Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell ; 186(22): 4898-4919.e25, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37827155

ABSTRACT

Expansions of repeat DNA tracts cause >70 diseases, and ongoing expansions in brains exacerbate disease. During expansion mutations, single-stranded DNAs (ssDNAs) form slipped-DNAs. We find the ssDNA-binding complexes canonical replication protein A (RPA1, RPA2, and RPA3) and Alternative-RPA (RPA1, RPA3, and primate-specific RPA4) are upregulated in Huntington disease and spinocerebellar ataxia type 1 (SCA1) patient brains. Protein interactomes of RPA and Alt-RPA reveal unique and shared partners, including modifiers of CAG instability and disease presentation. RPA enhances in vitro melting, FAN1 excision, and repair of slipped-CAGs and protects against CAG expansions in human cells. RPA overexpression in SCA1 mouse brains ablates expansions, coincident with decreased ATXN1 aggregation, reduced brain DNA damage, improved neuron morphology, and rescued motor phenotypes. In contrast, Alt-RPA inhibits melting, FAN1 excision, and repair of slipped-CAGs and promotes CAG expansions. These findings suggest a functional interplay between the two RPAs where Alt-RPA may antagonistically offset RPA's suppression of disease-associated repeat expansions, which may extend to other DNA processes.


Subject(s)
Replication Protein A , Trinucleotide Repeat Expansion , Animals , Humans , Mice , DNA/genetics , DNA Mismatch Repair , Huntington Disease/genetics , Proteins/genetics , Spinocerebellar Ataxias/genetics , Replication Protein A/metabolism
2.
Proc Natl Acad Sci U S A ; 120(52): e2310542120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38134200

ABSTRACT

Reciprocal exchanges of DNA between homologous chromosomes during meiosis, or crossovers (COs), shuffle genetic information in gametes and progeny. In many eukaryotes, the majority of COs (class I COs) are sensitive to a phenomenon called interference, which influences the occurrence of closely spaced double COs. Class I COs depend on a group of factors called ZMM (Zip, Msh, Mer) proteins including HEI10 (Human Enhancer of Invasion-10). However, how these proteins are recruited to class I CO sites is unclear. Here, we show that HEI10 forms foci on chromatin via a liquid-liquid phase separation (LLPS) mechanism that relies on residue Ser70. A HEI10S70F allele results in LLPS failure and a defect in class I CO formation. We further used immunoprecipitation-mass spectrometry to identify RPA1a (Replication Protein A 1) as a HEI10 interacting protein. Surprisingly, we find that RPA1a also undergoes phase separation and its ubiquitination and degradation are directly regulated by HEI10. We also show that HEI10 is required for the condensation of other class I CO factors. Thus, our results provide mechanistic insight into how meiotic class I CO formation is controlled by HEI10 coupling LLPS and ubiquitination.


Subject(s)
Arabidopsis Proteins , Crossing Over, Genetic , Meiosis , Chromosomes , Meiosis/genetics , Phase Separation , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism
3.
Ecotoxicol Environ Saf ; 272: 116032, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38306819

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs) have become contaminants widely distributed in the environment due to improper disposal and discharge. Previous study has found several components might involve in impairing enteric nervous system (ENS) development of zebrafish, including NSAIDs cinchophen. Deficient ENS development in fetal could lead to Hirschsprung disease (HSCR), a congenital neurocristopathy characterized by absence of enteric neurons in hindgut. However, the intrinsic mechanism of neurotoxicity of cinchophen is unclear. We confirmed that cinchophen could impair ENS development of zebrafish and transcriptome sequencing revealed that disfunction of Replication protein A1 (RPA1), which is involved in DNA replication and repairment, might be relevant to the neurotoxicity effects induced by cinchophen. Based on previous data of single cell RNA sequencing (scRNA-seq) of zebrafish gut cells, we observed that rpa1 mainly expressed in proliferating, differentiating ENS cells and neural crest progenitors. Interestingly, cinchophen induced apoptosis and impaired proliferation. Furthermore, cinchophen caused DNA damage and abnormal activation of ataxia telangiectasia mutated/ Rad3 related (ATM/ATR) and checkpoint kinase 2 (CHK2). Finally, molecular docking indicated cinchophen could bind and antagonize RPA1 more effectively. Our study might provide a better understanding and draw more attention to the role of environmental factors in the pathogenesis of HSCR. And the mechanism of cinchophen neurotoxicity would give theoretical guidance for clinical pharmacy.


