Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.784
Filter
1.
Cell Death Dis ; 15(5): 321, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719812

ABSTRACT

RAD18, an important ubiquitin E3 ligase, plays a dual role in translesion DNA synthesis (TLS) and homologous recombination (HR) repair. However, whether and how the regulatory mechanism of O-linked N-acetylglucosamine (O-GlcNAc) modification governing RAD18 and its function during these processes remains unknown. Here, we report that human RAD18, can undergo O-GlcNAcylation at Ser130/Ser164/Thr468, which is important for optimal RAD18 accumulation at DNA damage sites. Mechanistically, abrogation of RAD18 O-GlcNAcylation limits CDC7-dependent RAD18 Ser434 phosphorylation, which in turn significantly reduces damage-induced PCNA monoubiquitination, impairs Polη focus formation and enhances UV sensitivity. Moreover, the ubiquitin and RAD51C binding ability of RAD18 at DNA double-strand breaks (DSBs) is O-GlcNAcylation-dependent. O-GlcNAcylated RAD18 promotes the binding of RAD51 to damaged DNA during HR and decreases CPT hypersensitivity. Our findings demonstrate a novel role of RAD18 O-GlcNAcylation in TLS and HR regulation, establishing a new rationale to improve chemotherapeutic treatment.


Subject(s)
Acetylglucosamine , DNA-Binding Proteins , Proliferating Cell Nuclear Antigen , Rad51 Recombinase , Recombinational DNA Repair , Ubiquitin-Protein Ligases , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Acetylglucosamine/metabolism , Rad51 Recombinase/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Phosphorylation , DNA Replication , Ubiquitination , DNA Breaks, Double-Stranded , DNA-Directed DNA Polymerase/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA Damage , DNA/metabolism , HEK293 Cells , Ultraviolet Rays , Protein Binding , Glycosylation , Translesion DNA Synthesis
2.
Nat Commun ; 15(1): 4292, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769345

ABSTRACT

Deficiencies in the BRCA1 tumor suppressor gene are the main cause of hereditary breast and ovarian cancer. BRCA1 is involved in the Homologous Recombination DNA repair pathway and, together with BARD1, forms a heterodimer with ubiquitin E3 activity. The relevance of the BRCA1/BARD1 ubiquitin E3 activity for tumor suppression and DNA repair remains controversial. Here, we observe that the BRCA1/BARD1 ubiquitin E3 activity is not required for Homologous Recombination or resistance to Olaparib. Using TULIP2 methodology, which enables the direct identification of E3-specific ubiquitination substrates, we identify substrates for BRCA1/BARD1. We find that PCNA is ubiquitinated by BRCA1/BARD1 in unperturbed conditions independently of RAD18. PCNA ubiquitination by BRCA1/BARD1 avoids the formation of ssDNA gaps during DNA replication and promotes continuous DNA synthesis. These results provide additional insight about the importance of BRCA1/BARD1 E3 activity in Homologous Recombination.


Subject(s)
BRCA1 Protein , DNA Replication , Phthalazines , Piperazines , Proliferating Cell Nuclear Antigen , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , Ubiquitination , Humans , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Proliferating Cell Nuclear Antigen/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Phthalazines/pharmacology , Piperazines/pharmacology , Homologous Recombination , Female , HEK293 Cells , Cell Line, Tumor , DNA/metabolism
3.
BMC Cancer ; 24(1): 616, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773406

ABSTRACT

BACKGROUND: DNA-Damaged Binding protein 2 (DDB2) is a protein involved in the early step of Nucleotide Excision Repair. Recently, it has been reported that DDB2 is involved in epithelial-to-mesenchymal transition (EMT), key process in tumour invasiveness and metastasis formation. However, its role is not completely known. METHODS: Boyden chamber and cell adhesion assays, and ICELLigence analysis were performed to detect HEK293 adhesion and invasion. Western blotting and gelatine zymography techniques were employed to assess the EMT protein levels and MMP enzymatic activity. Immunofluorescence analysis and pull-down assays facilitated the detection of NF-kB sub-cellular localization and interaction. RESULTS: We have previously demonstrated that the loss of DDB2-PCNA binding favours genome instability, and increases cell proliferation and motility. Here, we have investigated the phenotypic and molecular EMT-like changes after UV DNA damage, in HEK293 clones stably expressing DDB2Wt protein or a mutant form unable to interact with PCNA (DDB2PCNA-), as well as in HeLa cells transiently expressing the same DDB2 constructs. Cells expressing DDB2PCNA- showed morphological modifications along with a reduced expression of E-cadherin, an increased activity of MMP-9 and an improved ability to migrate, in concomitance with a significant upregulation of EMT-associated Transcription Factors (TFs), whose expression has been reported to favour tumour invasion. We observed a higher expression of c-Myc oncogene, NF-kB, both regulating cell proliferation and metastatic process, as well as ZEB1, a TF significantly associated with tumorigenic potential and cell migratory ability. Interestingly, a novel interaction of DDB2 with NF-kB was detected and found to be increased in cells expressing the DDB2PCNA-, suggesting a direct modulation of NF-kB by DDB2. CONCLUSION: These results highlight the role of DDB2-PCNA interaction in counteracting EMT since DDB2PCNA- protein induces in HEK293 transformed cells a gain of function contributing to the acquisition of a more aggressive phenotype.


