1
|
Iacovacci J, Brough R, Moughari FA, Alexander J, Kemp H, Tutt ANJ, Natrajan R, Lord CJ, Haider S. Proteogenomic discovery of RB1-defective phenocopy in cancer predicts disease outcome, response to treatment, and therapeutic targets. SCIENCE ADVANCES 2025; 11:eadq9495. [PMID: 40138429 PMCID: PMC11939072 DOI: 10.1126/sciadv.adq9495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Genomic defects caused by truncating mutations or deletions in the Retinoblastoma tumor suppressor gene (RB1) are frequently observed in many cancer types leading to dysregulation of the RB pathway. Here, we propose an integrative proteogenomic approach that predicts cancers with dysregulation in the RB pathway. A subset of these cancers, which we term as "RBness," lack RB1 genomic defects and yet phenocopy the transcriptional profile of RB1-defective cancers. We report RBness as a pan-cancer phenomenon, associated with patient outcome and chemotherapy response in multiple cancer types, and predictive of CDK4/6 inhibitor response in estrogen-positive breast cancer. Using RNA interference and a CRISPR-Cas9 screen in isogenic models, we find that RBness cancers also phenocopy synthetic lethal vulnerabilities of cells with RB1 genomic defects. In summary, our findings suggest that dysregulation of the RB pathway in cancers lacking RB1 genomic defects provides a molecular rationale for how these cancers could be treated.
Collapse
Affiliation(s)
- Jacopo Iacovacci
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano 20133, Italy
| | - Rachel Brough
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Fatemeh Ahmadi Moughari
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - John Alexander
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Harriet Kemp
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Andrew N. J. Tutt
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Christopher J. Lord
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
2
|
Stewart J, Krastev DB, Brough R, Zatreanu D, Song F, Baxter JS, Sridhar S, Frankum J, Konde A, Yang W, Haider S, Alexander J, Betteridge K, Gulati A, Attygalle AD, Vroobel K, Natrajan R, Khalique S, Roumeliotis TI, Choudhary JS, Yeung J, Wicks AJ, Marlow R, Banerjee S, Pettitt SJ, Tutt ANJ, Lord CJ. PPP2R1A mutations cause ATR inhibitor sensitivity in ovarian clear cell carcinoma. Oncogene 2025; 44:618-629. [PMID: 39939726 PMCID: PMC11850283 DOI: 10.1038/s41388-024-03265-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 02/14/2025]
Abstract
Identification of ARID1A/ATR synthetic lethality led to ATR inhibitor phase II trials in ovarian clear cell carcinoma (OCCC), a cancer of unmet need. Using multiple CRISPR-Cas9 mutagenesis and interference screens, we show that inactivation of protein phosphatase 2A (PP2A) subunits, including PPP2R1A, enhance ATRi sensitivity in ARID1A mutant OCCC. Analysis of a new OCCC cohort indicates that 52% possess oncogenic PPP2R1A p.R183 mutations and of these, one half possessed both ARID1A as well as PPP2R1A mutations. Using CRISPR-prime editing to generate new isogenic models of PPP2R1A mutant OCCC, we found that PPP2R1A p.R183W and p.R183P mutations cause ATRi-induced S phase stress, premature mitotic entry, genomic instability and ATRi sensitivity in OCCC tumour cells. p.R183 mutation also enhanced both in vitro and in vivo ATRi sensitivity in preclinical models of ARID1A mutant OCCC. These results argue for the assessment of PPP2R1A mutations as a biomarker of ATRi sensitivity.
Collapse
Affiliation(s)
- James Stewart
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Dragomir B Krastev
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Rachel Brough
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Diana Zatreanu
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Feifei Song
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Joseph S Baxter
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Sandhya Sridhar
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Jessica Frankum
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Asha Konde
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - William Yang
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Syed Haider
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - John Alexander
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Kai Betteridge
- Light microscopy Facility, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Aditi Gulati
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Ayoma D Attygalle
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Katherine Vroobel
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Rachael Natrajan
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Saira Khalique
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | | | - Jyoti S Choudhary
- Functional Proteomics Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Jason Yeung
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Andrew J Wicks
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Rebecca Marlow
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Stephen J Pettitt
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Andrew N J Tutt
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Christopher J Lord
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK.
