Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(2)2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38256274

ABSTRACT

Cancer stands as the leading global cause of mortality, with rare cancer comprising 230 distinct subtypes characterized by infrequent incidence. Despite the inherent challenges in addressing the diagnosis and treatment of rare cancers due to their low occurrence rates, several biomedical breakthroughs have led to significant advancement in both areas. This review provides a comprehensive overview of state-of-the-art diagnostic techniques that encompass new-generation sequencing and multi-omics, coupled with the integration of artificial intelligence and machine learning, that have revolutionized rare cancer diagnosis. In addition, this review highlights the latest innovations in rare cancer therapeutic options, comprising immunotherapy, targeted therapy, transplantation, and drug combination therapy, that have undergone clinical trials and significantly contribute to the tumor remission and overall survival of rare cancer patients. In this review, we summarize recent breakthroughs and insights in the understanding of rare cancer pathophysiology, diagnosis, and therapeutic modalities, as well as the challenges faced in the development of rare cancer diagnosis data interpretation and drug development.


Subject(s)
Artificial Intelligence , Neoplasms , Humans , Drug Therapy, Combination , Immunotherapy , Drug Development , Neoplasms/diagnosis , Neoplasms/therapy
2.
Biomed Pharmacother ; 169: 115877, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37951025

ABSTRACT

DNA repair is a vital mechanism in cells that protects against DNA damage caused by internal and external factors. It involves a network of signaling pathways that monitor and transmit damage signals, activating various cellular activities to repair DNA damage and maintain genomic integrity. Dysfunctions in this repair pathway are strongly associated with the development and progression of cancer. However, they also present an opportunity for targeted therapy in breast cancer. Extensive research has focused on developing inhibitors that play a crucial role in the signaling pathway of DNA repair, particularly due to the remarkable success of PARP1 inhibitors (PARPis) in treating breast cancer patients with BRCA1/2 mutations. In this review, we summarize the current research progress and clinical implementation of BRCA and BRCAness in targeted treatments for the DNA repair pathway. Additionally, we present advancements in diverse inhibitors of DNA repair, both as individual and combined approaches, for treating breast cancer. We also discuss the clinical application of DNA repair-targeted therapy for breast cancer, including the rationale, indications, and summarized clinical data for patients with different breast cancer subtypes. We assess their influence on cancer progression, survival rates, and major adverse reactions. Last, we anticipate forthcoming advancements in targeted therapy for cancer treatment and emphasize prospective areas of development.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly(ADP-ribose) Polymerases/metabolism , DNA Repair , DNA Damage
3.
Int J Mol Sci ; 24(12)2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37373360

ABSTRACT

DNA double-strand breaks (DSBs) are the most lethal DNA damages which lead to severe genome instability. Phosphorylation is one of the most important protein post-translation modifications involved in DSBs repair regulation. Kinases and phosphatases play coordinating roles in DSB repair by phosphorylating and dephosphorylating various proteins. Recent research has shed light on the importance of maintaining a balance between kinase and phosphatase activities in DSB repair. The interplay between kinases and phosphatases plays an important role in regulating DNA-repair processes, and alterations in their activity can lead to genomic instability and disease. Therefore, study on the function of kinases and phosphatases in DSBs repair is essential for understanding their roles in cancer development and therapeutics. In this review, we summarize the current knowledge of kinases and phosphatases in DSBs repair regulation and highlight the advancements in the development of cancer therapies targeting kinases or phosphatases in DSBs repair pathways. In conclusion, understanding the balance of kinase and phosphatase activities in DSBs repair provides opportunities for the development of novel cancer therapeutics.


Subject(s)
DNA Breaks, Double-Stranded , Neoplasms , Humans , Protein Kinases/genetics , Phosphoric Monoester Hydrolases/genetics , DNA Repair , Neoplasms/genetics , DNA
4.
Int J Mol Sci ; 24(5)2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36902170

ABSTRACT

DNA damage is a double-edged sword in cancer cells. On the one hand, DNA damage exacerbates gene mutation frequency and cancer risk. Mutations in key DNA repair genes, such as breast cancer 1 (BRCA1) and/or breast cancer 2 (BRCA2), induce genomic instability and promote tumorigenesis. On the other hand, the induction of DNA damage using chemical reagents or radiation kills cancer cells effectively. Cancer-burdening mutations in key DNA repair-related genes imply relatively high sensitivity to chemotherapy or radiotherapy because of reduced DNA repair efficiency. Therefore, designing specific inhibitors targeting key enzymes in the DNA repair pathway is an effective way to induce synthetic lethality with chemotherapy or radiotherapy in cancer therapeutics. This study reviews the general pathways involved in DNA repair in cancer cells and the potential proteins that could be targeted for cancer therapeutics.