Subject(s)
Cell Movement , DNA Damage , DNA-Binding Proteins , Epithelial-Mesenchymal Transition , NF-kappa B , Proliferating Cell Nuclear Antigen , Ultraviolet Rays , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , NF-kappa B/metabolism , Ultraviolet Rays/adverse effects , HEK293 Cells , Proliferating Cell Nuclear Antigen/metabolism , HeLa Cells , Signal Transduction , Cell Adhesion , Cell Proliferation , Protein Binding , Mutation
4.
Artif Cells Nanomed Biotechnol ; 52(1): 300-308, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38753524

ABSTRACT

Lung cancer is a dangerous disease that is lacking in an ideal therapy. Here, we evaluated the anti-lung cancer effect in nude mice of a fully human single-chain antibody (scFv) against the associated antigen 7 transmembrane receptor (Ts7TMR), which is also called G protein-coupled receptor, between A549 cells and Trichinella spiralis (T. spiralis). Our data showed that anti-Ts7TMR scFv could inhibit lung cancer growth in a dose-dependent manner, with a tumour inhibition rate of 59.1%. HE staining did not reveal any obvious tissue damage. Mechanistically, immunohistochemical staining revealed that the scFv down-regulated the expression of PCNA and VEGF in tumour tissues. Overall, this study found that anti-Ts7TMR scFv could inhibit A549 lung cancer growth by suppressing cell proliferation and angiogenesis, which may provide a new strategy for treating lung cancer.


Subject(s)
Cell Proliferation , Lung Neoplasms , Mice, Nude , Single-Chain Antibodies , Trichinella spiralis , Animals , Humans , Trichinella spiralis/immunology , Mice , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/drug therapy , A549 Cells , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Proliferating Cell Nuclear Antigen/immunology , Proliferating Cell Nuclear Antigen/metabolism , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism , Neovascularization, Pathologic/immunology
5.
FASEB J ; 38(9): e23630, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38713100

ABSTRACT

Heat shock proteins (HSPs) are a group of highly conserved proteins found in a wide range of organisms. In recent years, members of the HSP family were overexpressed in various tumors and widely involved in oncogenesis, tumor development, and therapeutic resistance. In our previous study, DNAJC24, a member of the DNAJ/HSP40 family of HSPs, was found to be closely associated with the malignant phenotype of hepatocellular carcinoma. However, its relationship with other malignancies needs to be further explored. Herein, we demonstrated that DNAJC24 exhibited upregulated expression in LUAD tissue samples and predicted poor survival in LUAD patients. The upregulation of DNAJC24 expression promoted proliferation and invasion of LUAD cells in A549 and NCI-H1299 cell lines. Further studies revealed that DNAJC24 could regulate the PI3K/AKT signaling pathway by affecting AKT phosphorylation. In addition, a series of experiments such as Co-IP and mass spectrometry confirmed that DNAJC24 could directly interact with PCNA and promoted the malignant phenotypic transformation of LUAD. In conclusion, our results suggested that DNAJC24 played an important role in the progression of LUAD and may serve as a specific prognostic biomarker for LUAD patients. The DNAJC24/PCNA/AKT axis may be a potential target for future individualized and precise treatment of LUAD patients.