Subject(s)
DNA Damage , Quinolines , Zebrafish , Animals , Zebrafish/genetics , Molecular Docking Simulation , Apoptosis , Anti-Inflammatory Agents, Non-Steroidal
4.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125953

ABSTRACT

Targeting CDC20 can enhance the radiosensitivity of tumor cells, but the function and mechanism of CDC20 on DNA damage repair response remains vague. To examine that issue, tumor cell lines, including KYSE200, KYSE450, and HCT116, were utilized to detect the expression, function, and underlying mechanism of CDC20 in radio-chemoresistance. Western blot and immunofluorescence staining were employed to confirm CDC20 expression and location, and radiation could upregulate the expression of CDC20 in the cell nucleus. The homologous recombination (HR) and non-homologous end joining (NHEJ) reporter gene systems were utilized to explore the impact of CDC20 on DNA damage repair, indicating that CDC20 could promote HR repair and radio/chemo-resistance. In the early stages of DNA damage, CDC20 stabilizes the RPA1 protein through protein-protein interactions, activating the ATR-mediated signaling cascade, thereby aiding in genomic repair. In the later stages, CDC20 assists in the subsequent steps of damage repair by the ubiquitin-mediated degradation of RPA1. CCK-8 and colony formation assay were used to detect the function of CDC20 in cell vitality and proliferation, and targeting CDC20 can exacerbate the increase in DNA damage levels caused by cisplatin or etoposide. A tumor xenograft model was conducted in BALB/c-nu/nu mice to confirm the function of CDC20 in vivo, confirming the in vitro results. In conclusion, this study provides further validation of the potential clinical significance of CDC20 as a strategy to overcome radio-chemoresistance via uncovering a novel role of CDC20 in regulating RPA1 during DNA damage repair.


Subject(s)
Cdc20 Proteins , DNA Damage , Drug Resistance, Neoplasm , Radiation Tolerance , Replication Protein A , Humans , Animals , Replication Protein A/metabolism , Replication Protein A/genetics , Mice , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Drug Resistance, Neoplasm/genetics , Cdc20 Proteins/metabolism , Cdc20 Proteins/genetics , Cell Line, Tumor , Mice, Inbred BALB C , Mice, Nude , DNA Repair/drug effects , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Cisplatin/pharmacology , HCT116 Cells , Gene Expression Regulation, Neoplastic/drug effects
5.
Plant J ; 102(4): 688-702, 2020 05.
Article in English | MEDLINE | ID: mdl-31849122

ABSTRACT

The type three effector AvrRpm1Pma from Pseudomonas syringae pv. maculicola (Pma) triggers an RPM1-mediated immune response linked to phosphorylation of RIN4 (RPM1-interacting protein 4) in Arabidopsis. However, the effector-resistance (R) gene interaction is not well established with different AvrRpm1 effectors from other pathovars. We investigated the AvrRpm1-triggered immune responses in Nicotiana species and isolated Rpa1 (Resistance to Pseudomonas syringae pv. actinidiae 1) via a reverse genetic screen in Nicotiana tabacum. Transient expression and gene silencing were performed in combination with co-immunoprecipitation and growth assays to investigate the specificity of interactions that lead to inhibition of pathogen growth. Two closely related AvrRpm1 effectors derived from Pseudomonas syringae pv. actinidiae biovar 3 (AvrRpm1Psa ) and Pseudomonas syringae pv. syringae strain B728a (AvrRpm1Psy ) trigger immune responses mediated by RPA1, a nucleotide-binding leucine-rich repeat protein with an N-terminal coiled-coil domain. In a display of contrasting specificities, RPA1 does not respond to AvrRpm1Pma , and correspondingly AvrRpm1Psa and AvrRpm1Psy do not trigger the RPM1-mediated response, demonstrating that separate R genes mediate specific immune responses to different AvrRpm1 effectors. AvrRpm1Psa co-immunoprecipitates with RPA1, and both proteins co-immunoprecipitate with RIN4. In contrast with RPM1, however, RPA1 was not activated by the phosphomimic RIN4T166D and silencing of RIN4 did not affect the RPA1 activity. Delivery of AvrRpm1Psa by Pseudomonas syringae pv. tomato (Pto) in combination with transient expression of Rpa1 resulted in inhibition of the pathogen growth in N. benthamiana. Psa growth was also inhibited by RPA1 in N. tabacum.


