in the SP140 locus have been linked to Crohn's disease (CD), suggesting a role in inflammation. RESULTS: We report the development of the first small molecule SP140 inhibitor (GSK761) and utilize this to elucidate SP140 function in macrophages. We show that SP140 is highly expressed in CD mucosal macrophages and in in vitro-generated inflammatory macrophages. SP140 inhibition through GSK761 reduced monocyte-to-inflammatory macrophage differentiation and lipopolysaccharide (LPS)-induced inflammatory activation, while inducing the generation of CD206+ regulatory macrophages that were shown to associate with a therapeutic response to anti-TNF in CD patients. SP140 preferentially occupies transcriptional start sites in inflammatory macrophages, with enrichment at gene loci encoding pro-inflammatory cytokines/chemokines and inflammatory pathways. GSK761 specifically reduces SP140 chromatin binding and thereby expression of SP140-regulated genes. GSK761 inhibits the expression of cytokines, including TNF, by CD14+ macrophages isolated from CD intestinal mucosa. CONCLUSIONS: This study identifies SP140 as a druggable epigenetic therapeutic target for CD.


Crohn Disease , Tumor Necrosis Factor Inhibitors , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Crohn Disease/genetics , Crohn Disease/metabolism , Cytokines/genetics , Cytokines/metabolism , Epigenesis, Genetic , Humans , Macrophages , Transcription Factors/genetics
15.
Stem Cell Res Ther ; 13(1): 290, 2022 07 07.
Article En | MEDLINE | ID: mdl-35799276

AIMS: To explore the function of phosphorylation of KAP1 (p-KAP1) at the serine-824 site (S824) in the proliferation and apoptosis of endogenous neural stem cells (NSCs) after cerebral ischemic/reperfusion (I/R). METHODS: The apoptosis and proliferation of C17.2 cells transfected with the p-KAP1-expression plasmids and the expression of proliferation cell nuclear antigen (PCNA) and p-KAP1 were detected by immunofluorescence and Western blotting after the Oxygen Glucose deprivation/reperfusion model (OGD/R). The interaction of p-KAP1 and CUL4A with PCNA was analyzed by immunoprecipitation. In the rats MCAO model, we performed the adeno-associated virus (AAV) 2/9 gene delivery of p-KAP1 mutants to verify the proliferation of endogenous NSCs and the colocalization of PCNA and CUL4A by immunofluorescence. RESULTS: The level of p-KAP1 was significantly down-regulated in the stroke model in vivo and in vitro. Simulated p-KAP1(S824) significantly increased the proliferation of C17.2 cells and the expression of PCNA after OGD/R. Simulated p-KAP1(S824) enhanced the binding of p-KAP1 and PCNA and decreased the interaction between PCNA and CUL4A in C17.2 cells subjected to OGD/R. The AAV2/9-mediated p-KAP1(S824) increased endogenous NSCs proliferation, PCNA expression, p-KAP1 binding to PCNA, and improved neurological function in the rat MCAO model. CONCLUSIONS: Our findings confirmed that simulated p-KAP1(S824) improved the survival and proliferation of endogenous NSCs. The underlying mechanism is that highly expressed p-KAP1(S824) promotes binding to PCNA, and inhibits the binding of CUL4A to PCNA. This reduced CUL4A-mediated ubiquitination degradation to increase the stability of PCNA and promote the survival and proliferation of NSCs.


Brain Ischemia , Neural Stem Cells , Reperfusion Injury , Tripartite Motif-Containing Protein 28 , Animals , Antigens, Nuclear/metabolism , Brain Ischemia/metabolism , Brain Ischemia/therapy , Ischemia/metabolism , Neural Stem Cells/metabolism , Phosphorylation , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/therapy , Transcription Factors/metabolism , Tripartite Motif-Containing Protein 28/metabolism
16.
Sci Rep ; 12(1): 8134, 2022 05 17.
Article En | MEDLINE | ID: mdl-35581289

The maintenance of cellular homeostasis in living organisms requires a balance between anabolic and catabolic reactions. Macroautophagy (autophagy herein) is determined as one of the major catabolic reactions. Autophagy is an evolutionarily conserved stress response pathway that is activated by various insults including DNA damage. All sorts of damage to DNA potentially cause loss of genetic information and trigger genomic instability. Most of these lesions are repaired by the activation of DNA damage response following DNA repair mechanisms. Here we describe, a novel protein complex containing the autophagy protein ATG5 and the non-homologous end-joining repair system proteins. We discovered for the first time that ATG5 interacted with both Ku80 (XRCC5) and Ku70 (XRCC6). This novel interaction is facilitated mainly via Ku70. Our results suggest that this interaction is dynamic and enhanced upon genotoxic stresses. Strikingly, we identified that ATG5-Ku70 interaction is necessary for DNA repair and effective recovery from genotoxic stress. Therefore, our results are demonstrating a novel, direct, dynamic, and functional interaction between ATG5 and Ku70 proteins that plays a crucial role in DNA repair under genotoxic stress conditions.