Subject(s)
Cell Proliferation , HSP40 Heat-Shock Proteins , Proliferating Cell Nuclear Antigen , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Phosphorylation , HSP40 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/genetics , Male , Cell Line, Tumor , Female , Mice, Nude , Gene Expression Regulation, Neoplastic , Mice , Signal Transduction , Animals , Disease Progression , Mice, Inbred BALB C , Middle Aged , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics
6.
Nat Commun ; 15(1): 3734, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702312

ABSTRACT

Mutations in DNA damage response (DDR) factors are associated with human infertility, which affects up to 15% of the population. The DDR is required during germ cell development and meiosis. One pathway implicated in human fertility is DNA translesion synthesis (TLS), which allows replication impediments to be bypassed. We find that TLS is essential for pre-meiotic germ cell development in the embryo. Loss of the central TLS component, REV1, significantly inhibits the induction of human PGC-like cells (hPGCLCs). This is recapitulated in mice, where deficiencies in TLS initiation (Rev1-/- or PcnaK164R/K164R) or extension (Rev7 -/-) result in a > 150-fold reduction in the number of primordial germ cells (PGCs) and complete sterility. In contrast, the absence of TLS does not impact the growth, function, or homeostasis of somatic tissues. Surprisingly, we find a complete failure in both activation of the germ cell transcriptional program and in DNA demethylation, a critical step in germline epigenetic reprogramming. Our findings show that for normal fertility, DNA repair is required not only for meiotic recombination but for progression through the earliest stages of germ cell development in mammals.


Subject(s)
DNA Demethylation , DNA Repair , DNA-Directed DNA Polymerase , Germ Cells , Animals , Humans , Mice , Germ Cells/metabolism , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Male , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Female , DNA Damage , Mice, Knockout , Meiosis/genetics , DNA Replication , Proliferating Cell Nuclear Antigen/metabolism , Epigenesis, Genetic , Translesion DNA Synthesis
8.
BMC Genomics ; 25(1): 358, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38605318

ABSTRACT

BACKGROUND: Hox gene family is an important transcription factor that regulates cell process, and plays a role in the process of adipocytes differentiation and fat deposition. Previous transcriptome sequencing studies have indicated that the Homeobox A9 gene (HOXA9) is a candidate gene for regulating the process of bovine lipid metabolism, but the function and specific mechanism of action remain unclear. Therefore, this study aims to explore the role of HOXA9 in the proliferation, differentiation and apoptosis of bovine preadipocytes through gain-of-function and lose-of-function. RESULT: It found HOXA9 highly expressed in bovine adipose tissue, and its expression level changed significantly during adipocytes differentiation process. It gave a hint that HOXA9 may be involved in the process of bovine lipid metabolism. The results of HOXA9 gain-of-function experiments indicated that HOXA9 appeared to act as a negative regulator not only in the differentiation but also in the proliferation of bovine preadipocytes, which is mainly reflected that overexpression of HOXA9 down-regulate the mRNA and protein expression level of PPARγ, CEBPα and FABP4 (P < 0.05). The mRNA expression level of CDK1, CDK2, PCNA, CCNA2, CCNB1, CCND1 and CCNE2, as well as the protein expression of CDK2 also significantly decreased. The decrease of lipid droplets content was the main characteristic of the phenotype (P < 0.01), which further supported the evidence that HOXA9 was a negative regulator of preadipocytes differentiation. The decrease of cell proliferation rate and EdU positive rate, as well as the limitation of transition of preadipocytes from G0/G1 phase to S phase also provided evidence for the inhibition of proliferation. Apart from this above, we noted an interesting phenomenon that overexpression of HOXA9 showed in a significant upregulation of both mRNA and protein level of apoptosis markers, accompanied by a significant increase in cell apoptosis rate. These data led us not to refute the fact that HOXA9 played an active regulatory role in apoptosis. HOXA9 loss-of-function experiments, however, yielded the opposite results. Considering that HOXA9 acts as a transcription factor, we predicted its target genes. Dual luciferase reporter assay system indicated that overexpression of HOXA9 inhibits activity of PCNA promoter. CONCLUSION: Taken together, we demonstrated for the first time that HOXA9 played a role as a negative regulatory factor in the differentiation and proliferation of preadipocytes, but played a positive regulatory role in apoptosis, and it may play a regulatory role by targeting PCNA. This study provides basic data for further exploring the regulatory network of intramuscular fat deposition in bovine.