Subject(s)
Arabidopsis/genetics , Bacterial Proteins/immunology , Nicotiana/genetics , Plant Diseases/immunology , Plant Immunity , Proteins/metabolism , Pseudomonas syringae/immunology , Arabidopsis/immunology , Arabidopsis/microbiology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Leucine-Rich Repeat Proteins , Phosphorylation , Plant Diseases/microbiology , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Interaction Maps , Proteins/genetics , Nicotiana/immunology , Nicotiana/microbiology
6.
Proc Natl Acad Sci U S A ; 115(44): E10352-E10361, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30309964

ABSTRACT

NRF2 regulates cellular redox homeostasis, metabolic balance, and proteostasis by forming a dimer with small musculoaponeurotic fibrosarcoma proteins (sMAFs) and binding to antioxidant response elements (AREs) to activate target gene transcription. In contrast, NRF2-ARE-dependent transcriptional repression is unreported. Here, we describe NRF2-mediated gene repression via a specific seven-nucleotide sequence flanking the ARE, which we term the NRF2-replication protein A1 (RPA1) element (NRE). Mechanistically, RPA1 competes with sMAF for NRF2 binding, followed by interaction of NRF2-RPA1 with the ARE-NRE and eduction of promoter activity. Genome-wide in silico and RNA-seq analyses revealed this NRF2-RPA1-ARE-NRE complex mediates negative regulation of many genes with diverse functions, indicating that this mechanism is a fundamental cellular process. Notably, repression of MYLK, which encodes the nonmuscle myosin light chain kinase, by the NRF2-RPA1-ARE-NRE complex disrupts vascular integrity in preclinical inflammatory lung injury models, illustrating the translational significance of NRF2-mediated transcriptional repression. Our findings reveal a gene-suppressive function of NRF2 and a subset of negatively regulated NRF2 target genes, underscoring the broad impact of NRF2 in physiological and pathological settings.


Subject(s)
NF-E2-Related Factor 2/genetics , Replication Protein A/genetics , Repressor Proteins/genetics , Transcription, Genetic/genetics , Transcriptional Activation/genetics , A549 Cells , Animals , Cell Line , Cell Line, Tumor , DNA-Binding Proteins/genetics , Genome/genetics , Humans , Mice , Promoter Regions, Genetic/genetics , Response Elements/genetics
7.
Biochem Biophys Res Commun ; 498(3): 424-430, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29477843

ABSTRACT

As the sixth most prevalent cancer, hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide. Human replication protein A (RPA), a three-subunit protein, plays a central role in eukaryotic DNA replication, homologous recombination, and excision repair, including RPA1, RPA2 and RPA3. Recently, some studies focusing on the relation between RPA1 and carcinogenesis have demonstrated that RPA1 is a candidate oncogene and influences tumor biological behaviors in many cancers such as esophageal carcinoma, colon cancer, urothelial carcinomas, etc. However, the characteristic role of RPA1 in HCC and the detailed potential mechanism remain unknown. To identify the real effects of RPA1 on HCC and its potential pathway participating in the changes of liver cancer cells, we have conducted this study and demonstrated that RPA1 is up-regulated both in liver cancer cell lines and HCC tissues, which is associated with poorer prognosis, advanced TNM stage and larger tumor size. Stable knock-down of RPA1 by specific small hairpin RNA (shRNA) contributes to the impaired proliferate ability of SK-HEP-1 cells both in vitro and vivo. Consistently, upregulation of RPA1 in HuH-7 cells by specific adenovirus promotes tumor cells' proliferation. Furthermore, cyclin-dependent-kinase 4(CDK4)/Cyclin-D pathway is found to be well associated with RPA1 induced proliferation. In conclusion, RPA1 plays a pivotal role as a potential oncogene in HCC and promotes tumor proliferation via CDK4/Cyclin-D pathway.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Liver Neoplasms/metabolism , Replication Protein A/metabolism , Signal Transduction , Animals , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Replication Protein A/genetics
8.
New Phytol ; 220(2): 476-487, 2018 10.
Article in English | MEDLINE | ID: mdl-29974976