DNA End-Joining Repair , DNA Repair , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Instability , Humans , Ku Autoantigen/metabolism
17.
Cell Rep ; 39(6): 110794, 2022 05 10.
Article En | MEDLINE | ID: mdl-35545041

The mammalian KU70 is a pleiotropic protein functioning in DNA repair and cytoplasmic suppression of apoptosis. We report a regulatory mechanism by which KU70's cytoplasmic function is enabled due to a methylation at K570 of KU70 by SET-domain-containing protein 4 (SETD4). While SETD4 silencing reduces the level of methylated KU70, over-expression of SETD4 enhances methylation of KU70. Mutations of Y272 and Y284 of SETD4 abrogate methylation of KU70. Although SETD4 is predominantly a nuclear protein, the methylated KU70 is enriched in the cytoplasm. SETD4 knockdown enhances staurosporine (STS)-induced apoptosis and cell killing. Over-expression of the wild-type (WT) SETD4, but not the SETD4-Y272/Y284F mutant, suppresses STS-induced apoptosis. The KU70-K570R (mouse Ku70-K568R) mutation dampens the anti-apoptosis activity of KU70. Our study identifies KU70 as a non-histone substrate of SETD4, discovers a post-translational modification of KU70, and uncovers a role for SETD4 and KU70-K570 methylation in the suppression of apoptosis.


Apoptosis , DNA Repair , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Apoptosis/genetics , Cytoplasm/metabolism , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Mammals/metabolism , Methylation , Methyltransferases , Mice , Protein Processing, Post-Translational
18.
Biochimie ; 199: 139-149, 2022 Aug.
Article En | MEDLINE | ID: mdl-35430316

DNA-dependent protein kinase (DNA-PK) is a key player in the NHEJ repair pathway. DNA-PK and its subunits, Ku70, Ku80, and catalytic subunit (DNA-PKcs), also participate in other cellular processes; however, there are still no systemic data on the effect of depletion of Ku70, Ku80 and DNA-PKcs on cell functions in the same cell line. Here, we analyzed transcriptome changes in HEK 293T cells after depletion of each DNA-PK subunit. Depletion of various DNA-PK subunits resulted in dramatic differences in the number of differentially expressed genes: only 7 genes changed more than 2-fold in DNA-PKcs-deficient cells, 29 genes in Ku80-deficient, 219 genes in Ku70-deficient. All DNA-PKcs-dependent genes were stress-related and depended on both Ku70 and Ku80. Two-thirds of Ku80-dependent genes were also differentially expressed in the Ku70-deficient line. Most Ku70-dependent genes were altered exclusively in Ku70-depleted cells, indicating that Ku70 is involved in the regulation of more processes than Ku80. GO enrichment analysis showed the effect of Ku70 knockdown on cell adhesion and matrix organization, protein degradation, cell proliferation, and differentiation. Depletion of Ku70, but not Ku80, provided greater cell motility and disassembly of cell-cell contacts. These data clearly indicate that Ku70 is more functionally important for the cell life than DNA-PKcs and even Ku80.


Antigens, Nuclear , DNA-Activated Protein Kinase , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , DNA/metabolism , DNA Repair , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Nuclear Proteins/metabolism
19.
J Virol ; 96(8): e0027922, 2022 04 27.
Article En | MEDLINE | ID: mdl-35353002