Subject(s)
Adipocytes , Genes, Homeobox , Animals , Cattle , Adipocytes/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Cell Differentiation/genetics , Cell Proliferation , Transcription Factors/metabolism , Apoptosis/genetics , RNA, Messenger/metabolism , Adipogenesis/genetics
9.
Int J Mol Sci ; 25(7)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38612465

ABSTRACT

Ulcerative colitis (UC) is a relapsing and reoccurring inflammatory bowel disease. The treatment effect of Alhagi maurorum and stem cell extracts on UC remains unclear. The aim of the present study was to investigate the protective role of Alhagi maurorum combined with stem cell extract on the intestinal mucosal barrier in an intestinal inflammation mouse model. Sixty mice were randomly divided into a control group, model group, Alhagi group, MSC group, and MSC/Alhagi group. MSC and Alhagi extract were found to reduce the disease activity index (DAI) scores in mice with colitis, alleviate weight loss, improve intestinal inflammation in mice (p < 0.05), preserve the integrity of the ileal wall and increase the number of goblet cells and mucin in colon tissues. Little inflammatory cell infiltration was observed in the Alhagi, MSC, or MSC/Alhagi groups, and the degree of inflammation was significantly alleviated compared with that in the model group. The distribution of PCNA and TNF-alpha in the colonic tissues of the model group was more disperse than that in the normal group (p < 0.05), and the fluorescence intensity was lower. After MSC/Alhagi intervention, PCNA and TNF-alpha were distributed along the cellular membrane in the MSC/Alhagi group (p < 0.05). Compared with that in the normal control group, the intensity was slightly reduced, but it was still stronger than that in the model group. In conclusion, MSC/Alhagi can alleviate inflammatory reactions in mouse colonic tissue, possibly by strengthening the protective effect of the intestinal mucosal barrier.


Subject(s)
Colitis, Ulcerative , Fabaceae , Mesenchymal Stem Cells , Animals , Mice , Colitis, Ulcerative/drug therapy , Stem Cell Factor , Proliferating Cell Nuclear Antigen , Tumor Necrosis Factor-alpha , Inflammation , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
10.
Aging (Albany NY) ; 16(8): 7311-7330, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38687509

ABSTRACT

Cholangiocarcinoma, a prevalent hepatic malignancy, exhibits a progressively rising incidence. While Eukaryotic translation initiation factor 3 subunit B (EIF3B) has been implicated in the occurrence and development of various cancers, its specific roles in cholangiocarcinoma remain unexplored. Immunohistochemical (IHC) analysis was employed to detect EIF3B/PCNA expression in cholangiocarcinoma. Cells were manipulated using short hairpin RNA (shRNA)-mediated lentiviruses or overexpression plasmids. Statistical significance was assessed using the Student's t-test and one-way ANOVA, with P < 0.05 considered statistically significant. EIF3B exhibited robust expression in cholangiocarcinoma, demonstrating a significant correlation with the pathological grade of cholangiocarcinoma patients. Furthermore, modulation of EIF3B expression, either depletion or elevation, demonstrated the ability to inhibit or enhance cholangiocarcinoma cell survival and migration in vitro. Mechanistically, we identified Proliferating Cell Nuclear Antigen (PCNA) as a downstream gene of EIF3B, driving cholangiocarcinoma. EIF3B stabilized PCNA by inhibiting PCNA ubiquitination, a process mediated by E3 ligase SYVN1. Similar to EIF3B, PCNA levels were also abundant in cholangiocarcinoma, and knocking down PCNA impeded cholangiocarcinoma development. Intriguingly, silencing PCNA attenuated the promotion induced by EIF3B overexpression. Furthermore, the elevated P21 protein level in shEIF3B RBE cells was partially attenuated after UC2288 (P21 signaling pathway inhibitor) treatment. Our findings underscored the potential of EIF3B as a therapeutic target for cholangiocarcinoma. Unraveling its functions holds promise for the development of more specific and effective targeted therapy strategies.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Eukaryotic Initiation Factor-3 , Proliferating Cell Nuclear Antigen , Ubiquitin-Protein Ligases , Ubiquitination , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Humans , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/genetics , Cell Line, Tumor , Eukaryotic Initiation Factor-3/metabolism , Eukaryotic Initiation Factor-3/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Cell Proliferation , Male , Female , Animals
11.
Proc Natl Acad Sci U S A ; 121(18): e2319727121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38669181

ABSTRACT

The DNA sliding clamp PCNA is a multipurpose platform for DNA polymerases and many other proteins involved in DNA metabolism. The topologically closed PCNA ring needs to be cracked open and loaded onto DNA by a clamp loader, e.g., the well-studied pentameric ATPase complex RFC (RFC1-5). The CTF18-RFC complex is an alternative clamp loader found recently to bind the leading strand DNA polymerase ε and load PCNA onto leading strand DNA, but its structure and the loading mechanism have been unknown. By cryo-EM analysis of in vitro assembled human CTF18-RFC-DNA-PCNA complex, we have captured seven loading intermediates, revealing a detailed PCNA loading mechanism onto a 3'-ss/dsDNA junction by CTF18-RFC. Interestingly, the alternative loader has evolved a highly mobile CTF18 AAA+ module likely to lower the loading activity, perhaps to avoid competition with the RFC and to limit its role to leading strand clamp loading. To compensate for the lost stability due to the mobile AAA+ module, CTF18 has evolved a unique ß-hairpin motif that reaches across RFC2 to interact with RFC5, thereby stabilizing the pentameric complex. Further, we found that CTF18 also contains a separation pin to locally melt DNA from the 3'-end of the primer; this ensures its ability to load PCNA to any 3'-ss/dsDNA junction, facilitated by the binding energy of the E-plug to the major groove. Our study reveals unique structural features of the human CTF18-RFC and contributes to a broader understanding of PCNA loading by the alternative clamp loaders.