ABSTRACT

DNA replication is a fundamental process for the faithful transmission of genetic information in all living organisms. Many endogenous and environmental signals impede fork progression during DNA synthesis, which induces replication errors and DNA replication stress. Chromatin remodeling factors regulate nucleosome occupancy and the histone composition of the nucleosome in chromatin; however, whether chromatin remodeling factors are involved in the DNA replication stress response in plants is unknown. We reveal that chromatin remodeling factor CHR18 plays important roles in DNA replication stress in Arabidopsis thaliana by interacting with the DNA replication protein RPA1A. According to the genetic analysis, the loss of function of either CHR18 or RPA1A confers a high sensitivity to DNA replication stress in Arabidopsis. CHR18 interacts with RPA1A in both yeast cells and tobacco epidermal cells. The coexpression of RPA1A and CHR18 enhances the accumulation of CHR18 in nuclear foci in plants. CHR18 is a typical nuclear-localized chromatin remodeling factor with ATPase activity. Our results demonstrate that during DNA synthesis in plants, RPA1A interacts with CHR18 and recruits CHR18 to nuclear foci to resolve DNA replication stress, which is important for cell propagation and root growth in Arabidopsis plants.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Stress, Physiological , Adenosine Triphosphatases/metabolism , Arabidopsis Proteins/chemistry , Cell Nucleus/metabolism , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , Mutation/genetics , Plant Leaves/metabolism , Protein Binding , Protein Interaction Mapping
9.
Biochem J ; 473(14): 2131-9, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27208176

ABSTRACT

Cell proliferation was inhibited following forced over-expression of miR-30a in the ovary cancer cell line A2780DX5 and the gastric cancer cell line SGC7901R. Interestingly, miR-30a targets the DNA replication protein RPA1, hinders the replication of DNA and induces DNA fragmentation. Furthermore, ataxia telangiectasia mutated (ATM) and checkpoint kinase 2 (CHK2) were phosphorylated after DNA damage, which induced p53 expression, thus triggering the S-phase checkpoint, arresting cell cycle progression and ultimately initiating cancer cell apoptosis. Therefore, forced miR-30a over-expression in cancer cells can be a potential way to inhibit tumour development.


Subject(s)
Cell Proliferation/physiology , DNA Replication/physiology , MicroRNAs/physiology , Replication Protein A/metabolism , Apoptosis/genetics , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cellular Senescence/genetics , Cellular Senescence/physiology , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Comet Assay , DNA Replication/genetics , Histones/metabolism , Humans , Immunohistochemistry , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Interference/physiology , Replication Protein A/genetics
10.
Toxicol Appl Pharmacol ; 273(2): 229-41, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23732082

ABSTRACT

Safety pharmacology (SP) is an essential part of the drug development process that aims to identify and predict adverse effects prior to clinical trials. SP studies are described in the International Conference on Harmonisation (ICH) S7A and S7B guidelines. The core battery and supplemental SP studies evaluate effects of a new chemical entity (NCE) at both anticipated therapeutic and supra-therapeutic exposures on major organ systems, including cardiovascular, central nervous, respiratory, renal and gastrointestinal. This review outlines the current practices and emerging concepts in SP studies including frontloading, parallel assessment of core battery studies, use of non-standard species, biomarkers, and combining toxicology and SP assessments. Integration of the newer approaches to routine SP studies may significantly enhance the scope of SP by refining and providing mechanistic insight to potential adverse effects associated with test compounds.