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
| |
Collapse
|
3
|
Haider S, Brough R, Madera S, Iacovacci J, Gulati A, Wicks A, Alexander J, Pettitt SJ, Tutt ANJ, Lord CJ. The transcriptomic architecture of common cancers reflects synthetic lethal interactions. Nat Genet 2025; 57:522-529. [PMID: 40033056 PMCID: PMC11906352 DOI: 10.1038/s41588-025-02108-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
To maintain cell fitness, deleterious genetic alterations are buffered by compensatory changes in additional genes. In cancer, buffering processes could be targeted by synthetic lethality. However, despite the large-scale identification of synthetic lethal effects in preclinical models, evidence that these operate clinically is limited. This impedes the application of synthetic lethal approaches. By integrating molecular profiling data from >9,000 cancers with synthetic lethal screens, we show that transcriptomic buffering of tumor suppressor gene (TSG) loss by hyperexpression of synthetic lethal partners is a common phenomenon, extending to multiple TSGs and histotypes. Transcriptomic buffering is also notable in cancers that phenocopy TSG loss, such as BRCAness cancers, where expression of BRCA1/2 synthetic lethal genes correlates with clinical outcome. Synthetic lethal genes that exhibit transcriptomic buffering also represent more robust synthetic lethal effects. These observations have implications for understanding how tumor cells tolerate TSG loss, in part explain transcriptomic architectures in cancer and provide insight into target selection.
Collapse
Affiliation(s)
- Syed Haider
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK.
| | - Rachel Brough
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK
| | - Santiago Madera
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK
| | - Jacopo Iacovacci
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK
| | - Aditi Gulati
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK
| | - Andrew Wicks
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK
| | - John Alexander
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK
| | - Stephen J Pettitt
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK.
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK.
| | - Andrew N J Tutt
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK.
| | - Christopher J Lord
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, London, UK.
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK.
| |
Collapse
|
4
|
Palczewski MB, Kuschman HP, Hoffman BM, Kathiresan V, Yang H, Glynn SA, Wilson DL, Kool ET, Montfort WR, Chang J, Petenkaya A, Chronis C, Cundari TR, Sappa S, Islam K, McVicar DW, Fan Y, Chen Q, Meerzaman D, Sierk M, Thomas DD. Nitric oxide inhibits ten-eleven translocation DNA demethylases to regulate 5mC and 5hmC across the genome. Nat Commun 2025; 16:1732. [PMID: 39966373 PMCID: PMC11836389 DOI: 10.1038/s41467-025-56928-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
DNA methylation at cytosine bases (5-methylcytosine, 5mC) is a heritable epigenetic mark regulating gene expression. While enzymes that metabolize 5mC are well-characterized, endogenous signaling molecules that regulate DNA methylation machinery have not been described. We report that physiological nitric oxide (NO) concentrations reversibly inhibit the DNA demethylases TET and ALKBH2 by binding to the mononuclear non-heme iron atom forming a dinitrosyliron complex (DNIC) and preventing cosubstrates from binding. In cancer cells treated with exogenous NO, or endogenously synthesizing NO, 5mC and 5-hydroxymethylcytosine (5hmC) increase, with no changes in DNA methyltransferase activity. 5mC is also significantly increased in NO-producing patient-derived xenograft tumors from mice. Genome-wide methylome analysis of cells chronically treated with NO (10 days) shows enrichment of 5mC and 5hmC at gene-regulatory loci, correlating with altered expression of NO-regulated tumor-associated genes. Regulation of DNA methylation is distinctly different from canonical NO signaling and represents a unique epigenetic role for NO.