Subject(s)
ATPases Associated with Diverse Cellular Activities , Cryoelectron Microscopy , Nuclear Proteins , Proliferating Cell Nuclear Antigen , Replication Protein C , Humans , Cryoelectron Microscopy/methods , DNA/metabolism , DNA/chemistry , DNA Replication , Models, Molecular , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/chemistry , Protein Binding , Replication Protein C/metabolism , Replication Protein C/chemistry
12.
Microb Biotechnol ; 17(4): e14471, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38646975

ABSTRACT

Proliferating cell nuclear antigen (PCNA) is an essential factor for DNA metabolism. The influence of PCNA on DNA replication and repair, combined with the high expression rate of PCNA in various tumours renders PCNA a promising target for cancer therapy. In this context, an autodisplay-based screening method was developed to identify peptidic PCNA interaction inhibitors. A 12-mer randomized peptide library consisting of 2.54 × 106 colony-forming units was constructed and displayed at the surface of Escherichia coli BL21 (DE3) cells by autodisplay. Cells exhibiting an enhanced binding to fluorescent mScarlet-I-PCNA were enriched in four sorting rounds by flow cytometry. This led to the discovery of five peptide variants with affinity to mScarlet-I-PCNA. Among these, P3 (TCPLRWITHDHP) exhibited the highest binding signal. Subsequent flow cytometric analysis revealed a dissociation constant of 0.62 µM for PCNA-P3 interaction. Furthermore, the inhibition of PCNA interactions was investigated using p15, a PIP-box containing protein involved in DNA replication and repair. P3 inhibited the PCNA-p1551-70 interaction with a half maximal inhibitory activity of 16.2 µM, characterizing P3 as a potent inhibitor of the PCNA-p15 interaction.


Subject(s)
Escherichia coli , Peptide Library , Proliferating Cell Nuclear Antigen , Protein Binding , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Flow Cytometry , Drug Evaluation, Preclinical/methods , Cell Surface Display Techniques/methods , Humans , Peptides/metabolism , Peptides/genetics , Peptides/chemistry , Peptides/pharmacology
13.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 85-89, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38678624

ABSTRACT

We aimed to explore the effects of silencing NOD-like receptor protein 3 (NLRP3) on proliferation of psoriasis-like HaCaT cells and expressions of cytokines. HaCaT cells were treated with human keratinocyte growth factor (KGF) and were divided into KGF group, negative control group, NLRP3-RNAi group and control group. Cells proliferation was detected by CCK8, cell clone formation rate was detected by clone formation assay, distribution of cells cycle was detected by flow cytometry, expressions of cyclin B1 (Cyclin B1), cyclin-dependent kinase 2 (CDK2), Ki67 and proliferating cell nuclear antigen (PCNA) proteins were detected by Western blot, and levels of interleukin (IL)-17, IL-23, IL-6 and tumor necrosis factor α (TNF-α) were detected by enzyme-linked immunosorbent assay. Compared with control group, expressions of NLRP3 mRNA and protein, proliferation rate and clonal formation rate were increased in KGF group, percentage of cells in G0/G1 phase was decreased, percentage of cells in S phase was increased, expressions of Cyclin B1, CDK2, Ki67 and PCNA proteins were increased, and levels of IL-17, IL-23, IL-6 and TNF-α were increased. Compared with negative control group, expressions of NLRP3 mRNA and protein, proliferation rate and clonal formation rate were decreased in NLRP3-RNAi group, percentage of cells in G0/G1 phase was increased, percentage of cells in S phase was decreased, expressions of Cyclin B1, CDK2, Ki67 and PCNA proteins were decreased, and levels of IL-17, IL-23, IL-6 and TNF-α were decreased. Silencing NLRP3 gene can inhibit the proliferation of psoriasis-like HaCaT cells, arrest cell cycle, inhibit the expressions of cell proliferation-related proteins and reduce levels of pro-inflammatory factors.