Subject(s)
Drug Discovery/standards , Drug-Related Side Effects and Adverse Reactions/metabolism , Pharmaceutical Preparations/standards , Animals , Drug Discovery/methods , Drug Discovery/trends , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Drug Evaluation, Preclinical/trends , Drug Interactions/physiology , Drug-Related Side Effects and Adverse Reactions/prevention & control , Humans , Pharmaceutical Preparations/metabolism
11.
Front Oncol ; 13: 1229507, 2023.
Article in English | MEDLINE | ID: mdl-37869077

ABSTRACT

Replication Protein A (RPA) is single-strand DNA binding protein that plays a key role in the replication and repair of DNA. RPA is a heterotrimer made of 3 subunits - RPA1, RPA2, and RPA3. Germline pathogenic variants affecting RPA1 were recently described in patients with Telomere Biology Disorders (TBD), also known as dyskeratosis congenita or short telomere syndrome. Premature telomere shortening is a hallmark of TBD and results in bone marrow failure and predisposition to hematologic malignancies. Building on the finding that somatic mutations in RPA subunit genes occur in ~1% of cancers, we hypothesized that germline RPA alterations might be enriched in human cancers. Because germline RPA1 mutations are linked to early onset TBD with predisposition to myelodysplastic syndromes, we interrogated pediatric cancer cohorts to define the prevalence and spectrum of rare/novel and putative damaging germline RPA1, RPA2, and RPA3 variants. In this study of 5,993 children with cancer, 75 (1.25%) harbored heterozygous rare (non-cancer population allele frequency (AF) < 0.1%) variants in the RPA heterotrimer genes, of which 51 cases (0.85%) had ultra-rare (AF < 0.005%) or novel variants. Compared with Genome Aggregation Database (gnomAD) non-cancer controls, there was significant enrichment of ultra-rare and novel RPA1, but not RPA2 or RPA3, germline variants in our cohort (adjusted p-value < 0.05). Taken together, these findings suggest that germline putative damaging variants affecting RPA1 are found in excess in children with cancer, warranting further investigation into the functional role of these variants in oncogenesis.

12.
Biology (Basel) ; 12(4)2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37106808

ABSTRACT

Patients with metastatic colorectal cancer (mCRC) carrying BRAFV600E mutation have worse response to chemotherapy and poor prognosis. The BRAFV600E inhibitor vemurafenib has shown modest efficacy as monotherapy in BRAF-mutated mCRC due to the development of resistance. The aim of this study was to conduct a comparative proteomics profiling of the secretome from vemurafenib-sensitive vs. -resistant colon cancer cells harboring BRAFV600E mutation in order to identify specific secretory features potentially associated with changes in the resistant cells' phenotype. Towards this aim, we employed two complementary proteomics approaches including two-dimensional gel electrophoresis coupled with MALDI-TOF/TOF mass spectrometry and label-free quantitative LC-MS/MS analysis. Obtained results pointed to aberrant regulation of DNA replication and endoplasmic reticulum stress as the major secretome features associated with chemoresistant phenotype. Accordingly, two proteins implicated in these processes including RPA1 and HSPA5/GRP78 were discussed in more details in the context of biological networks and their importance as potential secretome targets for further functional and clinical evaluation. Expression patterns of RPA1 and HSPA5/GRP78 in tumor tissues from colon cancer patients were also found in additional in silico analyses to be associated with BRAFV600E mutation status, which opens the possibility to extrapolate our findings and their clinical implication to other solid tumors harboring BRAFV600E mutation, such as melanoma.