Sp100 (speckled protein 100 kDa) is a constituent component of nuclear structure PML (promyelocytic leukemia) bodies, playing important roles in mediating intrinsic and innate immunity. The Sp100 gene encodes four isoforms with distinct roles in the transcriptional regulation of both cellular and viral genes. Since Sp100 is a primary intranuclear target of infected-cell protein 0 (ICP0), an immediate early E3 ligase encoded by herpes simplex virus 1 (HSV-1), previous investigations attempting to analyze the functions of individual Sp100 variants during HSV-1 infection mostly avoided using a wild-type virus. Therefore, the role of Sp100 under natural infection by HSV-1 remains to be clarified. Here, we reappraised the antiviral capacity of four Sp100 isoforms during infection by a nonmutated HSV-1, examined the molecular behavior of the Sp100 protein in detail, and revealed the following intriguing observations. First, Sp100 isoform A (Sp100A) inhibited wild-type HSV-1 propagation in HEp-2, Sp100-/-, and PML-/- cells. Second, endogenous Sp100 is located in both the nucleus and the cytoplasm. During HSV-1 infection, the nuclear Sp100 level decreased drastically upon the detection of ICP0 in the same subcellular compartment, but cytosolic Sp100 remained stable. Third, transfected Sp100A showed subcellular localizations similar to those of endogenous Sp100 and matched the protein size of endogenous cytosolic Sp100. Fourth, HSV-1 infection induced increased secretion of endogenous Sp100 and ectopically expressed Sp100A, which copurified with extracellular vesicles (EVs) but not infectious virions. Fifth, the Sp100A level in secreting cells positively correlated with its level in EVs, and EV-associated Sp100A restricted HSV-1 in recipient cells. IMPORTANCE Previous studies show that the PML body component Sp100 protein is immediately targeted by ICP0 of HSV-1 in the nucleus during productive infection. Therefore, extensive studies investigating the interplay of Sp100 isoforms with HSV-1 were conducted using a mutant virus lacking ICP0 or in the absence of infection. The role of Sp100 variants during natural HSV-1 infection remains blurry. Here, we report that Sp100A potently and independently inhibited wild-type HSV-1 and that during HSV-1 infection, cytosolic Sp100 remained stable and was increasingly secreted into the extracellular space, in association with EVs. Furthermore, the Sp100A level in secreting cells positively correlated with its level in EVs and the anti-HSV-1 potency of these EVs in recipient cells. In summary, this study implies an active antiviral role of Sp100A during wild-type HSV-1 infection and reveals a novel mechanism of Sp100A to restrict HSV-1 through extracellular communications.


Antigens, Nuclear , Autoantigens , Herpes Simplex , Herpesvirus 1, Human , Host Microbial Interactions , Promyelocytic Leukemia Nuclear Bodies , Antigens, Nuclear/metabolism , Antiviral Agents/metabolism , Autoantigens/metabolism , Herpes Simplex/genetics , Herpesvirus 1, Human/metabolism , Humans , Promyelocytic Leukemia Nuclear Bodies/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
20.
Hum Pathol ; 124: 67-75, 2022 06.
Article En | MEDLINE | ID: mdl-35339566

The differential diagnosis between lymphoplasmacytic lymphoma (LPL) and marginal zone B-cell lymphoma, particularly splenic type (SMZL), can be challenging on onset of bone marrow biopsy (BMB) since morphology and phenotype are not specific and clinical features can overlap or be mildly developed at diagnosis. The LPL-specific L265P mutation in the MYD88 gene is not available in all laboratories, and genetic aberrancies identified in SMZL (del7q, mutations of NOTCH2 and KLF2) are seldom searched in routine practice. The study aim is to investigate the potential role of myeloid nuclear differentiation antigen (MNDA) expression in this specific differential diagnosis. We report MNDA reactivity in 559 patients with small B-cell lymphoma including bone marrow biopsies from 90 LPL and 91 SMZL cases. MYD88 p.Leu265Pro mutation status was assessed and confirmed as positive in 24 of 90 LPL cases, which served as the test set. MNDA staining was negative in 23 of 24 LPL cases in the test set (96%). In the 157 remaining cases (66 LPL, 91 SMZL), which served as the validation set, the MYD88 p.Leu265Pro mutation was unavailable and MNDA was more frequently expressed in SMZL (p < 0.00001). In addition, immunohistochemical features more consistent with SMZL (i.e., presence of CD23+ follicular dendritic cell meshworks, polytypic plasma cells, DBA44 reactivity) were more often present in MNDA-positive cases (statistically significant for 2 such parameters). On the widest case series so far published focusing on LPL and SMZL immunohistochemical diagnosis at onset of BMB, we demonstrated that MNDA expression significantly support the diagnosis of SMZL. This observation may be of particular help in cases where the MYD88 p.Leu265Pro mutational status and/or SMZL-related genetic aberrations are unavailable.


Leukemia, Lymphocytic, Chronic, B-Cell , Lymphoma, B-Cell, Marginal Zone , Splenic Neoplasms , Waldenstrom Macroglobulinemia , Antigens, Differentiation , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Biomarkers , Biopsy , Bone Marrow/pathology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, B-Cell, Marginal Zone/diagnosis , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, B-Cell, Marginal Zone/pathology , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Splenic Neoplasms/diagnosis , Waldenstrom Macroglobulinemia/diagnosis , Waldenstrom Macroglobulinemia/genetics , Waldenstrom Macroglobulinemia/pathology
...