Subject(s)
Cell Proliferation , Cytokines , NLR Family, Pyrin Domain-Containing 3 Protein , Proliferating Cell Nuclear Antigen , Psoriasis , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Cell Proliferation/genetics , Psoriasis/genetics , Psoriasis/metabolism , Psoriasis/pathology , Cytokines/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin B1/metabolism , Cyclin B1/genetics , Gene Silencing , Ki-67 Antigen/metabolism , Ki-67 Antigen/genetics , HaCaT Cells , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Cell Cycle/genetics , Interleukin-17/metabolism , Interleukin-17/genetics , RNA Interference , Interleukin-23/metabolism , Interleukin-23/genetics , Interleukin-6/metabolism , Interleukin-6/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
14.
Indian J Ophthalmol ; 72(Suppl 3): S501-S504, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38648458

ABSTRACT

PURPOSE: Pterygium is a common ocular surface disease defined by fibrovascular conjunctival growth extending onto the cornea. However, its pathogenesis remains unclear. This study aimed to determine the role of CD44, proliferating cell nuclear antigen (PCNA), and E-cadherin in pterygium formation and recurrence. METHODS: Sixty patients with pterygium participated in the study, and we collected conjunctival samples from 30 patients to form a control group. CD44, PCNA, and E-cadherin expressions in surgically excised pterygium were compared with tissue samples from the control group. RESULTS: We observed that the percentages of CD44 and PCNA were statistically higher in the primary pterygium group and recurrent pterygium group than in the control group (P < 0.001 and P < 0.001, respectively). Conversely, E-cadherin values were statistically higher in the control group than in the primary and recurrent pterygium groups (P = 0.013 and P < 0.001, respectively). CONCLUSION: Cell proliferation and cell adhesion factors may play important roles in the pathogenesis of pterygium.


Subject(s)
Cadherins , Conjunctiva , Hyaluronan Receptors , Proliferating Cell Nuclear Antigen , Pterygium , Female , Humans , Male , Biomarkers/metabolism , Cadherins/metabolism , Cadherins/biosynthesis , Conjunctiva/metabolism , Conjunctiva/pathology , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Hyaluronan Receptors/biosynthesis , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/biosynthesis , Pterygium/diagnosis , Pterygium/metabolism , Pterygium/pathology
15.
Folia Histochem Cytobiol ; 62(1): 25-36, 2024.
Article in English | MEDLINE | ID: mdl-38563050

ABSTRACT

INTRODUCTION: Endometriosis (EMs), manifested by pain and infertility, is a chronic inflammatory disease. The precise pathophysiology of this disease remains uncertain. Insulin-like growth factor-2 mRNA-binding protein 1 (IGF2BP1) and polypyrimidine tract-binding protein 1 (PTBP1) have both been found to regulate proliferation, apoptosis, and invasion. This study aimed to investigate the effects of IGF2BP1/PTBP1 in treating EMs. MATERIALS AND METHODS: qRT-PCR and western blotting were employed to quantify IGF2BP1 and PTBP1 expression in six patients with EMs (mean age 33.83 years). The correlation analysis, STRING database prediction, and RNA immunoprecipitation were utilized to identify the relationship between IGF2BP1 and PTBP1. Ectopic endometrial volume, weight, HE staining, and IGF2BP1 silencing were utilized to estimate the effects of IGF2BP1 in EMs model rats. qRT-PCR, CCK-8, 5-ethynyl-2'-deoxyuridine (EDU) labeling, Transwell assay, and flow cytometry were utilized to assess the effects of IGF2BP1/PTBP1 on the proliferation, migration, invasion, and apoptosis of ectopic endometrial stromal cells (eESCs). Furthermore, western blotting was employed to evaluate expressions of PCNA, VEGF, and E-cadherin in EMs rats and eESCs. RESULTS: The mRNA and protein levels of IGF2BP1 and PTBP1 in the ectopic and eutopic endometrium of EMs patients were significantly increased. RNA immunoprecipitation revealed a close interaction of IGF2BP1 with PTBP1. Additionally, the endometrial volume, weight, and histopathologic scores in rats were significantly reduced after IGF2BP1 silencing. IGF2BP1 silencing also decreased the expression of PCNA and VEGF, and increased E-cadherin expression in endometrial tissues of EMs rats. Moreover, IGF2BP1 silencing inhibited proliferation, migration, and invasion and promoted apoptosis through PTBP1 in eESCs. CONCLUSIONS: IGF2BP1 exhibits potential beneficial properties in the management of EMs by interacting with PTBP1, thereby highlighting IGF2BP1 as a promising therapeutic target for EMs.