13.
Adv Sci (Weinh) ; 10(11): e2206344, 2023 04.
Article in English | MEDLINE | ID: mdl-36721037

ABSTRACT

The peripheral T cell pool is maintained at dynamic homeostasis through fine-tuning of thymic output and self-renewal of naïve T cells. Lymphopenia or reduced lymphocyte number is implicated in autoimmune diseases, yet little is known about the homeostatic mechanisms. Here, it is reported that the replication protein A1 (RPA1) plays a critical role in T cell homeostasis. Utilizing T cell-specific Rpa1-deficient (Rpa1fl/fl Cd4-cre) mice, loss of Rpa1 results in lymphopenia through restraining peripheral T cell population and limiting TCR repertoire diversity. Moreover, Rpa1fl/fl Cd4-cre mice exhibit increased susceptibility to inflammatory diseases, including colitis and hepatitis. Clinical analysis reveals that the expression of RPA1 is reduced in patients with ulcerative colitis or other autoinflammatory diseases. Mechanistically, depletion of RPA1 activates ZBP1-RIPK3 signaling through triggering the genomic DNA leakage into cytosol, consequently resulting in T cell necroptosis. This necroptotic T cell death induced by RPA1 deficiency allows the release of damage-associated molecular patterns (DAMPs), which in turn recruits leukocytes and exacerbates inflammatory response. Reciprocally, chemical or genetic inhibition of necroptosis signaling can ameliorate the Rpa1 deficiency-induced inflammatory damage. The studies thus uncover the importance of RPA1-ZBP1-RIPK3 axis in T cell homeostasis and provide a promising strategy for autoinflammatory disease treatment.


Subject(s)
Colitis , Necroptosis , Replication Protein A , Animals , Mice , Colitis/metabolism , Homeostasis , Lymphopenia , RNA-Binding Proteins/metabolism , T-Lymphocytes/metabolism , Replication Protein A/metabolism
14.
Cell Rep ; 40(2): 111071, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35830798

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease, with a prevalence of 25% worldwide. However, the underlying molecular mechanism involved in the development and progression of the NAFLD spectrum remains unclear. Single-stranded DNA-binding protein replication protein A1 (RPA1) participates in DNA replication, recombination, and damage repair. Here, we show that Rpa1+/- mice develop fatty liver disease during aging and in response to a high-fat diet. Liver-specific deletion of Rpa1 results in downregulation of genes related to fatty acid oxidation and impaired fatty acid oxidation, which leads to hepatic steatosis and hepatocellular carcinoma. Mechanistically, RPA1 binds gene regulatory regions, chromatin-remodeling factors, and HNF4A and remodels chromatin architecture, through which RPA1 promotes HNF4A transcriptional activity and fatty acid ß oxidation. Collectively, our data demonstrate that RPA1 is an important regulator of NAFLD through controlling chromatin accessibility.


Subject(s)
Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Chromatin/metabolism , Diet, High-Fat , Fatty Acids/metabolism , Homeostasis , Lipid Metabolism , Lipids , Liver/metabolism , Liver Neoplasms/pathology , Mice , Non-alcoholic Fatty Liver Disease/pathology
15.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 37(5): 490-494, 2021 Sep.
Article in Zh | MEDLINE | ID: mdl-34816659

ABSTRACT

Objective: To investigate the effects of RPA1 silencing on the invasion, migration and cell cycle of human nasopharyngeal carcinoma CNE-2R cells. Methods: shRNA technology was used to construct CNE-2R cell lines with RPA1 low-expression, which were verified by RT-PCR and Western blotting. The following assays were performed using the three 3 groups: control group(CNE-2),negative control group(NC-shRNA) and RPA1 down-regulation group(RPA1-shRNA). The effects of RPA silence on the proliferation, invasion, migration, and cell cycle of CNE-2R cells were detected using Cell Counting Kit-8, clone formation experiment, Transwell, scratch test and flow cytometry, respectively. The expressions of Chk2, p-Chk2, Cdc 25c and p-cdc25c were tested by Western blot assay. Results: The expressions of RPA1 mRNA and protein in the RPA1-shRNA group were lower than those in the CNE-2 and NC-shRNA groups significantly (P<0.01 and 0.05). Compared with CNE-2 and NC-shRNA groups, the abilities of proliferation, invasion and migration of RPA1-shRNA group were decreased and the cell cycle in the RPA1-shRNA group was blocked in the G2/M phase (P<0.01). The expressions of Chk2 and Cdc25c in RPA1-shRNA group cells were lower than those in CNE-2R and NC-shRNA group cells (P<0.05), while the expressions of p-Chk2 and p-cdc25c were higher than those in the other groups (P<0.05). Conclusion: After RPA1 silenced, the proliferation and migration of radio resistant human nasopharyngeal carcinoma CNE-2R cells was inhibited, resulting in cell cycle arrested in the G2/M phase.