Collapse
Affiliation(s)
- Marianne B Palczewski
- Department of Pharmaceutical Sciences, University of Illinois Chicago, College of Pharmacy, Chicago, IL, USA
| | - Hannah Petraitis Kuschman
- Department of Pharmaceutical Sciences, University of Illinois Chicago, College of Pharmacy, Chicago, IL, USA
| | - Brian M Hoffman
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Venkatesan Kathiresan
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Hao Yang
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Sharon A Glynn
- Discipline of Pathology, University of Galway, College of Medicine, Nursing and Health Sciences, School of Medicine, Galway, Ireland
| | - David L Wilson
- Department of Chemistry, Stanford University, School of Humanities and Sciences, Stanford, CA, USA
| | - Eric T Kool
- Department of Chemistry, Stanford University, School of Humanities and Sciences, Stanford, CA, USA
| | - William R Montfort
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Jenny Chang
- Dr. Mary and Neal Cancer Center at Houston Methodist, Weill Cornell Medical College, Houston, NY, USA
| | - Aydolun Petenkaya
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering, Chicago, IL, USA
| | - Constantinos Chronis
- Department of Biochemistry and Molecular Genetics, University of Illinois Chicago, College of Medicine, Chicago, IL, USA
| | - Thomas R Cundari
- Department of Chemistry, University of North Texas, Denton, TX, USA
| | - Sushma Sappa
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kabirul Islam
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel W McVicar
- Cancer Innovation Laboratory, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Yu Fan
- National Cancer Institute, Center for Biomedical Informatics and Information Technology, Bethesda, USA
| | - Qingrong Chen
- National Cancer Institute, Center for Biomedical Informatics and Information Technology, Bethesda, USA
| | - Daoud Meerzaman
- Cancer Innovation Laboratory, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Michael Sierk
- Cancer Innovation Laboratory, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Douglas D Thomas
- Department of Pharmaceutical Sciences, University of Illinois Chicago, College of Pharmacy, Chicago, IL, USA.
| |
Collapse
|
5
|
Drew Y, Zenke FT, Curtin NJ. DNA damage response inhibitors in cancer therapy: lessons from the past, current status and future implications. Nat Rev Drug Discov 2025; 24:19-39. [PMID: 39533099 DOI: 10.1038/s41573-024-01060-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 11/16/2024]
Abstract
The DNA damage response (DDR) is a network of proteins that coordinate DNA repair and cell-cycle checkpoints to prevent damage being transmitted to daughter cells. DDR defects lead to genomic instability, which enables tumour development, but they also create vulnerabilities that can be used for cancer therapy. Historically, this vulnerability has been taken advantage of using DNA-damaging cytotoxic drugs and radiotherapy, which are more toxic to tumour cells than to normal tissues. However, the discovery of the unique sensitivity of tumours defective in the homologous recombination DNA repair pathway to PARP inhibition led to the approval of six PARP inhibitors worldwide and to a focus on making use of DDR defects through the development of other DDR-targeting drugs. Here, we analyse the lessons learnt from PARP inhibitor development and how these may be applied to new targets to maximize success. We explore why, despite so much research, no other DDR inhibitor class has been approved, and only a handful have advanced to later-stage clinical trials. We discuss why more reliable predictive biomarkers are needed, explore study design from past and current trials, and suggest alternative models for monotherapy and combination studies. Targeting multiple DDR pathways simultaneously and potential combinations with anti-angiogenic agents or immune checkpoint inhibitors are also discussed.
Collapse
Affiliation(s)
- Yvette Drew
- BC Cancer Vancouver Centre and Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frank T Zenke
- Research Unit Oncology, EMD Serono, Billerica, MA, USA
| | - Nicola J Curtin
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
6
|
Herrera-Abreu MT, Guan J, Khalid U, Ning J, Costa MR, Chan J, Li Q, Fortin JP, Wong WR, Perampalam P, Biton A, Sandoval W, Vijay J, Hafner M, Cutts R, Wilson G, Frankum J, Roumeliotis TI, Alexander J, Hickman O, Brough R, Haider S, Choudhary J, Lord CJ, Swain A, Metcalfe C, Turner NC. Inhibition of GPX4 enhances CDK4/6 inhibitor and endocrine therapy activity in breast cancer. Nat Commun 2024; 15:9550. [PMID: 39500869 PMCID: PMC11538343 DOI: 10.1038/s41467-024-53837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
CDK4/6 inhibition in combination with endocrine therapy is the standard of care for estrogen receptor (ER+) breast cancer, and although cytostasis is frequently observed, new treatment strategies that enhance efficacy are required. Here, we perform two independent genome-wide CRISPR screens to identify genetic determinants of CDK4/6 and endocrine therapy sensitivity. Genes involved in oxidative stress and ferroptosis modulate sensitivity, with GPX4 as the top sensitiser in both screens. Depletion or inhibition of GPX4 increases sensitivity to palbociclib and giredestrant, and their combination, in ER+ breast cancer models, with GPX4 null xenografts being highly sensitive to palbociclib. GPX4 perturbation additionally sensitises triple negative breast cancer (TNBC) models to palbociclib. Palbociclib and giredestrant induced oxidative stress and disordered lipid metabolism, leading to a ferroptosis-sensitive state. Lipid peroxidation is promoted by a peroxisome AGPAT3-dependent pathway in ER+ breast cancer models, rather than the classical ACSL4 pathway. Our data demonstrate that CDK4/6 and ER inhibition creates vulnerability to ferroptosis induction, that could be exploited through combination with GPX4 inhibitors, to enhance sensitivity to the current therapies in breast cancer.