Subject(s)
Endometriosis , Adult , Animals , Female , Humans , Rats , Cadherins/metabolism , Cell Proliferation , Endometriosis/pathology , Endometrium/pathology , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Polypyrimidine Tract-Binding Protein/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
Mol Biol Rep ; 51(1): 518, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622261

ABSTRACT

BACKGROUND: Cold atmospheric plasma (CAP) has been widely used in biomedical research, especially in vitro cancer therapy. Cutaneous squamous cell carcinoma (CSCC) is a malignant tumor originating from epidermal keratinocytes. However, the mechanism of CAP therapy on CSCC remains unclear. METHODS AND RESULTS: The animal models of CSCC induced by 7,12-dimethylbenz(a) anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA) were constructed. For the CAP treatment group, after each TPA application, CAP was administered for 3 min twice weekly after drying. HE staining were used to detect the pathological status of tumor tissue in each group. The levels of PCNA, Bcl-2, Bax, MMP2 and MMP9 were evaluated by western blot and qPCR. TUNEL staining were used to detect apoptosis in tumor tissues. In vivo, serum samples were used for ELISA of total ROS. MTT assay was used to detect the viability of A431 cells. Western blot and qPCR were used to detect the levels of PCNA, Bcl-2, Bax, MMP2 and MMP9 in A431 cells. A431 cell proliferation was examined by colony formation assay. The proportions of apoptosis of A431 cells were detected by flow cytometry. Transwell assessed the ability of A431 cells migration and proliferation. We found that CAP could induce skin cancer cells apoptosis and inhibit the progress of skin cancer. Through experiments in vitro, reactive oxygen species (ROS) generated by N-acetylcysteine (NAC) and CAP inhibited the proliferation and migration of A431 skin cancer cells while promoting apoptosis. CONCLUSIONS: These evidences suggest the protective effect of CAP in CSCC, and CAP has the potential clinical application of CSCC.


Subject(s)
Carcinoma, Squamous Cell , Plasma Gases , Skin Neoplasms , Animals , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Reactive Oxygen Species/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Plasma Gases/pharmacology , Proliferating Cell Nuclear Antigen/genetics , bcl-2-Associated X Protein , Apoptosis , Cell Line, Tumor , Cell Proliferation
17.
Viruses ; 16(4)2024 04 15.
Article in English | MEDLINE | ID: mdl-38675954

ABSTRACT

The first point-of-care (PoC) test (v-RetroFel®; modified version 2021) determining the presence of FeLV p27 antigen and FeLV anti-p15E antibodies has become recently commercially available to identify different feline leukaemia virus (FeLV) infection outcomes. This study aimed to assess this PoC test's performance concerning FeLV p27 antigen and FeLV anti-p15E antibody detection. Sensitivity, specificity, positive and negative predictive values (PPV, NPV) were assessed after ten minutes (recommended) and 20 min (prolonged) incubation times. The test results were evaluated as either positive or negative. Serum samples from 934 cats were included, originating from Italy (n = 269), Portugal (n = 240), Germany (n = 318), and France (n = 107). FeLV p27 antigen and anti-p15E antibodies were measured by reference standard ELISAs and compared to the PoC test results. The PoC test was easy to perform and the results easy to interpret. Sensitivity and specificity for FeLV p27 antigen were 82.8% (PPV: 57.8%) and 96.0% (NPV: 98.8%) after both, ten and 20 minues of incubation time. Sensitivity and specificity for anti-p15E antibodies were 31.4% (PPV: 71.6%) and 96.9% (NPV: 85.1%) after ten minutes incubation time; sensitivity was improved by a prolonged incubation time (20 min) to 40.0% (PPV: 76.3%), while specificity remained the same (96.9%, NPV: 86.7%). Despite the improved sensitivity using the prolonged incubation time, lower than ideal sensitivities for both p27 antigen and especially anti-p15E antibodies were found, indicating that the PoC test in its current version needs further improvement prior to application in the field.