Subject(s)
DNA Damage , DNA Repair , Neoplasms , Humans , BRCA1 Protein/genetics , DNA Repair/genetics , Mutation , Neoplasms/genetics , BRCA2 Protein/genetics
5.
Mol Cell ; 83(7): 1043-1060.e10, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36854302

ABSTRACT

Repair of DNA double-strand breaks (DSBs) elicits three-dimensional (3D) chromatin topological changes. A recent finding reveals that 53BP1 assembles into a 3D chromatin topology pattern around DSBs. How this formation of a higher-order structure is configured and regulated remains enigmatic. Here, we report that SLFN5 is a critical factor for 53BP1 topological arrangement at DSBs. Using super-resolution imaging, we find that SLFN5 binds to 53BP1 chromatin domains to assemble a higher-order microdomain architecture by driving damaged chromatin dynamics at both DSBs and deprotected telomeres. Mechanistically, we propose that 53BP1 topology is shaped by two processes: (1) chromatin mobility driven by the SLFN5-LINC-microtubule axis and (2) the assembly of 53BP1 oligomers mediated by SLFN5. In mammals, SLFN5 deficiency disrupts the DSB repair topology and impairs non-homologous end joining, telomere fusions, class switch recombination, and sensitivity to poly (ADP-ribose) polymerase inhibitor. We establish a molecular mechanism that shapes higher-order chromatin topologies to safeguard genomic stability.


Subject(s)
Chromatin , DNA Repair , Animals , Chromatin/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Mammals/metabolism , Telomere-Binding Proteins/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Cell Cycle Proteins/metabolism
7.
Nat Cancer ; 3(9): 1088-1104, 2022 09.
Article in English | MEDLINE | ID: mdl-36138131

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers. Characterization of genetic alterations will improve our understanding and therapies for this disease. Here, we report that PDAC with elevated expression of METTL16, one of the 'writers' of RNA N6-methyladenosine modification, may benefit from poly-(ADP-ribose)-polymerase inhibitor (PARPi) treatment. Mechanistically, METTL16 interacts with MRE11 through RNA and this interaction inhibits MRE11's exonuclease activity in a methyltransferase-independent manner, thereby repressing DNA end resection. Upon DNA damage, ATM phosphorylates METTL16 resulting in a conformational change and autoinhibition of its RNA binding. This dissociates the METTL16-RNA-MRE11 complex and releases inhibition of MRE11. Concordantly, PDAC cells with high METTL16 expression show increased sensitivity to PARPi, especially when combined with gemcitabine. Thus, our findings reveal a role for METTL16 in homologous recombination repair and suggest that a combination of PARPi with gemcitabine could be an effective treatment strategy for PDAC with elevated METTL16 expression.


Subject(s)
Carcinoma, Pancreatic Ductal , MRE11 Homologue Protein , Methyltransferases , Pancreatic Neoplasms , Adenosine Diphosphate Ribose , Carcinoma, Pancreatic Ductal/drug therapy , DNA , Exonucleases/genetics , Humans , MRE11 Homologue Protein/genetics , Methyltransferases/genetics , Pancreatic Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics , RNA , Synthetic Lethal Mutations , Pancreatic Neoplasms
8.
Clin Pharmacol Ther ; 111(6): 1296-1306, 2022 06.
Article in English | MEDLINE | ID: mdl-35288936

ABSTRACT

Approximately one-third of patients with metastatic castration-resistant prostate cancer (CRPC) exhibited primary abiraterone resistance. To identify alternative treatment for abiraterone nonresponders, we performed drug discovery analyses using the L1000 database using differentially expressed genes identified in tumor biopsies and patient-derived xenograft (PDX) tumors between abiraterone responders and nonresponders enrolled in PROMOTE trial. This approach identified 3 drugs, including topoisomerase II (TOP2) inhibitor mitoxantrone, CDK4/6 inhibitor palbociclib, and pan-CDK inhibitor PHA-793887. These drugs significantly suppressed the growth of abiraterone-resistant cell lines and PDX models. Moreover, we identified 11 genes targeted by all 3 drugs that were associated with worse outcomes in both the PROMOTE and Stand Up To Cancer cohorts. This 11-gene panel might also function as biomarkers to select the 3 alternative therapies for this subgroup of patients with CRPC, warranting further clinical investigation.


Subject(s)
Complementary Therapies , Prostatic Neoplasms, Castration-Resistant , Androstenes , Biomarkers , Humans , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Treatment Outcome
9.
Nat Cell Biol ; 23(8): 894-904, 2021 08.
Article in English | MEDLINE | ID: mdl-34354233

ABSTRACT

The shieldin complex functions as the downstream effector of 53BP1-RIF1 to promote DNA double-strand break end-joining by restricting end resection. The SHLD2 subunit binds to single-stranded DNA ends and blocks end resection through OB-fold domains. Besides blocking end resection, it is unclear how the shieldin complex processes SHLD2-bound single-stranded DNA and promotes non-homologous end-joining. Here, we identify a downstream effector of the shieldin complex, ASTE1, as a structure-specific DNA endonuclease that specifically cleaves single-stranded DNA and 3' overhang DNA. ASTE1 localizes to DNA damage sites in a shieldin-dependent manner. Loss of ASTE1 impairs non-homologous end-joining, leads to hyper-resection and causes defective immunoglobulin class switch recombination. ASTE1 deficiency also causes resistance to poly(ADP-ribose) polymerase inhibitors in BRCA1-deficient cells owing to restoration of homologous recombination. These findings suggest that ASTE1-mediated 3' single-stranded DNA end cleavage contributes to the control of DSB repair choice by 53BP1, RIF1 and shieldin.


Subject(s)
DNA End-Joining Repair , Deoxyribonuclease I/physiology , Proteins/physiology , Animals , Cell Cycle Proteins/physiology , DNA/metabolism , DNA-Binding Proteins/physiology , Female , Genomic Instability , HEK293 Cells , Humans , Immunoglobulin Class Switching/physiology , Male , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins
10.
Cancer Discov ; 11(11): 2726-2737, 2021 11.
Article in English | MEDLINE | ID: mdl-34099454

ABSTRACT

Immune checkpoint blockade (ICB) has revolutionized cancer therapy. However, the response of patients to ICB is difficult to predict. Here, we examined 81 patients with lung cancer under ICB treatment and found that patients with MET amplification were resistant to ICB and had a poor progression-free survival. Tumors with MET amplifications had significantly decreased STING levels and antitumor T-cell infiltration. Furthermore, we performed deep single-cell RNA sequencing on more than 20,000 single immune cells and identified an immunosuppressive signature with increased subsets of XIST- and CD96-positive exhausted natural killer (NK) cells and decreased CD8+ T-cell and NK-cell populations in patients with MET amplification. Mechanistically, we found that oncogenic MET signaling induces phosphorylation of UPF1 and downregulates tumor cell STING expression via modulation of the 3'-UTR length of STING by UPF1. Decreased efficiency of ICB by MET amplification can be overcome by inhibiting MET. SIGNIFICANCE: We suggest that the combination of MET inhibitor together with ICB will overcome ICB resistance induced by MET amplification. Our report reveals much-needed information that will benefit the treatment of patients with primary MET amplification or EGFR-tyrosine kinase inhibitor resistant-related MET amplification.This article is highlighted in the In This Issue feature, p. 2659.


Subject(s)
Lung Neoplasms , Proto-Oncogene Proteins c-met , CD8-Positive T-Lymphocytes , Gene Amplification , Humans , Immunotherapy , Killer Cells, Natural , Lung Neoplasms/therapy , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Oncogenes , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism
11.
Pharmacogenet Genomics ; 31(7): 155-164, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34001842

ABSTRACT

OBJECTIVES: We previously discovered that the single nucleotide polymorphisms (SNP) rs9940645 in the ZNF423 gene regulate ZNF423 expression and serve as a potential biomarker for response to selective estrogen receptor modulators (SERMs). Here we explored pathways involved in ZNF423-mediated SERMs response and drugs that potentially sensitize SERMs. METHODS: RNA sequencing and label-free quantitative proteomics were performed to identify genes and pathways that are regulated by ZNF423 and the ZNF423 SNP. Both cultured cells and mouse xenograft models with different ZNF423 SNP genotypes were used to study the cellular responses to metformin. RESULTS: We identified ribosome and AMP-activated protein kinase (AMPK) signaling as potential pathways regulated by ZNF423 or ZNF423 rs9940645 SNP. Moreover, using clustered regularly interspaced short palindromic repeats/Cas9-engineered ZR75-1 breast cancer cells with different ZNF423 SNP genotypes, striking differences in cellular responses to metformin, either alone or in the combination of tamoxifen, were observed in both cell culture and the mouse xenograft model. CONCLUSIONS: We found that AMPK signaling is modulated by the ZNF423 rs9940645 SNP in estrogen and SERM-dependent fashion. The ZNF423 rs9940645 SNP affects metformin response in breast cancer and could be a potential biomarker for tailoring the metformin treatment.


Subject(s)
Breast Neoplasms , Metformin , AMP-Activated Protein Kinases/genetics , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Estrogens , Female , Humans , Metformin/pharmacology , Mice , Polymorphism, Single Nucleotide/genetics , Selective Estrogen Receptor Modulators , Tamoxifen
12.
Nat Commun ; 12(1): 2187, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846346

ABSTRACT

The RNA-sensing pathway contributes to type I interferon (IFN) production induced by DNA damaging agents. However, the potential involvement of RNA sensors in DNA repair is unknown. Here, we found that retinoic acid-inducible gene I (RIG-I), a key cytosolic RNA sensor that recognizes RNA virus and initiates the MAVS-IRF3-type I IFN signaling cascade, is recruited to double-stranded breaks (DSBs) and suppresses non-homologous end joining (NHEJ). Mechanistically, RIG-I interacts with XRCC4, and the RIG-I/XRCC4 interaction impedes the formation of XRCC4/LIG4/XLF complex at DSBs. High expression of RIG-I compromises DNA repair and sensitizes cancer cells to irradiation treatment. In contrast, depletion of RIG-I renders cells resistant to irradiation in vitro and in vivo. In addition, this mechanism suggests a protective role of RIG-I in hindering retrovirus integration into the host genome by suppressing the NHEJ pathway. Reciprocally, XRCC4, while suppressed for its DNA repair function, has a critical role in RIG-I immune signaling through RIG-I interaction. XRCC4 promotes RIG-I signaling by enhancing oligomerization and ubiquitination of RIG-I, thereby suppressing RNA virus replication in host cells. In vivo, silencing XRCC4 in mouse lung promotes influenza virus replication in mice and these mice display faster body weight loss, poorer survival, and a greater degree of lung injury caused by influenza virus infection. This reciprocal regulation of RIG-I and XRCC4 reveals a new function of RIG-I in suppressing DNA repair and virus integration into the host genome, and meanwhile endues XRCC4 with a crucial role in potentiating innate immune response, thereby helping host to prevail in the battle against virus.


Subject(s)
DEAD Box Protein 58/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/immunology , A549 Cells , Animals , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/radiation effects , DNA Repair/radiation effects , Dose-Response Relationship, Radiation , Genome, Human , HEK293 Cells , Humans , Mice , Radiation, Ionizing , Retroviridae/metabolism , Virus Replication/radiation effects
13.
Clin Transl Med ; 11(3): e341, 2021 03.
Article in English | MEDLINE | ID: mdl-33784003

ABSTRACT

PARP inhibitors induce DNA lesions, the repair of which are highly dependent on homologous recombination (HR), and preferentially kill HR- deficient cancers. However, cancer cells have developed several mechanisms to transform HR and confer drug resistance to PARP inhibition. Therefore, there is a great clinical interest in exploring new therapies that induce HR deficiency (HRD), thereby sensitizing cancer cells to PARP inhibitors. Here, we found that GSK2578215A, a high-selective and effective leucine-rich repeat kinase 2 (LRRK2) inhibitor, or LRRK2 depletion suppresses HR preventing the recruitment of RAD51 to DNA damage sites through disruption of the interaction of RAD51 and BRCA2. Moreover, LRRK2 inhibition or depletion increases the susceptibility of ovarian cancer cells to Olaparib in vitro and in vivo. In clinical specimens, LRRK2 high expression is high related with advanced clinical characteristics and poor survival of ovarian cancer patients. All these findings indicate ovarian cancers expressing high levels of LRRK2 are more resistant to treatment potentially through promoting HR. Furthermore, combination treatment with an LRRK2 and PARP inhibitor may be a novel strategy to improve the effectiveness of LRRK2 expression ovarian cancers.


Subject(s)
Antineoplastic Agents/therapeutic use , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Recombinational DNA Repair/drug effects , Animals , Cell Line, Tumor , DNA Breaks, Double-Stranded , Disease Models, Animal , Drug Synergism , Female , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Recombinational DNA Repair/genetics
14.
Nucleic Acids Res ; 49(6): 3322-3337, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33704464

ABSTRACT

RPA is a critical factor for DNA replication and replication stress response. Surprisingly, we found that chromatin RPA stability is tightly regulated. We report that the GDP/GTP exchange factor DOCK7 acts as a critical replication stress regulator to promote RPA stability on chromatin. DOCK7 is phosphorylated by ATR and then recruited by MDC1 to the chromatin and replication fork during replication stress. DOCK7-mediated Rac1/Cdc42 activation leads to the activation of PAK1, which subsequently phosphorylates RPA1 at S135 and T180 to stabilize chromatin-loaded RPA1 and ensure proper replication stress response. Moreover, DOCK7 is overexpressed in ovarian cancer and depleting DOCK7 sensitizes cancer cells to camptothecin. Taken together, our results highlight a novel role for DOCK7 in regulation of the replication stress response and highlight potential therapeutic targets to overcome chemoresistance in cancer.


Subject(s)
Chromatin/metabolism , DNA Replication , GTPase-Activating Proteins/physiology , Guanine Nucleotide Exchange Factors/physiology , Replication Protein A/metabolism , Animals , Cell Line, Tumor , DNA Repair , Female , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Phosphorylation , Proteolysis , Signal Transduction , Stress, Physiological/genetics , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
15.
DNA Repair (Amst) ; 100: 103063, 2021 04.
Article in English | MEDLINE | ID: mdl-33592542

ABSTRACT

The DNA replication stress-induced checkpoint activated through the TopBP1-ATR axis is important for maintaining genomic stability. However, the regulation of TopBP1 in DNA-damage responses remains unclear. In this study, we identify the deubiquitinating enzyme (DUB) USP13 as an important regulator of TopBP1. Mechanistically, USP13 binds to TopBP1 and stabilizes TopBP1 by deubiquitination. Depletion of USP13 impedes ATR activation and hypersensitizes cells to replication stress-inducing agents. Furthermore, high USP13 expression enhances the replication stress response, promotes cancer cell chemoresistance, and is correlated with poor prognosis of cancer patients. Overall, these findings suggest that USP13 is a novel deubiquitinating enzyme for TopBP1 and coordinates the replication stress response.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Checkpoints , DNA Damage , DNA Replication , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA/metabolism , HEK293 Cells , Humans
17.
Nucleic Acids Res ; 48(22): 12711-12726, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33237263

ABSTRACT

PrimPol has been recently identified as a DNA damage tolerant polymerase that plays an important role in replication stress response. However, the regulatory mechanisms of PrimPol are not well defined. In this study, we identify that the deubiquitinase USP36 interferes with degradation of PrimPol to regulate the replication stress response. Mechanistically, USP36 is deubiquitinated following DNA replication stress, which in turn facilitates its upregulation and interaction with PrimPol. USP36 deubiquitinates K29-linked polyubiquitination of PrimPol and increases its protein stability. Depletion of USP36 results in replication stress-related defects and elevates cell sensitivity to DNA-damage agents, such as cisplatin and olaparib. Moreover, USP36 expression positively correlates with the level of PrimPol protein and poor prognosis in patient samples. These findings indicate that the regulation of PrimPol K29-linked ubiquitination by USP36 plays a critical role in DNA replication stress and chemotherapy response.


Subject(s)
DNA Primase/genetics , DNA Replication/drug effects , DNA-Directed DNA Polymerase/genetics , Multifunctional Enzymes/genetics , Ovarian Neoplasms/genetics , Ubiquitin Thiolesterase/genetics , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage/drug effects , Deubiquitinating Enzymes/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Phthalazines/pharmacology , Piperazines/pharmacology , Polyubiquitin/genetics , Prognosis , Protein Stability/drug effects , Proteolysis/drug effects
18.
Nat Commun ; 11(1): 5362, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097710

ABSTRACT

Human C-terminal binding protein (CtBP)-interacting protein (CtIP) is a central regulator to initiate DNA end resection and homologous recombination (HR). Several studies have shown that post-translational modifications control the activity or expression of CtIP. However, it remains unclear whether and how cells restrain CtIP activity in unstressed cells and activate CtIP when needed. Here, we identify that USP52 directly interacts with and deubiquitinates CtIP, thereby promoting DNA end resection and HR. Mechanistically, USP52 removes the ubiquitination of CtIP to facilitate the phosphorylation and activation of CtIP at Thr-847. In addition, USP52 is phosphorylated by ATM at Ser-1003 after DNA damage, which enhances the catalytic activity of USP52. Furthermore, depletion of USP52 sensitizes cells to PARP inhibition in a CtIP-dependent manner in vitro and in vivo. Collectively, our findings reveal the key role of USP52 and the regulatory complexity of CtIP deubiquitination in DNA repair.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Endodeoxyribonucleases/metabolism , Exoribonucleases/metabolism , Ubiquitination/physiology , Animals , DNA Damage , Exoribonucleases/genetics , Female , HEK293 Cells , Homologous Recombination , Humans , Mice , Mice, Nude , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Xenograft Model Antitumor Assays
19.
Exp Mol Med ; 52(10): 1705-1714, 2020 10.
Article in English | MEDLINE | ID: mdl-33122806

ABSTRACT

DNA end resection has a key role in double-strand break repair and DNA replication. Defective DNA end resection can cause malfunctions in DNA repair and replication, leading to greater genomic instability. DNA end resection is initiated by MRN-CtIP generating short, 3'-single-stranded DNA (ssDNA). This newly generated ssDNA is further elongated by multiple nucleases and DNA helicases, such as EXO1, DNA2, and BLM. Effective DNA end resection is essential for error-free homologous recombination DNA repair, the degradation of incorrectly replicated DNA and double-strand break repair choice. Because of its importance in DNA repair, DNA end resection is strictly regulated. Numerous mechanisms have been reported to regulate the initiation, extension, and termination of DNA end resection. Here, we review the general process of DNA end resection and its role in DNA replication and repair pathway choice.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA Replication , Animals , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Humans , Protein Binding , Signal Transduction
20.
Mol Cell ; 79(5): 824-835.e5, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32649882

ABSTRACT

DNA-protein crosslinks (DPCs) are highly toxic DNA lesions that threaten genomic integrity. Recent findings highlight that SPRTN, a specialized DNA-dependent metalloprotease, is a central player in proteolytic cleavage of DPCs. Previous studies suggest that SPRTN deubiquitination is important for its chromatin association and activation. However, the regulation and consequences of SPRTN deubiquitination remain unclear. Here we report that, in response to DPC induction, the deubiquitinase VCPIP1/VCIP135 is phosphorylated and activated by ATM/ATR. VCPIP1, in turn, deubiquitinates SPRTN and promotes its chromatin relocalization. Deubiquitination of SPRTN is required for its subsequent acetylation, which promotes SPRTN relocation to the site of chromatin damage. Furthermore, Vcpip1 knockout mice are prone to genomic instability and premature aging. We propose a model where two sequential post-translational modifications (PTMs) regulate SPRTN chromatin accessibility to repair DPCs and maintain genomic stability and a healthy lifespan.


Subject(s)
Aging/genetics , DNA Repair , DNA-Binding Proteins/metabolism , Acetylation , Aging/metabolism , Animals , Cell Line , DNA Damage , DNA-Binding Proteins/genetics , Deubiquitinating Enzymes/metabolism , Endopeptidases/metabolism , Female , Genomic Instability , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Domains , Protein Processing, Post-Translational , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...