Collapse
Affiliation(s)
- M T Herrera-Abreu
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - J Guan
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - U Khalid
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - J Ning
- Tumour Modelling Facility, Institute of Cancer Research, London, UK
| | - M R Costa
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - J Chan
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Q Li
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - J-P Fortin
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - W R Wong
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - P Perampalam
- ProCogia Inc. under contract to Hoffmann-La Roche Limited, Toronto, ON, Canada
| | - A Biton
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - W Sandoval
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - J Vijay
- Roche Informatics, Mississauga, ON, Canada
| | - M Hafner
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - R Cutts
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - G Wilson
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - J Frankum
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK
| | - T I Roumeliotis
- Functional proteomics team, The Institute of Cancer Research, London, UK
| | - J Alexander
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - O Hickman
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - R Brough
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK
| | - S Haider
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - J Choudhary
- Functional proteomics team, The Institute of Cancer Research, London, UK
| | - C J Lord
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK
| | - A Swain
- Tumour Modelling Facility, Institute of Cancer Research, London, UK
| | - C Metcalfe
- Genentech, 1 DNA Way, South San Francisco, CA, USA.
| | - N C Turner
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
- Breast Unit, The Royal Marsden Hospital, London, UK.
| |
Collapse
|
7
|
Zhang F, Sun J, Zhang L, Li R, Wang Y, Geng H, Shen C, Li L, Chen L. PARP inhibition leads to synthetic lethality with key splicing-factor mutations in myelodysplastic syndromes. Br J Cancer 2024; 131:231-242. [PMID: 38806724 PMCID: PMC11263539 DOI: 10.1038/s41416-024-02729-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Splicing factors are frequently mutated in patients with myelodysplastic syndromes and acute myeloid leukaemia. Recent studies have revealed convergent molecular defects caused by splicing factor mutations, among which R-loop dysregulation and resultant genome instability are suggested as contributing factors to disease progression. On the other hand, understanding how mutant cells survive upon aberrant R-loop formation and genome instability is essential for developing novel therapeutics. METHODS The immunoprecipitation was performed to identify R-loops in association with PARP1/poly-ADP-ribosylation. The western blot, immunofluorescence, and flow cytometry assays were used to test the cell viability, cell cycle arrest, apoptosis, and ATM activation in mutant cells following the treatment of the PARP inhibitor. The Srsf2(P95H) knock-in murine hematopoietic cells and MLL-AF9 transformed leukaemia model were generated to investigate the potential of the PARP inhibitor as a therapy for haematological malignancies. RESULTS The disease-causing mutations in SRSF2 activate PARP and elevate the overall poly-ADP-ribosylation levels of proteins in response to R-loop dysregulation. In accordance, mutant cells are more vulnerable to the PARP inhibitors in comparison to the wild-type counterpart. Notably, the synthetic lethality was further validated in the Srsf2(P95H) knock-in murine hematopoietic cell and MLL-AF9 leukaemia model. CONCLUSIONS Our findings suggest that mutant cells antagonise the genome threat caused by R-loop disruption by PARP activation, thus making PARP targeting a promising therapeutic strategy for myeloid cancers with mutations in SRSF2.
Collapse
Affiliation(s)
- Fangliang Zhang
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jianai Sun
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lei Zhang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Ruiqi Li
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yanzhen Wang
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Huichao Geng
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chao Shen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ling Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA.
| | - Liang Chen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
8
|
Thomas D, Palczewski M, Kuschman H, Hoffman B, Yang H, Glynn S, Wilson D, Kool E, Montfort W, Chang J, Petenkaya A, Chronis C, Cundari T, Sappa S, Islam K, McVicar D, Fan Y, Chen Q, Meerzaman D, Sierk M. Nitric oxide inhibits ten-eleven translocation DNA demethylases to regulate 5mC and 5hmC across the genome. RESEARCH SQUARE 2024:rs.3.rs-4131804. [PMID: 38645113 PMCID: PMC11030528 DOI: 10.21203/rs.3.rs-4131804/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
DNA methylation at cytosine bases of eukaryotic DNA (5-methylcytosine, 5mC) is a heritable epigenetic mark that can regulate gene expression in health and disease. Enzymes that metabolize 5mC have been well-characterized, yet the discovery of endogenously produced signaling molecules that regulate DNA methyl-modifying machinery have not been described. Herein, we report that the free radical signaling molecule nitric oxide (NO) can directly inhibit the Fe(II)/2-OG-dependent DNA demethylases ten-eleven translocation (TET) and human AlkB homolog 2 (ALKBH2). Physiologic NO concentrations reversibly inhibited TET and ALKBH2 demethylase activity by binding to the mononuclear non-heme iron atom which formed a dinitrosyliron complex (DNIC) preventing cosubstrates (2-OG and O2) from binding. In cancer cells treated with exogenous NO, or cells endogenously synthesizing NO, there was a global increase in 5mC and 5-hydroxymethylcytosine (5hmC) in DNA, the substrates for TET, that could not be attributed to increased DNA methyltransferase activity. 5mC was also elevated in NO-producing cell-line-derived mouse xenograft and patient-derived xenograft tumors. Genome-wide DNA methylome analysis of cells chronically treated with NO (10 days) demonstrated enrichment of 5mC and 5hmC at gene-regulatory loci which correlated to changes in the expression of NO-regulated tumor-associated genes. Regulation of DNA methylation is distinctly different from canonical NO signaling and represents a novel epigenetic role for NO.
Collapse
Affiliation(s)
| | - Marianne Palczewski
- University of Illinois Chicago, College of Pharmacy, Department of Pharmaceutical Sciences
| | - Hannah Kuschman
- University of Illinois Chicago, College of Pharmacy, Department of Pharmaceutical Sciences
| | | | - Hao Yang
- Weinberg College of Arts and Sciences, Northwestern University, Department of Chemistry
| | - Sharon Glynn
- University of Galway, College of Medicine, Nursing and Health Sciences, School of Medicine, D. of Pathology
| | | | - Eric Kool
- Stanford University, Department of Chemistry, School of Humanities and Sciences
| | | | - Jenny Chang
- Houston Methodist, Department of Medicine and Oncology, Weill Cornell Medical College
| | - Aydolun Petenkaya
- University of Illinois Chicago, College of Medicine, Biochemistry and Molecular Genetics
| | - Constantinos Chronis
- University of Illinois Chicago, College of Medicine, Biochemistry and Molecular Genetics
| | | | - Sushma Sappa
- University of Pittsburgh, Department of Chemistry
| | | | - Daniel McVicar
- National Institutes of Health, National Cancer Institute, Center for Cancer Research
| | - Yu Fan
- National Cancer Institute, Center for Biomedical Informatics and Information Technology
| | - Qingrong Chen
- National Cancer Institute, Center for Biomedical Informatics and Information Technology
| | - Daoud Meerzaman
- National Cancer Institute, Center for Biomedical Informatics and Information Technology
| | - Michael Sierk
- National Cancer Institute, Center for Biomedical Informatics and Information Technology
| |
Collapse
|
9
|
Ngoi NYL, Pilié PG, McGrail DJ, Zimmermann M, Schlacher K, Yap TA. Targeting ATR in patients with cancer. Nat Rev Clin Oncol 2024; 21:278-293. [PMID: 38378898 DOI: 10.1038/s41571-024-00863-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 02/22/2024]
Abstract
Pharmacological inhibition of the ataxia telangiectasia and Rad3-related protein serine/threonine kinase (ATR; also known as FRAP-related protein (FRP1)) has emerged as a promising strategy for cancer treatment that exploits synthetic lethal interactions with proteins involved in DNA damage repair, overcomes resistance to other therapies and enhances antitumour immunity. Multiple novel, potent ATR inhibitors are being tested in clinical trials using biomarker-directed approaches and involving patients across a broad range of solid cancer types; some of these inhibitors have now entered phase III trials. Further insight into the complex interactions of ATR with other DNA replication stress response pathway components and with the immune system is necessary in order to optimally harness the potential of ATR inhibitors in the clinic and achieve hypomorphic targeting of the various ATR functions. Furthermore, a deeper understanding of the diverse range of predictive biomarkers of response to ATR inhibitors and of the intraclass differences between these agents could help to refine trial design and patient selection strategies. Key challenges that remain in the clinical development of ATR inhibitors include the optimization of their therapeutic index and the development of rational combinations with these agents. In this Review, we detail the molecular mechanisms regulated by ATR and their clinical relevance, and discuss the challenges that must be addressed to extend the benefit of ATR inhibitors to a broad population of patients with cancer.
Collapse
Affiliation(s)
- Natalie Y L Ngoi
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Patrick G Pilié
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel J McGrail
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Katharina Schlacher
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
10
|
Liu M, Dong Q, Chen B, Liu K, Zhao Z, Wang Y, Zhuang S, Han H, Shi X, Jin Z, Hui Y, Gu Y. Synthetic viability induces resistance to immune checkpoint inhibitors in cancer cells. Br J Cancer 2023; 129:1339-1349. [PMID: 37620409 PMCID: PMC10575993 DOI: 10.1038/s41416-023-02404-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 08/05/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have revolutionized the treatment for multiple cancers. However, most of patients encounter resistance. Synthetic viability (SV) between genes could induce resistance. In this study, we established SV signature to predict the efficacy of ICI treatment for melanoma. METHODS We collected features and predicted SV gene pairs by random forest classifier. This work prioritized SV gene pairs based on CRISPR/Cas9 screens. SV gene pairs signature were constructed to predict the response to ICI for melanoma patients. RESULTS This study predicted robust SV gene pairs based on 14 features. Filtered by CRISPR/Cas9 screens, we identified 1,861 SV gene pairs, which were also related with prognosis across multiple cancer types. Next, we constructed the six SV pairs signature to predict resistance to ICI for melanoma patients. This study applied the six SV pairs signature to divide melanoma patients into high-risk and low-risk. High-risk melanoma patients were associated with worse response after ICI treatment. Immune landscape analysis revealed that high-risk melanoma patients had lower natural killer cells and CD8+ T cells infiltration. CONCLUSIONS In summary, the 14 features classifier accurately predicted robust SV gene pairs for cancer. The six SV pairs signature could predict resistance to ICI.
Collapse
Affiliation(s)
- Mingyue Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Qi Dong
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Bo Chen
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Kaidong Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zhangxiang Zhao
- The Sino-Russian Medical Research Center of Jinan University, The Institute of Chronic Disease of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuquan Wang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuping Zhuang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Huiming Han
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xingyang Shi
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zixin Jin
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yang Hui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China.
| | - Yunyan Gu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.
| |
Collapse
|
11
|
Zhang Q, Yang M, Zhang P, Wu B, Wei X, Li S. Deciphering gastric inflammation-induced tumorigenesis through multi-omics data and AI methods. Cancer Biol Med 2023; 21:j.issn.2095-3941.2023.0129. [PMID: 37589244 PMCID: PMC11033716 DOI: 10.20892/j.issn.2095-3941.2023.0129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/26/2023] [Indexed: 08/18/2023] Open
Abstract
Gastric cancer (GC), the fifth most common cancer globally, remains the leading cause of cancer deaths worldwide. Inflammation-induced tumorigenesis is the predominant process in GC development; therefore, systematic research in this area should improve understanding of the biological mechanisms that initiate GC development and promote cancer hallmarks. Here, we summarize biological knowledge regarding gastric inflammation-induced tumorigenesis, and characterize the multi-omics data and systems biology methods for investigating GC development. Of note, we highlight pioneering studies in multi-omics data and state-of-the-art network-based algorithms used for dissecting the features of gastric inflammation-induced tumorigenesis, and we propose translational applications in early GC warning biomarkers and precise treatment strategies. This review offers integrative insights for GC research, with the goal of paving the way to novel paradigms for GC precision oncology and prevention.
Collapse
Affiliation(s)
- Qian Zhang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Mingran Yang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Peng Zhang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Bowen Wu
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xiaosen Wei
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Shao Li
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| |
Collapse
|
12
|
Zong C, Zhang Z, Gao L, He J, Wang Y, Li Q, Liu X, Yang J, Chen D, Huang R, Zheng G, Jin X, Wei W, Jia R, Shen J. APOBEC3B coordinates R-loop to promote replication stress and sensitize cancer cells to ATR/Chk1 inhibitors. Cell Death Dis 2023; 14:348. [PMID: 37270643 DOI: 10.1038/s41419-023-05867-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 06/05/2023]
Abstract
The cytidine deaminase, Apolipoprotein B mRNA editing enzyme catalytic subunit 3B (APOBEC3B, herein termed A3B), is a critical mutation driver that induces genomic instability in cancer by catalyzing cytosine-to-thymine (C-to-T) conversion and promoting replication stress (RS). However, the detailed function of A3B in RS is not fully determined and it is not known whether the mechanism of A3B action can be exploited for cancer therapy. Here, we conducted an immunoprecipitation-mass spectrometry (IP-MS) study and identified A3B to be a novel binding component of R-loops, which are RNA:DNA hybrid structures. Mechanistically, overexpression of A3B exacerbated RS by promoting R-loop formation and altering the distribution of R-loops in the genome. This was rescued by the R-loop gatekeeper, Ribonuclease H1 (RNASEH1, herein termed RNH1). In addition, a high level of A3B conferred sensitivity to ATR/Chk1 inhibitors (ATRi/Chk1i) in melanoma cells, which was dependent on R-loop status. Together, our results provide novel insights into the mechanistic link between A3B and R-loops in the promotion of RS in cancer. This will inform the development of markers to predict the response of patients to ATRi/Chk1i.
Collapse
Affiliation(s)
- Chunyan Zong
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhe Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Li Gao
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jie He
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yiran Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qian Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoting Liu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
| | - Jie Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
| | - Di Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rui Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Guopei Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoliang Jin
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China
| | - Wu Wei
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
- Lingang Laboratory, Shanghai, 200031, China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
13
|
Pan M, Sha Y, Qiu J, Chen Y, Liu L, Luo M, Huang A, Xia J. RAD51 Inhibition Shows Antitumor Activity in Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:7905. [PMID: 37175611 PMCID: PMC10178757 DOI: 10.3390/ijms24097905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the major type of liver cancer, causes a high annual mortality worldwide. RAD51 is the critical recombinase responsible for homologous recombination (HR) repair in DNA damage. In this study, we identified that RAD51 was upregulated in HCC and that RAD51 silencing or inhibition reduced the proliferation, migration, and invasion of HCC cells and enhanced cell apoptosis and DNA damage. HCC cells with the combinatorial treatments of RAD51 siRNA or inhibitor and sorafenib demonstrated a synergistic effect in inhibiting HCC cell proliferation, migration, and invasion, as well as inducing cell apoptosis and DNA damage. Single RAD51 silencing or sorafenib reduced RAD51 protein expression and weakened HR efficiency, and their combination almost eliminated RAD51 protein expression and inhibited HR efficiency further. An in vivo tumor model confirmed the RAD51 inhibitor's antitumor activity and synergistic antitumor activity with sorafenib in HCC. RNA-Seq and gene set enrichment analysis (GSEA) in RAD51-inactivated Huh7 cells indicated that RAD51 knockdown upregulated cell apoptosis and G1/S DNA damage checkpoint pathways while downregulating mitotic spindle and homologous recombination pathways. Our findings suggest that RAD51 inhibition exhibits antitumor activities in HCC and synergizes with sorafenib. Targeting RAD51 may provide a novel therapeutic approach in HCC.
Collapse
Affiliation(s)
- Mingang Pan
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Yu Sha
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Jianguo Qiu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yunmeng Chen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Lele Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Muyu Luo
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Jie Xia
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|