Subject(s)
Antibodies, Viral , Antigens, Viral , Leukemia Virus, Feline , Point-of-Care Testing , Proliferating Cell Nuclear Antigen , Animals , Cats , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cat Diseases/diagnosis , Cat Diseases/immunology , Cat Diseases/virology , Enzyme-Linked Immunosorbent Assay/methods , Leukemia Virus, Feline/immunology , Leukemia, Feline/diagnosis , Leukemia, Feline/immunology , Leukemia, Feline/virology , Point-of-Care Systems , Retroviridae Proteins, Oncogenic/chemistry , Retroviridae Proteins, Oncogenic/immunology , Sensitivity and Specificity
18.
Int J Mol Sci ; 25(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38542333

ABSTRACT

DNA Damage Tolerance (DDT) mechanisms allow cells to bypass lesions in the DNA during replication. This allows the cells to progress normally through the cell cycle in the face of abnormalities in their DNA. PCNA, a homotrimeric sliding clamp complex, plays a central role in the coordination of various processes during DNA replication, including the choice of mechanism used during DNA damage bypass. Mono-or poly-ubiquitination of PCNA facilitates an error-prone or an error-free bypass mechanism, respectively. In contrast, SUMOylation recruits the Srs2 helicase, which prevents local homologous recombination. The Elg1 RFC-like complex plays an important role in unloading PCNA from the chromatin. We analyze the interaction of mutations that destabilize PCNA with mutations in the Elg1 clamp unloader and the Srs2 helicase. Our results suggest that, in addition to its role as a coordinator of bypass mechanisms, the very presence of PCNA on the chromatin prevents homologous recombination, even in the absence of the Srs2 helicase. Thus, PCNA unloading seems to be a pre-requisite for recombinational repair.


Subject(s)
Saccharomyces cerevisiae Proteins , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , Homologous Recombination , DNA Replication , DNA/genetics , DNA/metabolism , Chromatin/genetics , Chromatin/metabolism , Carrier Proteins/metabolism
19.
Sci Rep ; 14(1): 7066, 2024 03 25.
Article in English | MEDLINE | ID: mdl-38528023

ABSTRACT

Maintenance of genome integrity is instrumental in preventing cancer. In addition to DNA repair pathways that prevent damage to DNA, damage tolerance pathways allow for the survival of cells that encounter DNA damage during replication. The Rad6/18 pathway is instrumental in this process, mediating damage bypass by ubiquitination of proliferating cell nuclear antigen. Previous studies have shown different roles of Rad18 in vivo and in tumorigenesis. Here, we show that B cells induce Rad18 expression upon proliferation induction. We have therefore analysed the role of Rad18 in B cell activation as well as in B cell lymphomagenesis mediated by an Eµ-Myc transgene. We find no activation defects or survival differences between Rad18 WT mice and two different models of Rad18 deficient tumour mice. Also, tumour subtypes do not differ between the mouse models. Accordingly, functions of Rad18 in B cell activation and tumorigenesis may be compensated for by other pathways in B cells.


Subject(s)
Lymphocyte Activation , Neoplasms , Ubiquitin-Protein Ligases , Animals , Mice , Carcinogenesis/genetics , DNA Damage , DNA Repair , DNA Replication , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , DNA-Binding Proteins/metabolism , B-Lymphocytes/metabolism , Lymphocyte Activation/genetics
20.
EMBO J ; 43(7): 1273-1300, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448672

ABSTRACT

MAGEA4 is a cancer-testis antigen primarily expressed in the testes but aberrantly overexpressed in several cancers. MAGEA4 interacts with the RING ubiquitin ligase RAD18 and activates trans-lesion DNA synthesis (TLS), potentially favouring tumour evolution. Here, we employed NMR and AlphaFold2 (AF) to elucidate the interaction mode between RAD18 and MAGEA4, and reveal that the RAD6-binding domain (R6BD) of RAD18 occupies a groove in the C-terminal winged-helix subdomain of MAGEA4. We found that MAGEA4 partially displaces RAD6 from the RAD18 R6BD and inhibits degradative RAD18 autoubiquitination, which could be countered by a competing peptide of the RAD18 R6BD. AlphaFold2 and cross-linking mass spectrometry (XL-MS) also revealed an evolutionary invariant intramolecular interaction between the catalytic RING and the DNA-binding SAP domains of RAD18, which is essential for PCNA mono-ubiquitination. Using interaction proteomics, we found that another Type-I MAGE, MAGE-C2, interacts with the RING ubiquitin ligase TRIM28 in a manner similar to the MAGEA4/RAD18 complex, suggesting that the MAGEA4 peptide-binding groove also serves as a ligase-binding cleft in other type-I MAGEs. Our data provide new insights into the mechanism and regulation of RAD18-mediated PCNA mono-ubiquitination.


Subject(s)
Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Ubiquitin/metabolism , Peptides/metabolism , DNA Damage
SELECTION OF CITATIONS
SEARCH DETAIL
...