Subject(s)
Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Replication Protein A/genetics , Apoptosis , Cell Cycle , Cell Division , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Gene Silencing , Humans , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics
16.
Front Genet ; 11: 855, 2020.
Article in English | MEDLINE | ID: mdl-32849834

ABSTRACT

Molecular responses to genotoxic stress, such as ionizing radiation, are intricately complex and involve hundreds of genes. Whether targeted overexpression of an endogenous gene can enhance resistance to ionizing radiation remains to be explored. In the present study we take an advantage of the CRISPR/dCas9 technology to moderately overexpress the RPA1 gene that encodes a key functional subunit of the replication protein A (RPA). RPA is a highly conserved heterotrimeric single-stranded DNA-binding protein complex involved in DNA replication, recombination, and repair. Dysfunction of RPA1 is detrimental for cells and organisms and can lead to diminished resistance to many stress factors. We demonstrate that HEK293T cells overexpressing RPA1 exhibit enhanced resistance to cell killing by gamma-radiation. Using the alkali comet assay, we show a remarkable acceleration of DNA breaks rejoining after gamma-irradiation in RPA1 overexpressing cells. However, the spontaneous rate of DNA damage was also higher in the presence of RPA1 overexpression, suggesting alterations in the processing of replication errors due to elevated activity of the RPA protein. Additionally, the analysis of the distributions of cells with different levels of DNA damage showed a link between the RPA1 overexpression and the kinetics of DNA repair within differentially damaged cell subpopulations. Our results provide knew knowledge on DNA damage stress responses and indicate that the concept of enhancing radioresistance by targeted alteration of the expression of a single gene is feasible, however undesired consequences should be considered and evaluated.

17.
Adv Sci (Weinh) ; 7(10): 1903727, 2020 May.
Article in English | MEDLINE | ID: mdl-32440486

ABSTRACT

Germline polymorphisms are linked with differential survival outcomes in cancers but are not well studied in nasopharyngeal carcinoma (NPC). Here, a two-phase association study is conducted to discover germline polymorphisms that are associated with the prognosis of NPC. The discovery phase includes two consecutive hospital cohorts of patients with NPC from Southern China. Exome-wide genotypes at 246 173 single nucleotide polymorphisms (SNPs) are determined, followed by survival analysis for each SNP under Cox proportional hazard regression model. Candidate SNP is replicated in another two independent cohorts from Southern China and Singapore. Meta-analysis of all samples (n = 5553) confirms that the presence of rs1131636-T, located in the 3'-UTR of RPA1, confers an inferior overall survival (HR = 1.33, 95% CI = 1.20-1.47, P = 6.31 × 10-8). Bioinformatics and biological assays show that rs1131636 has regulatory effects on upstream RPA1. Functional studies further demonstrate that RPA1 promotes the growth, invasion, migration, and radioresistance of NPC cells. Additionally, miR-1253 is identified as a suppressor for RPA1 expression, likely through regulation of its binding affinity to rs1131636 locus. Collectively, these findings provide a promising biomarker aiding in stratifying patients with poor survival, as well as a potential drug target for NPC.

18.
Biochim Biophys Acta Gen Subj ; 1864(7): 129607, 2020 07.
Article in English | MEDLINE | ID: mdl-32222548

ABSTRACT

BACKGROUND: Telomeres are chromosome end structures important in the maintenance of genome homeostasis. They are replenished by the action of telomerase and associated proteins, such as the OB (oligonucleotide/oligosaccharide-binding)-fold containing telomere-end binding proteins (TEBP) which plays an essential role in telomere maintenance and protection. The nature of TEBPs is well known in higher and some primitive eukaryotes, but it remains undetermined in trypanosomatids. Previous in silico searches have shown that there are no homologs of the classical TEPBs in trypanosomatids, including Leishmania sp. However, Replication Protein A subunit 1 (RPA-1), an OB-fold containing DNA-binding protein, was found co-localized with trypanosomatids telomeres and showed a high preference for the telomeric G-rich strand. METHODS AND RESULTS: We predicted the absence of structural homologs of OB-fold containing TEBPs in the Leishmania sp. genome using structural comparisons. We demonstrated by molecular docking that the ssDNA binding mode of LaRPA-1 shares features with the higher eukaryotes POT1 and RPA-1 crystal structures ssDNA binding mode. Using fluorescence spectroscopy, protein-DNA interaction assays, and FRET, we respectively show that LaRPA-1 shares some telomeric functions with the classical TEBPs since it can bind at least one telomeric repeat, protect the telomeric G-rich DNA from 3'-5' Exonuclease I digestion, and unfold telomeric G-quadruplex. CONCLUSIONS: Our results suggest that RPA-1 emerges as a TEBP in trypanosomatids, and in this context, we present two possible evolutionary landscapes of trypanosomatids RPA-1 that could reflect upon the evolution of OB-fold containing TEBPs from all eukaryotes.


Subject(s)
Leishmania , Telomere-Binding Proteins , DNA , Leishmania/genetics , Molecular Docking Simulation , Replication Protein A/chemistry , Replication Protein A/genetics , Replication Protein A/metabolism , Telomere/genetics , Telomere/metabolism , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/genetics
19.
G3 (Bethesda) ; 9(6): 1933-1943, 2019 06 05.
Article in English | MEDLINE | ID: mdl-30992318

ABSTRACT

Identifying protein localization is a useful tool in analyzing protein function. Using GFP-fusion tags, researchers can study the function of endogenous proteins in living tissue. However, these tags are considerably large, making them difficult to insert, and they can potentially affect the normal function of these proteins. To improve on these drawbacks, we have adopted the split sfGFP system for studying the localization of proteins in the Caenorhabditis elegans germline. This system divides the "super folder" GFP into 2 fragments, allowing researchers to use CRISPR/Cas9 to tag proteins more easily with the smaller subunit, while constitutively expressing the larger subunit from another locus. These two parts are able to stably interact, producing a functional GFP when both fragments are in the same cellular compartment. Our data demonstrate that the split sfGFP system can be adapted for use in C. elegans to tag endogenous proteins with relative ease. Strains containing the tags are homozygous viable and fertile. These small subunit tags produce fluorescent signals that matched the localization patterns of the wild-type protein in the gonad. Thus, our study shows that this approach could be used for tissue-specific GFP expression from an endogenous locus.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Germ Cells/metabolism , Green Fluorescent Proteins/genetics , Recombinant Fusion Proteins/genetics , Animals , Biomarkers , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cloning, Molecular , Fertility , Gene Expression , Green Fluorescent Proteins/metabolism , Molecular Imaging , Recombinant Fusion Proteins/metabolism
20.
Cell Rep ; 20(9): 1997-2009, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28854354

ABSTRACT

The RPA complex can integrate multiple stress signals into diverse responses by activating distinct DNA repair pathways. However, it remains unclear how RPA1 elects to activate a specific repair pathway during different types of DNA damage. Here, we report that PCAF/GCN5-mediated K163 acetylation of RPA1 is crucial for nucleotide excision repair (NER) but is dispensable for other DNA repair pathways. Mechanistically, we demonstrate that the acetylation of RPA1 is critical for the steady accumulation of XPA at damaged DNA sites and preferentially activates the NER pathway. DNA-PK phosphorylates and activates PCAF upon UV damage and consequently promotes the acetylation of RPA1. Moreover, the acetylation of RPA1 is tightly regulated by HDAC6 and SIRT1. Together, our results demonstrate that the K163 acetylation of RPA1 plays a key role in the repair of UV-induced DNA damage and reveal how the specific RPA1 modification modulates the choice of distinct DNA repair pathways.


Subject(s)
DNA Repair , Replication Protein A/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , DNA Damage , DNA-Activated Protein Kinase/metabolism , HEK293 Cells , HeLa Cells , Histone Deacetylase 6/metabolism , Humans , Lysine/metabolism , Protein Binding/radiation effects , Protein Stability/radiation effects , Sirtuin 1/metabolism , Ultraviolet Rays , Xeroderma Pigmentosum Group A Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL