1
|
Lee JH. ATM in immunobiology: From lymphocyte development to cancer immunotherapy. Transl Oncol 2025; 52:102268. [PMID: 39752906 PMCID: PMC11754496 DOI: 10.1016/j.tranon.2024.102268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/14/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Ataxia Telangiectasia Mutated (ATM) is a protein kinase traditionally known for its role in DNA damage response and cell cycle regulation. However, emerging research has revealed its multifaceted and crucial functions in the immune system. This comprehensive review explores the diverse roles of ATM in immune regulation, from lymphocyte development to its involvement in cancer immunotherapy. The review describes ATM's critical functions in V(D)J recombination and class switch recombination, highlighting its importance in adaptive immunity. It examines ATM's role in innate immunity, particularly in NF-κB signaling and cytokine production. Furthermore, the review analyzes the impact of ATM deficiency on oxidative stress and mitochondrial function in immune cells, providing insights into the immunological defects observed in Ataxia Telangiectasia (A-T). The article explores ATM's significance in maintaining hematopoietic stem cell function and its implications for bone marrow transplantation and gene therapy. Additionally, it addresses ATM's involvement in inflammation and immune senescence, linking DNA damage response to age-related immune decline. Finally, this review highlights the emerging role of ATM in cancer immunotherapy, where its inhibition shows promise in enhancing immune checkpoint blockade therapy. This review synthesizes current knowledge on ATM's functions in the immune system, offering insights into the pathophysiology of ATM-related disorders and potential therapeutic strategies for immune-related conditions and cancer immunotherapy.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
2
|
Khodyreva SN, Dyrkheeva NS, Lavrik OI. Proteins Associated with Neurodegenerative Diseases: Link to DNA Repair. Biomedicines 2024; 12:2808. [PMID: 39767715 PMCID: PMC11673744 DOI: 10.3390/biomedicines12122808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
The nervous system is susceptible to DNA damage and DNA repair defects, and if DNA damage is not repaired, neuronal cells can die, causing neurodegenerative diseases in humans. The overall picture of what is known about DNA repair mechanisms in the nervous system is still unclear. The current challenge is to use the accumulated knowledge of basic science on DNA repair to improve the treatment of neurodegenerative disorders. In this review, we summarize the current understanding of the function of DNA damage repair, in particular, the base excision repair and double-strand break repair pathways as being the most important in nervous system cells. We summarize recent data on the proteins involved in DNA repair associated with neurodegenerative diseases, with particular emphasis on PARP1 and ND-associated proteins, which are involved in DNA repair and have the ability to undergo liquid-liquid phase separation.
Collapse
Affiliation(s)
- Svetlana N. Khodyreva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 8 Akad. Lavrentyeva pr., Novosibirsk 630090, Russia;
| | - Nadezhda S. Dyrkheeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 8 Akad. Lavrentyeva pr., Novosibirsk 630090, Russia;
- Faculty of Natural Sciences, Novosibirsk State University, 2 Pirogova Str., Novosibirsk 630090, Russia
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 8 Akad. Lavrentyeva pr., Novosibirsk 630090, Russia;
- Faculty of Natural Sciences, Novosibirsk State University, 2 Pirogova Str., Novosibirsk 630090, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez pr., St. Petersburg 194223, Russia
| |
Collapse
|
3
|
Ashraf S, Deshpande N, Cheung Q, Asabere JB, Wong RJ, Gauthier AG, Parekh M, Adhikari Y, Melangath G, Jurkunas UV. Modulation of ATM enhances DNA repair in G2/M phase of cell cycle and averts senescence in Fuchs endothelial corneal dystrophy. Commun Biol 2024; 7:1482. [PMID: 39523410 PMCID: PMC11551145 DOI: 10.1038/s42003-024-07179-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Fuchs Endothelial Corneal Dystrophy (FECD) is an aging disorder characterized by expedited loss of corneal endothelial cells (CEnCs) and heightened DNA damage compared to normal CEnCs. We previously established that ultraviolet-A (UVA) light causes DNA damage and leads to FECD phenotype in a non-genetic mouse model. Here, we demonstrate that acute treatment with chemical stressor, menadione, or physiological stressors, UVA, and catechol estrogen (4-OHE2), results in an early and increased activation of ATM-mediated DNA damage response in FECD compared to normal CEnCs. Acute stress with UVA and 4OHE2 causes (i) greater cell-cycle arrest and DNA repair in G2/M phase, and (ii) greater cytoprotective senescence in NQO1-/- compared to NQO1+/+ cells, which was reversed upon ATM inhibition. Chronic stress with UVA and 4OHE2 results in ATM-driven cell-cycle arrest in G0/G1 phase, reduced DNA repair, and cytotoxic senescence, due to sustained damage. Likewise, UVA-induced cell-cycle reentry, gamma-H2AX foci, and senescence-associated heterochromatin were reduced in Atm-null mice. Remarkably, inhibiting ATM activation with KU-55933 restored DNA repair in G2/M phase and attenuated senescence in chronic cellular model of FECD lacking NQO1. This study provides insights into understanding the pivotal role of ATM in regulating cell-cycle, DNA repair, and senescence, in oxidative-stress disorders like FECD.
Collapse
Affiliation(s)
- Shazia Ashraf
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Neha Deshpande
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Queenie Cheung
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Jeffrey Boakye Asabere
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Raymond Jeff Wong
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Alex G Gauthier
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Mohit Parekh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yadav Adhikari
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Geetha Melangath
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Ula V Jurkunas
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
4
|
Hartlerode AJ, Mostafa AM, Orban SK, Benedeck R, Campbell K, Hoenerhoff MJ, Ferguson DO, Sekiguchi JM. Reduced levels of MRE11 cause disease phenotypes distinct from ataxia telangiectasia-like disorder. Hum Mol Genet 2024; 33:1605-1617. [PMID: 38888340 PMCID: PMC11373340 DOI: 10.1093/hmg/ddae101] [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: 12/21/2023] [Revised: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
The MRE11/RAD50/NBS1 (MRN) complex plays critical roles in cellular responses to DNA double-strand breaks. MRN is involved in end binding and processing, and it also induces cell cycle checkpoints by activating the ataxia-telangiectasia mutated (ATM) protein kinase. Hypomorphic pathogenic variants in the MRE11, RAD50, or NBS1 genes cause autosomal recessive genome instability syndromes featuring variable degrees of dwarfism, neurological defects, anemia, and cancer predisposition. Disease-associated MRN alleles include missense and nonsense variants, and many cause reduced protein levels of the entire MRN complex. However, the dramatic variability in the disease manifestation of MRN pathogenic variants is not understood. We sought to determine if low protein levels are a significant contributor to disease sequelae and therefore generated a transgenic murine model expressing MRE11 at low levels. These mice display dramatic phenotypes including small body size, severe anemia, and impaired DNA repair. We demonstrate that, distinct from ataxia telangiectasia-like disorder caused by MRE11 pathogenic missense or nonsense variants, mice and cultured cells expressing low MRE11 levels do not display the anticipated defects in ATM activation. Our findings indicate that ATM signaling can be supported by very low levels of the MRN complex and imply that defective ATM activation results from perturbation of MRN function caused by specific hypomorphic disease mutations. These distinct phenotypic outcomes underline the importance of understanding the impact of specific pathogenic MRE11 variants, which may help direct appropriate early surveillance for patients with these complicated disorders in a clinical setting.
Collapse
Affiliation(s)
- Andrea J Hartlerode
- Department of Pathology, University of Michigan Medical School, 109 Zina Pitcher Place, Rm 2067, Ann Arbor, MI 48109-2200, United States
- Department of Human Genetics, University of Michigan Medical School, 109 Zina Pitcher Place, Rm 2063, Ann Arbor, MI 48109-2200, United States
| | - Ahmed M Mostafa
- Department of Pathology, University of Michigan Medical School, 109 Zina Pitcher Place, Rm 2067, Ann Arbor, MI 48109-2200, United States
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Cairo, Egypt 11566
| | - Steven K Orban
- Department of Pathology, University of Michigan Medical School, 109 Zina Pitcher Place, Rm 2067, Ann Arbor, MI 48109-2200, United States
| | - Rachel Benedeck
- Department of Pathology, University of Michigan Medical School, 109 Zina Pitcher Place, Rm 2067, Ann Arbor, MI 48109-2200, United States
- Program in Biomedical Sciences PhD Program, University of Michigan Medical School, 1135 Catherine Street, Rm 2960, Ann Arbor, MI 48109, United States
| | - Koral Campbell
- Department of Pathology, University of Michigan Medical School, 109 Zina Pitcher Place, Rm 2067, Ann Arbor, MI 48109-2200, United States
- Program in Biomedical Sciences PhD Program, University of Michigan Medical School, 1135 Catherine Street, Rm 2960, Ann Arbor, MI 48109, United States
| | - Mark J Hoenerhoff
- In Vivo Animal Core, Unit for Laboratory Animal Medicine, University of Michigan Medical School, 2800 Plymouth Road, Ann Arbor, MI 48109, United States
| | - David O Ferguson
- Department of Pathology, University of Michigan Medical School, 109 Zina Pitcher Place, Rm 2067, Ann Arbor, MI 48109-2200, United States
| | - JoAnn M Sekiguchi
- Department of Human Genetics, University of Michigan Medical School, 109 Zina Pitcher Place, Rm 2063, Ann Arbor, MI 48109-2200, United States
| |
Collapse
|
5
|
Shen LP, Zhang WC, Deng JR, Qi ZH, Lin ZW, Wang ZD. Advances in the mechanism of small nucleolar RNA and its role in DNA damage response. Mil Med Res 2024; 11:53. [PMID: 39118131 PMCID: PMC11308251 DOI: 10.1186/s40779-024-00553-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Small nucleolar RNAs (snoRNAs) were previously regarded as a class of functionally conserved housekeeping genes, primarily involved in the regulation of ribosome biogenesis by ribosomal RNA (rRNA) modification. However, some of them are involved in several biological processes via complex molecular mechanisms. DNA damage response (DDR) is a conserved mechanism for maintaining genomic stability to prevent the occurrence of various human diseases. It has recently been revealed that snoRNAs are involved in DDR at multiple levels, indicating their relevant theoretical and clinical significance in this field. The present review systematically addresses four main points, including the biosynthesis and classification of snoRNAs, the mechanisms through which snoRNAs regulate target molecules, snoRNAs in the process of DDR, and the significance of snoRNA in disease diagnosis and treatment. It focuses on the potential functions of snoRNAs in DDR to help in the discovery of the roles of snoRNAs in maintaining genome stability and pathological processes.
Collapse
Affiliation(s)
- Li-Ping Shen
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wen-Cheng Zhang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jia-Rong Deng
- Graduate Collaborative Training Base of Academy of Military Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhen-Hua Qi
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zhong-Wu Lin
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zhi-Dong Wang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
- Graduate Collaborative Training Base of Academy of Military Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
6
|
Covino DA, Desimio MG, Giovinazzo A, de Oliveira BSP, Merolle M, Marazziti D, Pellegrini M, Doria M. Absence of ATM leads to altered NK cell function in mice. Clin Immunol 2024; 263:110233. [PMID: 38697554 DOI: 10.1016/j.clim.2024.110233] [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: 01/25/2024] [Revised: 03/28/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Ataxia-telangiectasia (A-T) is a rare disorder caused by genetic defects of A-T mutated (ATM) kinase, a key regulator of stress response, and characterized by neurodegeneration, immunodeficiency, and high incidence of cancer. Here we investigated NK cells in a mouse model of A-T (Atm-/-) showing that they are strongly impaired at killing tumor cells due to a block of early signaling events. On the other hand, in Atm-/- littermates with thymic lymphoma NK cell cytotoxicity is enhanced as compared with ATM-proficient mice, possibly via tumor-produced TNF-α. Results also suggest that expansion of exhausted NKG2D+ NK cells in Atm-/- mice is driven by low-level expression of stress-inducible NKG2D ligands, whereas development of thymoma expressing the high-affinity MULT1 ligand is associated with NKG2D down-regulation on NK cells. These results expand our understanding of immunodeficiency in A-T and encourage exploring NK cell biology in A-T patients in the attempt to identify cancer predictive biomarkers and novel therapeutic targets.
Collapse
Affiliation(s)
- Daniela Angela Covino
- Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria Giovanna Desimio
- Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alessandro Giovinazzo
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Monterotondo Scalo, 00015 Rome, Italy
| | | | - Matilde Merolle
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Monterotondo Scalo, 00015 Rome, Italy
| | - Daniela Marazziti
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Monterotondo Scalo, 00015 Rome, Italy
| | - Manuela Pellegrini
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Monterotondo Scalo, 00015 Rome, Italy
| | - Margherita Doria
- Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
7
|
Muvaffak A, Coleman KG. PARP inhibitor synthetic lethality in ATM biallelic mutant cancer cell lines is associated with BRCA1/2 and RAD51 downregulation. Front Oncol 2024; 14:1380633. [PMID: 38807759 PMCID: PMC11131418 DOI: 10.3389/fonc.2024.1380633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/10/2024] [Indexed: 05/30/2024] Open
Abstract
Background Ataxia telangiectasia-mutated (ATM) kinase is a central regulator of the DNA damage response (DDR) signaling pathway, and its function is critical for the maintenance of genomic stability in cells that coordinate a network of cellular processes, including DNA replication, DNA repair, and cell cycle progression. ATM is frequently mutated in human cancers, and approximately 3% of lung cancers have biallelic mutations in ATM, i.e., including 3.5% of lung adenocarcinomas (LUAD) and 1.4% of lung squamous cell carcinomas (LUSC). Methods We investigated the potential of targeting the DDR pathway in lung cancer as a potential therapeutic approach. In this context, we examined whether ATM loss is synthetically lethal with niraparib monotherapy. This exploration involved the use of hATM knockout (KO) isogenic cell lines containing hATM homozygous (-/-) and heterozygous (+/-) generated via CRISPR/Cas9 gene knockout technology in DLD-1, a human colorectal adenocarcinoma cell line. Subsequently, we extended our investigation to non-small cell lung cancer (NSCLC) patient derived xenograft (PDX) models for further validation of poly ADP-ribose polymerase inhibitor (PARPi) synthetic lethality in ATM mutant NSCLC models. Results Here, we demonstared that biallelic hATM deletion (-/-) in DLD-1 impairs homologous recombination (HR) repair function and sensitizes cells to the PARPi, niraparib. Niraparib also caused significant tumor regression in one-third of the NSCLC PDX models harboring deleterious biallelic ATM mutations. Loss of hATM (-/-) was concomitantly associated with low BRCA1 and BRCA2 protein expression in both the hATM (-/-) DLD-1 cell line and PARPi-sensitive ATM mutant NSCLC PDX models, suggesting a downstream effect on the impairment of HR-mediated DNA checkpoint signaling. Further analysis revealed that loss of ATM led to inhibition of phosphorylation of MRN (Mre11-Rad50-NBS1) complex proteins, which are required for ATM-mediated downstream phosphorylation of p53, BRCA1, and CHK2. Conclusions Taken together, our findings highlight that the synthetic lethality of niraparib in ATM-deficient tumors can be regulated through a subsequent effect on the modulation of BRCA1/2 expression and its effect on HR function.
Collapse
Affiliation(s)
- Asli Muvaffak
- Oncology, GlaxoSmithKline, Cambridge, MA, United States
| | | |
Collapse
|
8
|
Zhang C, Xiang C, Zhou K, Liu X, Qiao G, Zhao Y, Dong K, Sun K, Liu Z. Intestinal lysozyme1 deficiency alters microbiota composition and impacts host metabolism through the emergence of NAD +-secreting ASTB Qing110 bacteria. mSystems 2024; 9:e0121423. [PMID: 38364095 PMCID: PMC10949482 DOI: 10.1128/msystems.01214-23] [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: 11/14/2023] [Accepted: 01/26/2024] [Indexed: 02/18/2024] Open
Abstract
The intestine plays a pivotal role in nutrient absorption and host defense against pathogens, orchestrated in part by antimicrobial peptides secreted by Paneth cells. Among these peptides, lysozyme has multifaceted functions beyond its bactericidal activity. Here, we uncover the intricate relationship between intestinal lysozyme, the gut microbiota, and host metabolism. Lysozyme deficiency in mice led to altered body weight, energy expenditure, and substrate utilization, particularly on a high-fat diet. Interestingly, these metabolic benefits were linked to changes in the gut microbiota composition. Cohousing experiments revealed that the metabolic effects of lysozyme deficiency were microbiota-dependent. 16S rDNA sequencing highlighted differences in microbial communities, with ASTB_g (OTU60) highly enriched in lysozyme knockout mice. Subsequently, a novel bacterium, ASTB Qing110, corresponding to ASTB_g (OTU60), was isolated. Metabolomic analysis revealed that ASTB Qing110 secreted high levels of NAD+, potentially influencing host metabolism. This study sheds light on the complex interplay between intestinal lysozyme, the gut microbiota, and host metabolism, uncovering the potential role of ASTB Qing110 as a key player in modulating metabolic outcomes. IMPORTANCE The impact of intestinal lumen lysozyme on intestinal health is complex, arising from its multifaceted interactions with the gut microbiota. Lysozyme can both mitigate and worsen certain health conditions, varying with different scenarios. This underscores the necessity of identifying the specific bacterial responses elicited by lysozyme and understanding their molecular foundations. Our research reveals that a deficiency in intestinal lysozyme1 may offer protection against diet-induced obesity by altering bacterial populations. We discovered a strain of bacterium, ASTB Qing110, which secretes NAD+ and is predominantly found in lyz1-deficient mice. Qing110 demonstrates positive effects in both C. elegans and mouse models of ataxia telangiectasia. This study sheds light on the intricate role of lysozyme in influencing intestinal health.
Collapse
Affiliation(s)
- Chengye Zhang
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Chen Xiang
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Kaichen Zhou
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xingchen Liu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Guofeng Qiao
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Yabo Zhao
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Kemeng Dong
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Ke Sun
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Zhihua Liu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
9
|
Paull TT, Woolley PR. A-T neurodegeneration and DNA damage-induced transcriptional stress. DNA Repair (Amst) 2024; 135:103647. [PMID: 38377644 PMCID: PMC11707827 DOI: 10.1016/j.dnarep.2024.103647] [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: 12/20/2023] [Accepted: 02/08/2024] [Indexed: 02/22/2024]
Abstract
Loss of the ATM protein kinase in humans results in Ataxia-telangiectasia, a disorder characterized by childhood-onset neurodegeneration of the cerebellum as well as cancer predisposition and immunodeficiency. Although many aspects of ATM function are well-understood, the mechanistic basis of the progressive cerebellar ataxia that occurs in patients is not. Here we review recent progress related to the role of ATM in neurons and the cerebellum that comes from many sources: animal models, post-mortem brain tissue samples, and human neurons in culture. These observations have revealed new insights into the consequences of ATM loss on DNA damage, gene expression, and immune signaling in the brain. Many results point to the importance of reactive oxygen species as well as single-strand DNA breaks in the progression of molecular events leading to neuronal dysfunction. In addition, innate immunity signaling pathways appear to play a critical role in ATM functions in microglia, responding to various forms of nucleic acid sensors and regulating survival of neurons and other cell types. Overall, the results lead to an updated view of transcriptional stress and DNA damage resulting from ATM loss that results in changes in gene expression as well as neuroinflammation that contribute to the cerebellar neurodegeneration observed in patients.
Collapse
Affiliation(s)
- Tanya T Paull
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA.
| | - Phillip R Woolley
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA
| |
Collapse
|
10
|
Ajouaou Y, Magnani E, Madakashira B, Jenkins E, Sadler KC. atm Mutation and Oxidative Stress Enhance the Pre-Cancerous Effects of UHRF1 Overexpression in Zebrafish Livers. Cancers (Basel) 2023; 15:cancers15082302. [PMID: 37190230 DOI: 10.3390/cancers15082302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023] Open
Abstract
The ataxia-telangiectasia mutated (atm) gene is activated in response to genotoxic stress and leads to activation of the tp53 tumor suppressor gene which induces either senescence or apoptosis as tumor suppressive mechanisms. Atm also serves non-canonical functions in the response to oxidative stress and chromatin reorganization. We previously reported that overexpression of the epigenetic regulator and oncogene Ubiquitin Like with PHD and Ring Finger Domains 1 (UHRF1) in zebrafish hepatocytes resulted in tp53-dependent hepatocyte senescence, a small liver and larval lethality. We investigated the role of atm on UHRF1-mediated phenotypes by generating zebrafish atm mutants. atm-/- adults were viable but had reduction in fertility. Embryos developed normally but were protected from lethality caused by etoposide or H2O2 exposure and failed to fully upregulate Tp53 targets or oxidative stress response genes in response to these treatments. In contrast to the finding that Tp53 prevents the small liver phenotype caused by UHRF1 overexpression, atm mutation and exposure to H2O2 further reduced the liver size in UHRF1 overexpressing larvae whereas treatment with the antioxidant N-acetyl cysteine suppressed this phenotype. We conclude that UHRF1 overexpression in hepatocytes causes oxidative stress, and that loss of atm further enhances this, triggering elimination of these precancerous cells, leading to a small liver.
Collapse
Affiliation(s)
- Yousra Ajouaou
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Elena Magnani
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Bhavani Madakashira
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Eleanor Jenkins
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| |
Collapse
|
11
|
Tichy ED. Specialized Circuitry of Embryonic Stem Cells Promotes Genomic Integrity. Crit Rev Oncog 2023; 27:1-15. [PMID: 36734869 DOI: 10.1615/critrevoncog.2022042332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Embryonic stem cells (ESCs) give rise to all cell types of the organism. Given the importance of these cells in this process, ESCs must employ robust mechanisms to protect genomic integrity or risk catastrophic propagation of mutations throughout the organism. Should such an event occur in daughter cells that will eventually contribute to the germline, the overall species health could dramatically decline. This review describes several key mechanisms employed by ESCs that are unique to these cells, in order to maintain their genomic integrity. Additionally, the contributions of cell cycle regulators in modulating ESC differentiation, after DNA damage exposure, are also examined. Where data are available, findings reported in ESCs are extended to include observations described in induced pluripotent stem cells (IPSCs).
Collapse
Affiliation(s)
- Elisia D Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, 371 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104-6081
| |
Collapse
|
12
|
Kaur S, Kurokawa M. Regulation of Oocyte Apoptosis: A View from Gene Knockout Mice. Int J Mol Sci 2023; 24:ijms24021345. [PMID: 36674865 PMCID: PMC9861590 DOI: 10.3390/ijms24021345] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Apoptosis is a form of programmed cell death that plays a critical role in cellular homeostasis and development, including in the ovarian reserve. In humans, hundreds of thousands of oocytes are produced in the fetal ovary. However, the majority die by apoptosis before birth. After puberty, primordial follicles develop into mature follicles. While only a large dominant follicle is selected to ovulate, smaller ones undergo apoptosis. Despite numerous studies, the mechanism of oocyte death at the molecular level remains elusive. Over the last two and a half decades, many knockout mouse models disrupting key genes in the apoptosis pathway have been generated. In this review, we highlight some of the phenotypes and discuss distinct and overlapping roles of the apoptosis regulators in oocyte death and survival. We also review how the transcription factor p63 and its family members may trigger oocyte apoptosis in response to DNA damage.
Collapse
|
13
|
Osma-Garcia IC, Capitan-Sobrino D, Mouysset M, Aubert Y, Maloudi O, Turner M, Diaz-Muñoz MD. The splicing regulators TIA1 and TIAL1 are required for the expression of the DNA damage repair machinery during B cell lymphopoiesis. Cell Rep 2022; 41:111869. [PMID: 36543128 PMCID: PMC9794549 DOI: 10.1016/j.celrep.2022.111869] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 10/01/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
B cell lymphopoiesis requires dynamic modulation of the B cell transcriptome for timely coordination of somatic mutagenesis and DNA repair in progenitor B (pro-B) cells. Here, we show that, in pro-B cells, the RNA-binding proteins T cell intracellular antigen 1 (TIA1) and TIA1-like protein (TIAL1) act redundantly to enable developmental progression. They are global splicing regulators that control the expression of hundreds of mRNAs, including those involved in DNA damage repair. Mechanistically, TIA1 and TIAL1 bind to 5' splice sites for exon definition, splicing, and expression of DNA damage sensors, such as Chek2 and Rif1. In their absence, pro-B cells show exacerbated DNA damage, altered P53 expression, and increased cell death. Our study uncovers the importance of tight regulation of RNA splicing by TIA1 and TIAL1 for the expression of integrative transcriptional programs that control DNA damage sensing and repair during B cell development.
Collapse
Affiliation(s)
- Ines C. Osma-Garcia
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Inserm UMR1291, CNRS UMR5051, University Paul Sabatier, CHU Purpan, Toulouse 31024, France
| | - Dunja Capitan-Sobrino
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Inserm UMR1291, CNRS UMR5051, University Paul Sabatier, CHU Purpan, Toulouse 31024, France
| | - Mailys Mouysset
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Inserm UMR1291, CNRS UMR5051, University Paul Sabatier, CHU Purpan, Toulouse 31024, France
| | - Yann Aubert
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Inserm UMR1291, CNRS UMR5051, University Paul Sabatier, CHU Purpan, Toulouse 31024, France
| | - Orlane Maloudi
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Inserm UMR1291, CNRS UMR5051, University Paul Sabatier, CHU Purpan, Toulouse 31024, France
| | - Martin Turner
- Immunology Program, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Manuel D. Diaz-Muñoz
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Inserm UMR1291, CNRS UMR5051, University Paul Sabatier, CHU Purpan, Toulouse 31024, France,Corresponding author
| |
Collapse
|
14
|
Guan Y, Lin H, Leu NA, Ruthel G, Fuchs SY, Busino L, Luo M, Wang PJ. SCF ubiquitin E3 ligase regulates DNA double-strand breaks in early meiotic recombination. Nucleic Acids Res 2022; 50:5129-5144. [PMID: 35489071 PMCID: PMC9122608 DOI: 10.1093/nar/gkac304] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Homeostasis of meiotic DNA double strand breaks (DSB) is critical for germline genome integrity and homologous recombination. Here we demonstrate an essential role for SKP1, a constitutive subunit of the SCF (SKP1-Cullin-F-box) ubiquitin E3 ligase, in early meiotic processes. SKP1 restrains accumulation of HORMAD1 and the pre-DSB complex (IHO1-REC114-MEI4) on the chromosome axis in meiotic germ cells. Loss of SKP1 prior to meiosis leads to aberrant localization of DSB repair proteins and a failure in synapsis initiation in meiosis of both males and females. Furthermore, SKP1 is crucial for sister chromatid cohesion during the pre-meiotic S-phase. Mechanistically, FBXO47, a meiosis-specific F-box protein, interacts with SKP1 and HORMAD1 and targets HORMAD1 for polyubiquitination and degradation in HEK293T cells. Our results support a model wherein the SCF ubiquitin E3 ligase prevents hyperactive DSB formation through proteasome-mediated degradation of HORMAD1 and subsequent modulation of the pre-DSB complex during meiosis.
Collapse
Affiliation(s)
- Yongjuan Guan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Huijuan Lin
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
- Department of Tissue and Embryology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei Province, China
| | - N Adrian Leu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Gordon Ruthel
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Luca Busino
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mengcheng Luo
- Department of Tissue and Embryology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei Province, China
| | - P Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| |
Collapse
|
15
|
Chen K, Wang P, Chen J, Ying Y, Chen Y, Gilson E, Lu Y, Ye J. Loss of atm in Zebrafish as a Model of Ataxia-Telangiectasia Syndrome. Biomedicines 2022; 10:392. [PMID: 35203601 PMCID: PMC8962326 DOI: 10.3390/biomedicines10020392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/16/2022] Open
Abstract
Ataxia-telangiectasia mutated (ATM) is a key DNA damage signaling kinase that is mutated in humans with ataxia-telangiectasia (A-T) syndrome. This syndrome is characterized by neurodegeneration, immune abnormality, cancer predisposition, and premature aging. To better understand the function of ATM in vivo, we engineered a viable zebrafish model with a mutated atm gene. Zebrafish atm loss-of-function mutants show characteristic features of A-T-like motor disturbance, including coordination disorders, immunodeficiency, and tumorigenesis. The immunological disorder of atm homozygote fish is linked to the developmental blockade of hematopoiesis, which occurs at the adulthood stage and results in a decrease in infection defense but, with little effect on wound healing. Malignant neoplasms found in atm mutant fish were mainly nerve sheath tumors and myeloid leukemia, which rarely occur in A-T patients or Atm-/- mice. These results underscore the importance of atm during immune cell development. This zebrafish A-T model opens up a pathway to an improved understanding of the molecular basis of tumorigenesis in A-T and the cellular role of atm.
Collapse
Affiliation(s)
- Kehua Chen
- Department of Geriatrics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China; (K.C.); (P.W.); (J.C.); (Y.Y.)
- Medical Center on Aging of Ruijin Hospital, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
- International Laboratory in Hematology and Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China; (Y.C.); (E.G.)
| | - Peng Wang
- Department of Geriatrics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China; (K.C.); (P.W.); (J.C.); (Y.Y.)
- Medical Center on Aging of Ruijin Hospital, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
- International Laboratory in Hematology and Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China; (Y.C.); (E.G.)
| | - Jingrun Chen
- Department of Geriatrics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China; (K.C.); (P.W.); (J.C.); (Y.Y.)
- Medical Center on Aging of Ruijin Hospital, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Yiling Ying
- Department of Geriatrics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China; (K.C.); (P.W.); (J.C.); (Y.Y.)
- Medical Center on Aging of Ruijin Hospital, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
- International Laboratory in Hematology and Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China; (Y.C.); (E.G.)
| | - Yi Chen
- International Laboratory in Hematology and Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China; (Y.C.); (E.G.)
| | - Eric Gilson
- International Laboratory in Hematology and Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China; (Y.C.); (E.G.)
- Faculty of Medicine, University Côte d’Azur, CNRS, INSERM, IRCAN, 06107 Nice, France
- Department of Medical Genetics, CHU, 06107 Nice, France
| | - Yiming Lu
- Department of Geriatrics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China; (K.C.); (P.W.); (J.C.); (Y.Y.)
- Medical Center on Aging of Ruijin Hospital, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
- International Laboratory in Hematology and Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China; (Y.C.); (E.G.)
| | - Jing Ye
- Department of Geriatrics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China; (K.C.); (P.W.); (J.C.); (Y.Y.)
- Medical Center on Aging of Ruijin Hospital, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
- International Laboratory in Hematology and Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China; (Y.C.); (E.G.)
| |
Collapse
|
16
|
Douglas C, Maciulyte V, Zohren J, Snell DM, Mahadevaiah SK, Ojarikre OA, Ellis PJI, Turner JMA. CRISPR-Cas9 effectors facilitate generation of single-sex litters and sex-specific phenotypes. Nat Commun 2021; 12:6926. [PMID: 34862376 PMCID: PMC8642469 DOI: 10.1038/s41467-021-27227-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/05/2021] [Indexed: 11/09/2022] Open
Abstract
Animals are essential genetic tools in scientific research and global resources in agriculture. In both arenas, a single sex is often required in surplus. The ethical and financial burden of producing and culling animals of the undesired sex is considerable. Using the mouse as a model, we develop a synthetic lethal, bicomponent CRISPR-Cas9 strategy that produces male- or female-only litters with one hundred percent efficiency. Strikingly, we observe a degree of litter size compensation relative to control matings, indicating that our system has the potential to increase the yield of the desired sex in comparison to standard breeding designs. The bicomponent system can also be repurposed to generate postnatal sex-specific phenotypes. Our approach, harnessing the technological applications of CRISPR-Cas9, may be applicable to other vertebrate species, and provides strides towards ethical improvements for laboratory research and agriculture.
Collapse
Affiliation(s)
- Charlotte Douglas
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Valdone Maciulyte
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Jasmin Zohren
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Daniel M Snell
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | | | - Obah A Ojarikre
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | | | - James M A Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
17
|
Levi H, Bar E, Cohen-Adiv S, Sweitat S, Kanner S, Galron R, Mitiagin Y, Barzilai A. Dysfunction of cerebellar microglia in Ataxia-telangiectasia. Glia 2021; 70:536-557. [PMID: 34854502 DOI: 10.1002/glia.24122] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022]
Abstract
Ataxia-telangiectasia (A-T) is a multisystem autosomal recessive disease caused by mutations in the ATM gene and characterized by cerebellar atrophy, progressive ataxia, immunodeficiency, male and female sterility, radiosensitivity, cancer predisposition, growth retardation, insulin-resistant diabetes, and premature aging. ATM phosphorylates more than 1500 target proteins, which are involved in cell cycle control, DNA repair, apoptosis, modulation of chromatin structure, and other cytoplasmic as well as mitochondrial processes. In our quest to better understand the mechanisms by which ATM deficiency causes cerebellar degeneration, we hypothesized that specific vulnerabilities of cerebellar microglia underlie the etiology of A-T. Our hypothesis is based on the recent finding that dysfunction of glial cells affect a variety of process leading to impaired neuronal functionality (Song et al., 2019). Whereas astrocytes and neurons descend from the neural tube, microglia originate from the hematopoietic system, invade the brain at early embryonic stage, and become the innate immune cells of the central nervous system and important participants in development of synaptic plasticity. Here we demonstrate that microglia derived from Atm-/- mouse cerebellum display accelerated cell migration and are severely impaired in phagocytosis, secretion of neurotrophic factors, and mitochondrial activity, suggestive of apoptotic processes. Interestingly, no microglial impairment was detected in Atm-deficient cerebral cortex, and Atm deficiency had less impact on astroglia than microglia. Collectively, our findings validate the roles of glial cells in cerebellar attrition in A-T.
Collapse
Affiliation(s)
- Hadar Levi
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ela Bar
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Stav Cohen-Adiv
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Suzan Sweitat
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Kanner
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Galron
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yulia Mitiagin
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ari Barzilai
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
18
|
Lukaszewicz A, Lange J, Keeney S, Jasin M. De novo deletions and duplications at recombination hotspots in mouse germlines. Cell 2021; 184:5970-5984.e18. [PMID: 34793701 PMCID: PMC8616837 DOI: 10.1016/j.cell.2021.10.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/10/2021] [Accepted: 10/27/2021] [Indexed: 11/23/2022]
Abstract
Numerous DNA double-strand breaks (DSBs) arise during meiosis to initiate homologous recombination. These DSBs are usually repaired faithfully, but here, we uncover a distinct type of mutational event in which deletions form via joining of ends from two closely spaced DSBs (double cuts) within a single hotspot or at adjacent hotspots on the same or different chromatids. Deletions occur in normal meiosis but are much more frequent when DSB formation is dysregulated in the absence of the ATM kinase. Events between chromosome homologs point to multi-chromatid damage and aborted gap repair. Some deletions contain DNA from other hotspots, indicating that double cutting at distant sites creates substrates for insertional mutagenesis. End joining at double cuts can also yield tandem duplications or extrachromosomal circles. Our findings highlight the importance of DSB regulation and reveal a previously hidden potential for meiotic mutagenesis that is likely to affect human health and genome evolution.
Collapse
Affiliation(s)
- Agnieszka Lukaszewicz
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Julian Lange
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Scott Keeney
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Memorial Sloan Kettering Cancer Center, Howard Hughes Medical Institute, New York, NY 10065, USA.
| | - Maria Jasin
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
19
|
Cellular functions of the protein kinase ATM and their relevance to human disease. Nat Rev Mol Cell Biol 2021; 22:796-814. [PMID: 34429537 DOI: 10.1038/s41580-021-00394-2] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
The protein kinase ataxia telangiectasia mutated (ATM) is a master regulator of double-strand DNA break (DSB) signalling and stress responses. For three decades, ATM has been investigated extensively to elucidate its roles in the DNA damage response (DDR) and in the pathogenesis of ataxia telangiectasia (A-T), a human neurodegenerative disease caused by loss of ATM. Although hundreds of proteins have been identified as ATM phosphorylation targets and many important roles for this kinase have been identified, it is still unclear how ATM deficiency leads to the early-onset cerebellar degeneration that is common in all individuals with A-T. Recent studies suggest the existence of links between ATM deficiency and other cerebellum-specific neurological disorders, as well as the existence of broader similarities with more common neurodegenerative disorders. In this Review, we discuss recent structural insights into ATM regulation, and possible aetiologies of A-T phenotypes, including reactive oxygen species, mitochondrial dysfunction, alterations in transcription, R-loop metabolism and alternative splicing, defects in cellular proteostasis and metabolism, and potential pathogenic roles for hyper-poly(ADP-ribosyl)ation.
Collapse
|
20
|
Silva GLA, Tosi LRO, McCulloch R, Black JA. Unpicking the Roles of DNA Damage Protein Kinases in Trypanosomatids. Front Cell Dev Biol 2021; 9:636615. [PMID: 34422791 PMCID: PMC8377203 DOI: 10.3389/fcell.2021.636615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 07/13/2021] [Indexed: 12/31/2022] Open
Abstract
To preserve genome integrity when faced with DNA lesions, cells activate and coordinate a multitude of DNA repair pathways to ensure timely error correction or tolerance, collectively called the DNA damage response (DDR). These interconnecting damage response pathways are molecular signal relays, with protein kinases (PKs) at the pinnacle. Focused efforts in model eukaryotes have revealed intricate aspects of DNA repair PK function, including how they direct DDR pathways and how repair reactions connect to wider cellular processes, including DNA replication and transcription. The Kinetoplastidae, including many parasites like Trypanosoma spp. and Leishmania spp. (causative agents of debilitating, neglected tropical infections), exhibit peculiarities in several core biological processes, including the predominance of multigenic transcription and the streamlining or repurposing of DNA repair pathways, such as the loss of non-homologous end joining and novel operation of nucleotide excision repair (NER). Very recent studies have implicated ATR and ATM kinases in the DDR of kinetoplastid parasites, whereas DNA-dependent protein kinase (DNA-PKcs) displays uncertain conservation, questioning what functions it fulfills. The wide range of genetic manipulation approaches in these organisms presents an opportunity to investigate DNA repair kinase roles in kinetoplastids and to ask if further kinases are involved. Furthermore, the availability of kinase inhibitory compounds, targeting numerous eukaryotic PKs, could allow us to test the suitability of DNA repair PKs as novel chemotherapeutic targets. Here, we will review recent advances in the study of trypanosomatid DNA repair kinases.
Collapse
Affiliation(s)
- Gabriel L A Silva
- The Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.,Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luiz R O Tosi
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Richard McCulloch
- The Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer Ann Black
- The Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.,Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
21
|
Matsumoto Y, Asa ADDC, Modak C, Shimada M. DNA-Dependent Protein Kinase Catalytic Subunit: The Sensor for DNA Double-Strand Breaks Structurally and Functionally Related to Ataxia Telangiectasia Mutated. Genes (Basel) 2021; 12:genes12081143. [PMID: 34440313 PMCID: PMC8394720 DOI: 10.3390/genes12081143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 11/16/2022] Open
Abstract
The DNA-dependent protein kinase (DNA-PK) is composed of a DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and Ku70/Ku80 heterodimer. DNA-PK is thought to act as the “sensor” for DNA double-stranded breaks (DSB), which are considered the most deleterious type of DNA damage. In particular, DNA-PKcs and Ku are shown to be essential for DSB repair through nonhomologous end joining (NHEJ). The phenotypes of animals and human individuals with defective DNA-PKcs or Ku functions indicate their essential roles in these developments, especially in neuronal and immune systems. DNA-PKcs are structurally related to Ataxia–telangiectasia mutated (ATM), which is also implicated in the cellular responses to DSBs. DNA-PKcs and ATM constitute the phosphatidylinositol 3-kinase-like kinases (PIKKs) family with several other molecules. Here, we review the accumulated knowledge on the functions of DNA-PKcs, mainly based on the phenotypes of DNA-PKcs-deficient cells in animals and human individuals, and also discuss its relationship with ATM in the maintenance of genomic stability.
Collapse
|
22
|
Ando K, Nakagawara A. Acceleration or Brakes: Which Is Rational for Cell Cycle-Targeting Neuroblastoma Therapy? Biomolecules 2021; 11:biom11050750. [PMID: 34069817 PMCID: PMC8157238 DOI: 10.3390/biom11050750] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 11/27/2022] Open
Abstract
Unrestrained proliferation is a common feature of malignant neoplasms. Targeting the cell cycle is a therapeutic strategy to prevent unlimited cell division. Recently developed rationales for these selective inhibitors can be subdivided into two categories with antithetical functionality. One applies a “brake” to the cell cycle to halt cell proliferation, such as with inhibitors of cell cycle kinases. The other “accelerates” the cell cycle to initiate replication/mitotic catastrophe, such as with inhibitors of cell cycle checkpoint kinases. The fate of cell cycle progression or arrest is tightly regulated by the presence of tolerable or excessive DNA damage, respectively. This suggests that there is compatibility between inhibitors of DNA repair kinases, such as PARP inhibitors, and inhibitors of cell cycle checkpoint kinases. In the present review, we explore alterations to the cell cycle that are concomitant with altered DNA damage repair machinery in unfavorable neuroblastomas, with respect to their unique genomic and molecular features. We highlight the vulnerabilities of these alterations that are attributable to the features of each. Based on the assessment, we offer possible therapeutic approaches for personalized medicine, which are seemingly antithetical, but both are promising strategies for targeting the altered cell cycle in unfavorable neuroblastomas.
Collapse
Affiliation(s)
- Kiyohiro Ando
- Research Institute for Clinical Oncology, Saitama Cancer Center, 818 Komuro, Ina, Saitama 362-0806, Japan
- Correspondence: (K.A.); (A.N.); Tel.: +81-48-722-1111 (K.A.); +81-942-50-8829 (A.N.)
| | - Akira Nakagawara
- Saga International Carbon Particle Beam Radiation Cancer Therapy Center, Saga HIMAT Foundation, 3049 Harakoga-Machi, Saga 841-0071, Japan
- Correspondence: (K.A.); (A.N.); Tel.: +81-48-722-1111 (K.A.); +81-942-50-8829 (A.N.)
| |
Collapse
|
23
|
Lee JH, Ryu SW, Ender NA, Paull TT. Poly-ADP-ribosylation drives loss of protein homeostasis in ATM and Mre11 deficiency. Mol Cell 2021; 81:1515-1533.e5. [PMID: 33571423 PMCID: PMC8026623 DOI: 10.1016/j.molcel.2021.01.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/14/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
Loss of the ataxia-telangiectasia mutated (ATM) kinase causes cerebellum-specific neurodegeneration in humans. We previously demonstrated that deficiency in ATM activation via oxidative stress generates insoluble protein aggregates in human cells, reminiscent of protein dysfunction in common neurodegenerative disorders. Here, we show that this process is driven by poly-ADP-ribose polymerases (PARPs) and that the insoluble protein species arise from intrinsically disordered proteins associating with PAR-associated genomic sites in ATM-deficient cells. The lesions implicated in this process are single-strand DNA breaks dependent on reactive oxygen species, transcription, and R-loops. Human cells expressing Mre11 A-T-like disorder mutants also show PARP-dependent aggregation identical to ATM deficiency. Lastly, analysis of A-T patient cerebellum samples shows widespread protein aggregation as well as loss of proteins known to be critical in human spinocerebellar ataxias that is not observed in neocortex tissues. These results provide a hypothesis accounting for loss of protein integrity and cerebellum function in A-T.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA
| | - Seung W Ryu
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA
| | - Nicolette A Ender
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA
| | - Tanya T Paull
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA.
| |
Collapse
|
24
|
Natale VAI, Cole TJ, Rothblum-Oviatt C, Wright J, Crawford TO, Lefton-Greif MA, McGrath-Morrow SA, Schlechter H, Lederman HM. Growth in ataxia telangiectasia. Orphanet J Rare Dis 2021; 16:123. [PMID: 33691726 PMCID: PMC7945359 DOI: 10.1186/s13023-021-01716-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/26/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ataxia telangiectasia (A-T) is a DNA repair disorder that affects multiple body systems. Neurological problems and immunodeficiency are two important features of this disease. At this time, two main severity groups are defined in A-T: classic (the more severe form) and mild. Poor growth is a common problem in classic A-T. An objective of this study was to develop growth references for classic A-T. Another objective was to compare growth patterns in classic A-T and mild A-T with each other and with the general population, using the CDC growth references. A final objective was to examine the effects of chronic infection on height. RESULTS We found that classic A-T patients were smaller overall, and suffered from height and weight faltering that continued throughout childhood and adolescence. When compared to the CDC growth references, the median heights and weights for both male and female patients eventually fell to or below the 3rd centile on the CDC charts. Height faltering was more pronounced in females. Birthweight was lower in the classic A-T group compared to mild A-T and the general population, whereas birth length was not. Finally, we investigated height and BMI faltering in relation to number of infections and found no association. CONCLUSIONS Classic A-T appears to affect growth in utero. Although children appear to grow well in very early life, faltering begins early, and is unrelenting.
Collapse
Affiliation(s)
| | - Tim J Cole
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Jennifer Wright
- Division of Pediatric Allergy and Immunology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Thomas O Crawford
- Departments of Pediatrics and Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Maureen A Lefton-Greif
- Departments of Pediatrics, Otolaryngology-Head and Neck Surgery, and Physical Medicine and Rehabilitation, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sharon A McGrath-Morrow
- Children's Hospital of Philadelphia Division of Pulmonary Medicine and Sleep, Philadelphia, PA, USA
- Institute for Clinical and Translational Research, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Haley Schlechter
- Institute for Clinical and Translational Research, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Howard M Lederman
- Division of Pediatric Allergy and Immunology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
25
|
Linn E, Ghanem L, Bhakta H, Greer C, Avella M. Genes Regulating Spermatogenesis and Sperm Function Associated With Rare Disorders. Front Cell Dev Biol 2021; 9:634536. [PMID: 33665191 PMCID: PMC7921155 DOI: 10.3389/fcell.2021.634536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/20/2021] [Indexed: 12/26/2022] Open
Abstract
Spermatogenesis is a cell differentiation process that ensures the production of fertilizing sperm, which ultimately fuse with an egg to form a zygote. Normal spermatogenesis relies on Sertoli cells, which preserve cell junctions while providing nutrients for mitosis and meiosis of male germ cells. Several genes regulate normal spermatogenesis, some of which are not exclusively expressed in the testis and control multiple physiological processes in an organism. Loss-of-function mutations in some of these genes result in spermatogenesis and sperm functionality defects, potentially leading to the insurgence of rare genetic disorders. To identify genetic intersections between spermatogenesis and rare diseases, we screened public archives of human genetic conditions available on the Genetic and Rare Diseases Information Center (GARD), the Online Mendelian Inheritance in Man (OMIM), and the Clinical Variant (ClinVar), and after an extensive literature search, we identified 22 distinct genes associated with 21 rare genetic conditions and defective spermatogenesis or sperm function. These protein-coding genes regulate Sertoli cell development and function during spermatogenesis, checkpoint signaling pathways at meiosis, cellular organization and shape definition during spermiogenesis, sperm motility, and capacitation at fertilization. A number of these genes regulate folliculogenesis and oogenesis as well. For each gene, we review the genotype–phenotype association together with associative or causative polymorphisms in humans, and provide a description of the shared molecular mechanisms that regulate gametogenesis and fertilization obtained in transgenic animal models.
Collapse
Affiliation(s)
- Emma Linn
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Lillian Ghanem
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Hanisha Bhakta
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Cory Greer
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Matteo Avella
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| |
Collapse
|
26
|
Milanovic M, Shao Z, Estes VM, Wang XS, Menolfi D, Lin X, Lee BJ, Xu J, Cupo OM, Wang D, Zha S. FATC Domain Deletion Compromises ATM Protein Stability, Blocks Lymphocyte Development, and Promotes Lymphomagenesis. THE JOURNAL OF IMMUNOLOGY 2021; 206:1228-1239. [PMID: 33536256 DOI: 10.4049/jimmunol.2000967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/04/2021] [Indexed: 12/21/2022]
Abstract
Ataxia-telangiectasia mutated (ATM) kinase is a master regulator of the DNA damage response, and loss of ATM leads to primary immunodeficiency and greatly increased risk for lymphoid malignancies. The FATC domain is conserved in phosphatidylinositol-3-kinase-related protein kinases (PIKKs). Truncation mutation in the FATC domain (R3047X) selectively compromised reactive oxygen species-induced ATM activation in cell-free assays. In this article, we show that in mouse models, knock-in ATM-R3057X mutation (Atm RX , corresponding to R3047X in human ATM) severely compromises ATM protein stability and causes T cell developmental defects, B cell Ig class-switch recombination defects, and infertility resembling ATM-null. The residual ATM-R3057X protein retains minimal yet functional measurable DNA damage-induced checkpoint activation and significantly delays lymphomagenesis in Atm RX/RX mice compared with Atm -/- . Together, these results support a physiological role of the FATC domain in ATM protein stability and show that the presence of minimal residual ATM-R3057X protein can prevent growth retardation and delay tumorigenesis without restoring lymphocyte development and fertility.
Collapse
Affiliation(s)
- Maja Milanovic
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Zhengping Shao
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Verna M Estes
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Xiaobin S Wang
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032.,Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Demis Menolfi
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Xiaohui Lin
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Brian J Lee
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Jun Xu
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093
| | - Olivia M Cupo
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Dong Wang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093
| | - Shan Zha
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032; .,Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032.,Division of Pediatric Oncology, Hematology and Stem Cell Transplantation, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Immunology and Microbiology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| |
Collapse
|
27
|
Milanovic M, Sprinzen L, Menolfi D, Lee JH, Yamamoto K, Li Y, Lee BJ, Xu J, Estes VM, Wang D, Mckinnon PJ, Paull TT, Zha S. The Cancer-Associated ATM R3008H Mutation Reveals the Link between ATM Activation and Its Exchange. Cancer Res 2021; 81:426-437. [PMID: 33239428 PMCID: PMC8137556 DOI: 10.1158/0008-5472.can-20-2447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/14/2020] [Accepted: 11/18/2020] [Indexed: 11/16/2022]
Abstract
ATM kinase is a tumor suppressor and a master regulator of the DNA damage response. Most cancer-associated alterations to ATM are missense mutations at the PI3-kinase regulatory domain (PRD) or the kinase domain. Expression of kinase-dead (KD) ATM protein solely accelerates lymphomagenesis beyond ATM loss. To understand how PRD suppresses lymphomagenesis, we introduced the cancer-associated PRD mutation R3008H (R3016 in mouse) into mice. R3008H abrogated DNA damage- and oxidative stress-induced activation of ATM without consistently affecting ATM protein stability and recruitment. In contrast to the early embryonic lethality of AtmKD/KD mice, AtmR3016H (AtmR/R ) mice were viable, immunodeficient, and displayed spontaneous craniofacial abnormalities and delayed lymphomagenesis compared with Atm-/- controls. Mechanistically, R3008H rescued the tardy exchange of ATM-KD at DNA damage foci, indicating that PRD coordinates ATM activation with its exchange at DNA-breaks. Taken together, our results reveal a unique tumorigenesis profile for PRD mutations that is distinct from null or KD mutations. SIGNIFICANT: This study functionally characterizes the most common ATM missense mutation R3008H in cancer and identifies a unique role of PI3-kinase regulatory domain in ATM activation.
Collapse
Affiliation(s)
- Maja Milanovic
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York City, New York
| | - Lisa Sprinzen
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York City, New York
- Department of Pathology and Cell Biology, Pathobiology and Human Disease Graduate Program, Vagelos College for Physicians and Surgeons, Columbia University, New York, New York
| | - Demis Menolfi
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York City, New York
| | - Ji-Hoon Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas
| | - Kenta Yamamoto
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York City, New York
- Department of Pathology and Cell Biology, Pathobiology and Human Disease Graduate Program, Vagelos College for Physicians and Surgeons, Columbia University, New York, New York
| | - Yang Li
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Brian J Lee
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York City, New York
| | - Jun Xu
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California
| | - Verna M Estes
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York City, New York
| | - Dong Wang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California
| | - Peter J Mckinnon
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Tanya T Paull
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas
| | - Shan Zha
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York City, New York.
- Division of Pediatric Oncology, Hematology and Stem Cell Transplantation, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York City, New York
| |
Collapse
|
28
|
Rosenfield RL, Cooke DW, Radovick S. Puberty in the Female and Its Disorders. SPERLING PEDIATRIC ENDOCRINOLOGY 2021:528-626. [DOI: 10.1016/b978-0-323-62520-3.00016-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
29
|
Carrassa L, Colombo I, Damia G, Bertoni F. Targeting the DNA damage response for patients with lymphoma: Preclinical and clinical evidences. Cancer Treat Rev 2020; 90:102090. [DOI: 10.1016/j.ctrv.2020.102090] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
|
30
|
Sharma R, Lewis S, Wlodarski MW. DNA Repair Syndromes and Cancer: Insights Into Genetics and Phenotype Patterns. Front Pediatr 2020; 8:570084. [PMID: 33194896 PMCID: PMC7644847 DOI: 10.3389/fped.2020.570084] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022] Open
Abstract
DNA damage response is essential to human physiology. A broad spectrum of pathologies are displayed by individuals carrying monoallelic or biallelic loss-of-function mutations in DNA damage repair genes. DNA repair syndromes with biallelic disturbance of essential DNA damage response pathways manifest early in life with multi-systemic involvement and a high propensity for hematologic and solid cancers, as well as bone marrow failure. In this review, we describe classic biallelic DNA repair cancer syndromes arising from faulty single- and double-strand DNA break repair, as well as dysfunctional DNA helicases. These clinical entities include xeroderma pigmentosum, constitutional mismatch repair deficiency, ataxia telangiectasia, Nijmegen breakage syndrome, deficiencies of DNA ligase IV, NHEJ/Cernunnos, and ERCC6L2, as well as Bloom, Werner, and Rothmund-Thompson syndromes. To give an in-depth understanding of these disorders, we provide historical overview and discuss the interplay between complex biology and heterogeneous clinical manifestations.
Collapse
Affiliation(s)
- Richa Sharma
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Sara Lewis
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Marcin W. Wlodarski
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
31
|
Tran TN, Schimenti JC. A segregating human allele of SPO11 modeled in mice disrupts timing and amounts of meiotic recombination, causing oligospermia and a decreased ovarian reserve†. Biol Reprod 2020; 101:347-359. [PMID: 31074776 DOI: 10.1093/biolre/ioz089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 01/27/2023] Open
Abstract
A major challenge in medical genetics is to characterize variants of unknown significance (VUS). Doing so would help delineate underlying causes of disease and the design of customized treatments. Infertility has presented an especially difficult challenge with respect to not only determining if a given patient has a genetic basis, but also to identify the causative genetic factor(s). Though genome sequencing can identify candidate variants, in silico predictions of causation are not always sufficiently reliable so as to be actionable. Thus, experimental validation is crucial. Here, we describe the phenotype of mice containing a non-synonymous (proline-to-threonine at position 306) change in Spo11, corresponding to human SNP rs185545661. SPO11 is a topoisomerase-like protein that is essential for meiosis because it induces DNA double stranded breaks (DSBs) that stimulate pairing and recombination of homologous chromosomes. Although both male and female Spo11P306T/P306T mice were fertile, they had reduced sperm and oocytes, respectively. Spermatocyte chromosomes exhibited synapsis defects (especially between the X and Y chromosomes), elevated apoptotic cells, persistent markers of DSBs, and most importantly, fewer Type 1 crossovers that causes some chromosomes to have none. Spo11P306T/- mice were sterile and made fewer meiotic DSBs than Spo11+/- animals, suggesting that the Spo11P306T allele is a hypomorph and likely is delayed in making sufficient DSBs in a timely fashion. If the consequences are recapitulated in humans, it would predict phenotypes of premature ovarian failure, reduced sperm counts, and possible increased number of aneuploid gametes. These results emphasize the importance of deep phenotyping in order to accurately assess the impact of VUSs in reproduction genes.
Collapse
Affiliation(s)
- Tina N Tran
- Department of Biomedical Sciences and the Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - John C Schimenti
- Department of Biomedical Sciences and the Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| |
Collapse
|
32
|
Shiloh Y. The cerebellar degeneration in ataxia-telangiectasia: A case for genome instability. DNA Repair (Amst) 2020; 95:102950. [PMID: 32871349 DOI: 10.1016/j.dnarep.2020.102950] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/05/2020] [Accepted: 08/08/2020] [Indexed: 02/06/2023]
Abstract
Research on the molecular pathology of genome instability disorders has advanced our understanding of the complex mechanisms that safeguard genome stability and cellular homeostasis at large. Once the culprit genes and their protein products are identified, an ongoing dialogue develops between the research lab and the clinic in an effort to link specific disease symptoms to the functions of the proteins that are missing in the patients. Ataxi A-T elangiectasia (A-T) is a prominent example of this process. A-T's hallmarks are progressive cerebellar degeneration, immunodeficiency, chronic lung disease, cancer predisposition, endocrine abnormalities, segmental premature aging, chromosomal instability and radiation sensitivity. The disease is caused by absence of the powerful protein kinase, ATM, best known as the mobilizer of the broad signaling network induced by double-strand breaks (DSBs) in the DNA. In parallel, ATM also functions in the maintenance of the cellular redox balance, mitochondrial function and turnover and many other metabolic circuits. An ongoing discussion in the A-T field revolves around the question of which ATM function is the one whose absence is responsible for the most debilitating aspect of A-T - the cerebellar degeneration. This review suggests that it is the absence of a comprehensive role of ATM in responding to ongoing DNA damage induced mainly by endogenous agents. It is the ensuing deterioration and eventual loss of cerebellar Purkinje cells, which are very vulnerable to ATM absence due to a unique combination of physiological features, which kindles the cerebellar decay in A-T.
Collapse
Affiliation(s)
- Yosef Shiloh
- The David and Inez Myers Laboratory for Cancer Genetics, Department of Human Molecular Genetics and Biochemistry, Tel Aviv University Medical School, Tel Aviv, 69978, Israel.
| |
Collapse
|
33
|
Charaka V, Tiwari A, Pandita RK, Hunt CR, Pandita TK. Role of HP1β during spermatogenesis and DNA replication. Chromosoma 2020; 129:215-226. [PMID: 32651609 DOI: 10.1007/s00412-020-00739-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 11/25/2022]
Abstract
Heterochromatin protein 1β (HP1β), encoded by the Cbx1 gene, has been functionally linked to chromatin condensation, transcriptional regulation, and DNA damage repair. Here we report that testis-specific Cbx1 conditional knockout (Cbx1 cKO) impairs male germ cell development in mice. Depletion of HP1β negatively affected sperm maturation and increased seminiferous tubule degeneration in Cbx1 cKO mice. In addition, the spermatogonia have elevated γ-H2AX foci levels as do Cbx1 deficient mouse embryonic fibroblasts (MEFs) as compared to wild-type (WT) control MEFs. The increase in γ-H2AX foci in proliferating Cbx1 cKO cells indicates defective replication-dependent DNA damage repair. Depletion or loss of HP1β from human cells and MEFs increased DNA replication fork stalling and firing of new origins of replication, indicating defective DNA synthesis. Taken together, these results suggest that loss of HP1β in proliferating cells leads to DNA replication defects with associated DNA damage that impact spermatogenesis.
Collapse
Affiliation(s)
- Vijay Charaka
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Anjana Tiwari
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Raj K Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX, 77030, USA
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Clayton R Hunt
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Tej K Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
34
|
Yao Y, Li X, Chen W, Liu H, Mi L, Ren D, Mo A, Lu P. ATM Promotes RAD51-Mediated Meiotic DSB Repair by Inter-Sister-Chromatid Recombination in Arabidopsis. FRONTIERS IN PLANT SCIENCE 2020; 11:839. [PMID: 32670319 PMCID: PMC7329986 DOI: 10.3389/fpls.2020.00839] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/26/2020] [Indexed: 05/17/2023]
Abstract
Meiotic recombination ensures accurate homologous chromosome segregation during meiosis and generates novel allelic combinations among gametes. During meiosis, DNA double strand breaks (DSBs) are generated to facilitate recombination. To maintain genome integrity, meiotic DSBs must be repaired using appropriate DNA templates. Although the DNA damage response protein kinase Ataxia-telangiectasia mutated (ATM) has been shown to be involved in meiotic recombination in Arabidopsis, its mechanistic role is still unclear. In this study, we performed cytological analysis in Arabidopsis atm mutant, we show that there are fewer γH2AX foci, but more RAD51 and DMC1 foci on atm meiotic chromosomes. Furthermore, we observed an increase in meiotic Type I crossovers (COs) in atm. Our genetic analysis shows that the meiotic phenotype of atm rad51 double mutants is similar to the rad51 single mutant. Whereas, the atm dmc1 double mutant has a more severe chromosome fragmentation phenotype compared to both single mutants, suggesting that ATM functions in concert with RAD51, but in parallel to DMC1. Lastly, we show that atm asy1 double mutants also have more severe meiotic recombination defects. These data lead us to propose a model wherein ATM promotes RAD51-mediated meiotic DSB repair by inter-sister-chromatid (IS) recombination in Arabidopsis.
Collapse
Affiliation(s)
- Yuan Yao
- School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaojing Li
- School of Life Sciences, Fudan University, Shanghai, China
| | - Wanli Chen
- School of Life Sciences, Fudan University, Shanghai, China
| | - Hui Liu
- School of Life Sciences, Fudan University, Shanghai, China
| | - Limin Mi
- School of Life Sciences, Fudan University, Shanghai, China
| | - Ding Ren
- School of Life Sciences, Fudan University, Shanghai, China
| | - Aowei Mo
- School of Life Sciences, Fudan University, Shanghai, China
| | - Pingli Lu
- State Key Laboratory of Crop Stress Adaptation and Improvement, Key Laboratory of Plant Stress Biology, School of Life Sciences, Henan University, Kaifeng, China
| |
Collapse
|
35
|
Sulkowski PL, Oeck S, Dow J, Economos NG, Mirfakhraie L, Liu Y, Noronha K, Bao X, Li J, Shuch BM, King MC, Bindra RS, Glazer PM. Oncometabolites suppress DNA repair by disrupting local chromatin signalling. Nature 2020; 582:586-591. [PMID: 32494005 PMCID: PMC7319896 DOI: 10.1038/s41586-020-2363-0] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/28/2020] [Indexed: 01/06/2023]
Abstract
Deregulation of metabolism and disruption of genome integrity are hallmarks of cancer1. Increased levels of the metabolites 2-hydroxyglutarate, succinate and fumarate occur in human malignancies owing to somatic mutations in the isocitrate dehydrogenase-1 or -2 (IDH1 or IDH2) genes, or germline mutations in the fumarate hydratase (FH) and succinate dehydrogenase genes (SDHA, SDHB, SDHC and SDHD), respectively2-4. Recent work has made an unexpected connection between these metabolites and DNA repair by showing that they suppress the pathway of homology-dependent repair (HDR)5,6 and confer an exquisite sensitivity to inhibitors of poly (ADP-ribose) polymerase (PARP) that are being tested in clinical trials. However, the mechanism by which these oncometabolites inhibit HDR remains poorly understood. Here we determine the pathway by which these metabolites disrupt DNA repair. We show that oncometabolite-induced inhibition of the lysine demethylase KDM4B results in aberrant hypermethylation of histone 3 lysine 9 (H3K9) at loci surrounding DNA breaks, masking a local H3K9 trimethylation signal that is essential for the proper execution of HDR. Consequently, recruitment of TIP60 and ATM, two key proximal HDR factors, is substantially impaired at DNA breaks, with reduced end resection and diminished recruitment of downstream repair factors. These findings provide a mechanistic basis for oncometabolite-induced HDR suppression and may guide effective strategies to exploit these defects for therapeutic gain.
Collapse
Affiliation(s)
- Parker L Sulkowski
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Sebastian Oeck
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Medical Oncology, University of Duisburg-Essen, Essen, Germany
| | - Jonathan Dow
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Nicholas G Economos
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Lily Mirfakhraie
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Yanfeng Liu
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Katelyn Noronha
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Xun Bao
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Jing Li
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Brian M Shuch
- Department of Urology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Megan C King
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
36
|
Kafer GR, Cesare AJ. A Survey of Essential Genome Stability Genes Reveals That Replication Stress Mitigation Is Critical for Peri-Implantation Embryogenesis. Front Cell Dev Biol 2020; 8:416. [PMID: 32548123 PMCID: PMC7274024 DOI: 10.3389/fcell.2020.00416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/05/2020] [Indexed: 12/16/2022] Open
Abstract
Murine development demands that pluripotent epiblast stem cells in the peri-implantation embryo increase from approximately 120 to 14,000 cells between embryonic days (E) 4.5 and E7.5. This is possible because epiblast stem cells can complete cell cycles in under 3 h in vivo. To ensure conceptus fitness, epiblast cells must undertake this proliferative feat while maintaining genome integrity. How epiblast cells maintain genome health under such an immense proliferation demand remains unclear. To illuminate the contribution of genome stability pathways to early mammalian development we systematically reviewed knockout mouse data from 347 DDR and repair associated genes. Cumulatively, the data indicate that while many DNA repair functions are dispensable in embryogenesis, genes encoding replication stress response and homology directed repair factors are essential specifically during the peri-implantation stage of early development. We discuss the significance of these findings in the context of the unique proliferative demands placed on pluripotent epiblast stem cells.
Collapse
Affiliation(s)
| | - Anthony J. Cesare
- Genome Integrity Unit, Children’s Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
37
|
Chen J, Li P, Song L, Bai L, Huen MSY, Liu Y, Lu LY. 53BP1 loss rescues embryonic lethality but not genomic instability of BRCA1 total knockout mice. Cell Death Differ 2020; 27:2552-2567. [PMID: 32139898 PMCID: PMC7429965 DOI: 10.1038/s41418-020-0521-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/29/2022] Open
Abstract
BRCA1 is critical for DNA double-strand break (DSB) repair by homologous recombination (HR). BRCA1 deficient mice are embryonic lethal. Previous studies have shown that 53BP1 knockout (KO) rescues embryonic lethality of BRCA1 hypomorphic mutant mice by restoring HR. Here, we show that 53BP1 KO can partially rescue embryonic lethality of BRCA1 total KO mice, but HR is not restored in BRCA1-53BP1 double knockout (DKO) mice. As a result, BRCA1-53BP1 DKO cells are extremely sensitive to PARP inhibitors (PARPi). In addition to HR deficiency, BRCA1-53BP1 DKO cells have elevated microhomology-mediated end joining (MMEJ) activity and G2/M cell cycle checkpoint defects, causing severe genomic instability in these cells. Interestingly, BRCA1-53BP1 DKO mice rapidly develop thymic lymphoma that is 100% penetrant, which is not observed in any BRCA1 mutant mice rescued by 53BP1 KO. Taken together, our study reveals that 53BP1 KO can partially rescue embryonic lethality caused by complete BRCA1 loss without rescuing HR-related defects. This finding suggests that loss of 53BP1 can support the development of cancers with silenced BRCA1 expression without causing PARPi resistance.
Collapse
Affiliation(s)
- Jiyuan Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Li
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Licun Song
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Long Bai
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Michael S Y Huen
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yidan Liu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lin-Yu Lu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
38
|
Núñez FJ, Mendez FM, Kadiyala P, Alghamri MS, Savelieff MG, Garcia-Fabiani MB, Haase S, Koschmann C, Calinescu AA, Kamran N, Saxena M, Patel R, Carney S, Guo MZ, Edwards M, Ljungman M, Qin T, Sartor MA, Tagett R, Venneti S, Brosnan-Cashman J, Meeker A, Gorbunova V, Zhao L, Kremer DM, Zhang L, Lyssiotis CA, Jones L, Herting CJ, Ross JL, Hambardzumyan D, Hervey-Jumper S, Figueroa ME, Lowenstein PR, Castro MG. IDH1-R132H acts as a tumor suppressor in glioma via epigenetic up-regulation of the DNA damage response. Sci Transl Med 2020; 11:11/479/eaaq1427. [PMID: 30760578 DOI: 10.1126/scitranslmed.aaq1427] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 08/08/2018] [Accepted: 12/28/2018] [Indexed: 01/01/2023]
Abstract
Patients with glioma whose tumors carry a mutation in isocitrate dehydrogenase 1 (IDH1R132H) are younger at diagnosis and live longer. IDH1 mutations co-occur with other molecular lesions, such as 1p/19q codeletion, inactivating mutations in the tumor suppressor protein 53 (TP53) gene, and loss-of-function mutations in alpha thalassemia/mental retardation syndrome X-linked gene (ATRX). All adult low-grade gliomas (LGGs) harboring ATRX loss also express the IDH1R132H mutation. The current molecular classification of LGGs is based, partly, on the distribution of these mutations. We developed a genetically engineered mouse model harboring IDH1R132H, TP53 and ATRX inactivating mutations, and activated NRAS G12V. Previously, we established that ATRX deficiency, in the context of wild-type IDH1, induces genomic instability, impairs nonhomologous end-joining DNA repair, and increases sensitivity to DNA-damaging therapies. In this study, using our mouse model and primary patient-derived glioma cultures with IDH1 mutations, we investigated the function of IDH1R132H in the context of TP53 and ATRX loss. We discovered that IDH1R132H expression in the genetic context of ATRX and TP53 gene inactivation (i) increases median survival in the absence of treatment, (ii) enhances DNA damage response (DDR) via epigenetic up-regulation of the ataxia-telangiectasia-mutated (ATM) signaling pathway, and (iii) elicits tumor radioresistance. Accordingly, pharmacological inhibition of ATM or checkpoint kinases 1 and 2, essential kinases in the DDR, restored the tumors' radiosensitivity. Translation of these findings to patients with IDH1132H glioma harboring TP53 and ATRX loss could improve the therapeutic efficacy of radiotherapy and, consequently, patient survival.
Collapse
Affiliation(s)
- Felipe J Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Flor M Mendez
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mahmoud S Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Masha G Savelieff
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria B Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | - Neha Kamran
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Meghna Saxena
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rohin Patel
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marissa Z Guo
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marta Edwards
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mats Ljungman
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Environmental Health Science, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rebecca Tagett
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sriram Venneti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | - Alan Meeker
- Departments of Pathology, Oncology and Urology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Daniel M Kremer
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.,Graduate Program in Chemical Biology, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Li Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lindsey Jones
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Cameron J Herting
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA.,Graduate Division of Molecular and Systems Pharmacology, Emory University, Atlanta, GA 30322, USA
| | - James L Ross
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA.,Graduate Division of Cancer Biology, Emory University, Atlanta, GA 30322, USA
| | - Dolores Hambardzumyan
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shawn Hervey-Jumper
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria E Figueroa
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136-1000, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA. .,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA. .,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
39
|
Tan K, Song HW, Wilkinson MF. Single-cell RNAseq analysis of testicular germ and somatic cell development during the perinatal period. Development 2020; 147:dev.183251. [PMID: 31964773 DOI: 10.1242/dev.183251] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/03/2020] [Indexed: 12/22/2022]
Abstract
Pro-spermatogonia (SG) serve as the gateway to spermatogenesis. Using single-cell RNA sequencing (RNAseq), we studied the development of ProSG, their SG descendants and testicular somatic cells during the perinatal period in mice. We identified both gene and protein markers for three temporally distinct ProSG cell subsets, including a migratory cell population with a transcriptome distinct from the previously defined T1- and T2-ProSG stages. This intermediate (I)-ProSG subset translocates from the center of seminiferous tubules to the spermatogonial stem cell (SSC) 'niche' in its periphery soon after birth. We identified three undifferentiated SG subsets at postnatal day 7, each of which expresses distinct genes, including transcription factor and signaling genes. Two of these subsets have the characteristics of newly emergent SSCs. We also molecularly defined the development of Sertoli, Leydig and peritubular myoid cells during the perinatal period, allowing us to identify candidate signaling pathways acting between somatic and germ cells in a stage-specific manner during the perinatal period. Our study provides a rich resource for those investigating testicular germ and somatic cell developmental during the perinatal period.
Collapse
Affiliation(s)
- Kun Tan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Hye-Won Song
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Miles F Wilkinson
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA .,Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
40
|
Menolfi D, Zha S. ATM, ATR and DNA-PKcs kinases-the lessons from the mouse models: inhibition ≠ deletion. Cell Biosci 2020; 10:8. [PMID: 32015826 PMCID: PMC6990542 DOI: 10.1186/s13578-020-0376-x] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/14/2020] [Indexed: 01/11/2023] Open
Abstract
DNA damage, especially DNA double strand breaks (DSBs) and replication stress, activates a complex post-translational network termed DNA damage response (DDR). Our review focuses on three PI3-kinase related protein kinases-ATM, ATR and DNA-PKcs, which situate at the apex of the mammalian DDR. They are recruited to and activated at the DNA damage sites by their respective sensor protein complexes-MRE11/RAD50/NBS1 for ATM, RPA/ATRIP for ATR and KU70-KU80/86 (XRCC6/XRCC5) for DNA-PKcs. Upon activation, ATM, ATR and DNA-PKcs phosphorylate a large number of partially overlapping substrates to promote efficient and accurate DNA repair and to coordinate DNA repair with other DNA metabolic events (e.g., transcription, replication and mitosis). At the organism level, robust DDR is critical for normal development, aging, stem cell maintenance and regeneration, and physiological genomic rearrangements in lymphocytes and germ cells. In addition to endogenous damage, oncogene-induced replication stresses and genotoxic chemotherapies also activate DDR. On one hand, DDR factors suppress genomic instability to prevent malignant transformation. On the other hand, targeting DDR enhances the therapeutic effects of anti-cancer chemotherapy, which led to the development of specific kinase inhibitors for ATM, ATR and DNA-PKcs. Using mouse models expressing kinase dead ATM, ATR and DNA-PKcs, an unexpected structural function of these kinases was revealed, where the expression of catalytically inactive kinases causes more genomic instability than the loss of the proteins themselves. The spectrum of genomic instabilities and physiological consequences are unique for each kinase and depends on their activating complexes, suggesting a model in which the catalysis is coupled with DNA/chromatin release and catalytic inhibition leads to the persistence of the kinases at the DNA lesion, which in turn affects repair pathway choice and outcomes. Here we discuss the experimental evidences supporting this mode of action and their implications in the design and use of specific kinase inhibitors for ATM, ATR and DNA-PKcs for cancer therapy.
Collapse
Affiliation(s)
- Demis Menolfi
- Institute for Cancer Genetics, College of Physicians & Surgeons, Columbia University, New York, NY 10032 USA
| | - Shan Zha
- Institute for Cancer Genetics, College of Physicians & Surgeons, Columbia University, New York, NY 10032 USA
- Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY 10032 USA
- Division of Pediatric Oncology, Hematology and Stem Cell Transplantation, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY 10032 USA
| |
Collapse
|
41
|
Cheng ZY, Hsiao YT, Huang YP, Peng SF, Huang WW, Liu KC, Hsia TC, Way TD, Chung JG. Casticin Induces DNA Damage and Affects DNA Repair Associated Protein Expression in Human Lung Cancer A549 Cells (Running Title: Casticin Induces DNA Damage in Lung Cancer Cells). Molecules 2020; 25:E341. [PMID: 31952105 PMCID: PMC7024307 DOI: 10.3390/molecules25020341] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/09/2020] [Indexed: 12/20/2022] Open
Abstract
Casticin was obtained from natural plants, and it has been shown to exert biological functions; however, no report concerns the induction of DNA damage and repair in human lung cancer cells. The objective of this study was to investigate the effects and molecular mechanism of casticin on DNA damage and repair in human lung cancer A549 cells. Cell viability was determined by flow cytometric assay. The DNA damage was evaluated by 4',6-diamidino-2-phenylindole (DAPI) staining and electrophoresis which included comet assay and DNA gel electrophoresis. The protein levels associated with DNA damage and repair were analyzed by western blotting. The expression and translocation of p-H2A.X were observed by confocal laser microscopy. Casticin reduced total viable cell number and induced DNA condensation, fragmentation, and damage in A549 cells. Furthermore, casticin increased p-ATM at 6 h and increased p-ATR and BRCA1 at 6-24 h treatment but decreased p-ATM at 24-48 h, as well as decreased p-ATR and BRCA1 at 48 h. Furthermore, casticin decreased p-p53 at 6-24 h but increased at 48 h. Casticin increased p-H2A.X and MDC1 at 6-48 h treatment. In addition, casticin increased PARP (cleavage) at 6, 24, and 48 h treatment, DNA-PKcs and MGMT at 48 h in A549 cells. Casticin induced the expressions and nuclear translocation of p-H2AX in A549 cells by confocal laser microscopy. Casticin reduced cell number through DNA damage and condensation in human lung cancer A549 cells.
Collapse
Affiliation(s)
- Zheng-Yu Cheng
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| | - Yung-Ting Hsiao
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| | - Yi-Ping Huang
- Department of Physiology, College of Medicine, China Medical University, Taichung 404, Taiwan;
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
| | - Wen-Wen Huang
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| | - Kuo-Ching Liu
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan;
| | - Te-Chun Hsia
- Department of Respiratory Therapy, China Medical University, Taichung 404, Taiwan;
- Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Tzong-Der Way
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan; (Z.-Y.C.); (Y.-T.H.); (S.-F.P.); (W.-W.H.)
| |
Collapse
|
42
|
Innes CL, Hesse JE, Morales AJ, Helmink BA, Schurman SH, Sleckman BP, Paules RS. DNA damage responses in murine Pre-B cells with genetic deficiencies in damage response genes. Cell Cycle 2020; 19:67-83. [PMID: 31757180 PMCID: PMC6927727 DOI: 10.1080/15384101.2019.1693118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 10/29/2019] [Accepted: 11/07/2019] [Indexed: 01/11/2023] Open
Abstract
DNA damage can be generated in multiple ways from genotoxic and physiologic sources. Genotoxic damage is known to disrupt cellular functions and is lethal if not repaired properly. We compare the transcriptional programs activated in response to genotoxic DNA damage induced by ionizing radiation (IR) in abl pre-B cells from mice deficient in DNA damage response (DDR) genes Atm, Mre11, Mdc1, H2ax, 53bp1, and DNA-PKcs. We identified a core IR-specific transcriptional response that occurs in abl pre-B cells from WT mice and compared the response of the other genotypes to the WT response. We also identified genotype specific responses and compared those to each other. The WT response includes many processes involved in lymphocyte development and immune response, as well as responses associated with the molecular mechanisms of cancer, such as TP53 signaling. As expected, there is a range of similarity in transcriptional profiles in comparison to WT cells, with Atm-/- cells being the most different from the core WT DDR and Mre11 hypomorph (Mre11A/A) cells also very dissimilar to WT and other genotypes. For example, NF-kB-related signaling and CD40 signaling are deficient in both Atm-/- and Mre11A/A cells, but present in all other genotypes. In contrast, IR-induced TP53 signaling is seen in the Mre11A/A cells, while these responses are not seen in the Atm-/- cells. By examining the similarities and differences in the signaling pathways in response to IR when specific genes are absent, our results further illustrate the contribution of each gene to the DDR. The microarray gene expression data discussed in this paper have been deposited in NCBI's Gene Expression Omnibus (GEO) (http://www.ncbi.nlm.nih.gov/geo/) and are accessible under accession number GSE116388.
Collapse
Affiliation(s)
- Cynthia L. Innes
- Environmental Stress and Cancer Group, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Jill E. Hesse
- Environmental Stress and Cancer Group, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Abigail J. Morales
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Beth A. Helmink
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shepherd H. Schurman
- Clinical Research Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Barry P. Sleckman
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard S. Paules
- Environmental Stress and Cancer Group, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
43
|
van Gastel J, Leysen H, Santos-Otte P, Hendrickx JO, Azmi A, Martin B, Maudsley S. The RXFP3 receptor is functionally associated with cellular responses to oxidative stress and DNA damage. Aging (Albany NY) 2019; 11:11268-11313. [PMID: 31794429 PMCID: PMC6932917 DOI: 10.18632/aging.102528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/18/2019] [Indexed: 12/19/2022]
Abstract
DNA damage response (DDR) processes, often caused by oxidative stress, are important in aging and -related disorders. We recently showed that G protein-coupled receptor (GPCR) kinase interacting protein 2 (GIT2) plays a key role in both DNA damage and oxidative stress. Multiple tissue analyses in GIT2KO mice demonstrated that GIT2 expression affects the GPCR relaxin family peptide 3 receptor (RXFP3), and is thus a therapeutically-targetable system. RXFP3 and GIT2 play similar roles in metabolic aging processes. Gaining a detailed understanding of the RXFP3-GIT2 functional relationship could aid the development of novel anti-aging therapies. We determined the connection between RXFP3 and GIT2 by investigating the role of RXFP3 in oxidative stress and DDR. Analyzing the effects of oxidizing (H2O2) and DNA-damaging (camptothecin) stressors on the interacting partners of RXFP3 using Affinity Purification-Mass Spectrometry, we found multiple proteins linked to DDR and cell cycle control. RXFP3 expression increased in response to DNA damage, overexpression, and Relaxin 3-mediated stimulation of RXFP3 reduced phosphorylation of DNA damage marker H2AX, and repair protein BRCA1, moderating DNA damage. Our data suggests an RXFP3-GIT2 system that could regulate cellular degradation after DNA damage, and could be a novel mechanism for mitigating the rate of age-related damage accumulation.
Collapse
Affiliation(s)
- Jaana van Gastel
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Paula Santos-Otte
- Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Jhana O Hendrickx
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Abdelkrim Azmi
- Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Bronwen Martin
- Faculty of Pharmaceutical, Veterinary and Biomedical Science, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| |
Collapse
|
44
|
Riabinska A, Lehrmann D, Jachimowicz RD, Knittel G, Fritz C, Schmitt A, Geyer A, Heneweer C, Wittersheim M, Frenzel LP, Torgovnick A, Wiederstein JL, Wunderlich CM, Ortmann M, Paillard A, Wößmann W, Borkhardt A, Burdach S, Hansmann ML, Rosenwald A, Perner S, Mall G, Klapper W, Merseburg A, Krüger M, Grüll H, Persigehl T, Wunderlich FT, Peifer M, Utermöhlen O, Büttner R, Beleggia F, Reinhardt HC. ATM activity in T cells is critical for immune surveillance of lymphoma in vivo. Leukemia 2019; 34:771-786. [PMID: 31690822 DOI: 10.1038/s41375-019-0618-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/25/2019] [Accepted: 10/24/2019] [Indexed: 11/09/2022]
Abstract
The proximal DNA damage response kinase ATM is frequently inactivated in human malignancies. Germline mutations in the ATM gene cause Ataxia-telangiectasia (A-T), characterized by cerebellar ataxia and cancer predisposition. Whether ATM deficiency impacts on tumor initiation or also on the maintenance of the malignant state is unclear. Here, we show that Atm reactivation in initially Atm-deficient B- and T cell lymphomas induces tumor regression. We further find a reduced T cell abundance in B cell lymphomas from Atm-defective mice and A-T patients. Using T cell-specific Atm-knockout models, as well as allogeneic transplantation experiments, we pinpoint impaired immune surveillance as a contributor to cancer predisposition and development. Moreover, we demonstrate that Atm-deficient T cells display impaired proliferation capacity upon stimulation, due to replication stress. Altogether, our data indicate that T cell-specific restoration of ATM activity or allogeneic hematopoietic stem cell transplantation may prevent lymphomagenesis in A-T patients.
Collapse
Affiliation(s)
- Arina Riabinska
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany.
| | - Daria Lehrmann
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Ron Daniel Jachimowicz
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Gero Knittel
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Christian Fritz
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Anna Schmitt
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Aenne Geyer
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Carola Heneweer
- Department of Radiology, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Maike Wittersheim
- Institute of Pathology, University Hospital of Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Lukas P Frenzel
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50937, Germany
| | - Alessandro Torgovnick
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50937, Germany
| | - Janica Lea Wiederstein
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50937, Germany
| | | | - Monika Ortmann
- Institute of Pathology, University Hospital of Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Arlette Paillard
- Intitute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, 50937, Germany
| | - Wilhelm Wößmann
- Department of Pediatric Hematology and Oncology, Justus-Liebig-University, Giessen, 35390, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Heinrich Heine University, Medical Faculty, Düsseldorf, 40225, Germany
| | - Stefan Burdach
- Children's Cancer Research Center and Department of Pediatrics, Rechts der Isar Hospital, Technical University of Munich and Comprehensive Cancer Center Munich, Munich, 80333, Germany
| | - Martin-Leo Hansmann
- Institute of Pathology, University of Frankfurt, Medical School, Frankfurt, 60590, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center Mainfranken, Würzburg, 97080, Germany
| | - Sven Perner
- Institute of Pathology, University Hospital Schleswig-Holstein, Lübeck, 23538, Germany
| | - Gita Mall
- Institute of Forensic Medicine, University Hospital Jena, Jena, 07743, Germany
| | - Wolfram Klapper
- Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Kiel, 24105, Germany
| | - Andrea Merseburg
- Experimental Neurophysiology, German Center for Neurodegenerative Diseases, Bonn, 53175, Germany
| | - Marcus Krüger
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50937, Germany
| | - Holger Grüll
- Department of Radiology, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Thorsten Persigehl
- Department of Radiology, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | | | - Martin Peifer
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, 50937, Germany.,Department of Translational Genomics, Cologne, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Olaf Utermöhlen
- Intitute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, 50937, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, 50937, Germany.,German Center for Infection Research (DZIF), Bonn-Cologne, Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital of Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, 50937, Germany
| | - Filippo Beleggia
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany
| | - Hans Christian Reinhardt
- Clinic I of Internal Medicine, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, 50937, Germany. .,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50937, Germany. .,Center for Molecular Medicine Cologne, University of Cologne, Cologne, 50937, Germany.
| |
Collapse
|
45
|
Warren R, Domm W, Yee M, Campbell A, Malone J, Wright T, Mayer-Pröschel M, O'Reilly MA. Ataxia-telangiectasia mutated is required for the development of protective immune memory after influenza A virus infection. Am J Physiol Lung Cell Mol Physiol 2019; 317:L591-L601. [PMID: 31509427 PMCID: PMC6879906 DOI: 10.1152/ajplung.00031.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 08/21/2019] [Accepted: 09/01/2019] [Indexed: 01/26/2023] Open
Abstract
Ataxia-telangiectasia (A-T), caused by mutations in the A-T mutated (ATM) gene, is a neurodegenerative disorder affecting ∼1 in 40,000-100,000 children. Recurrent respiratory infections are a common and challenging comorbidity, often leading to the development of bronchiectasis in individuals with A-T. The role of ATM in development of immune memory in response to recurrent respiratory viral infections is not well understood. Here, we infect wild-type (WT) and Atm-null mice with influenza A virus (IAV; HKx31, H3N2) and interrogate the immune memory with secondary infections designed to challenge the B cell memory response with homologous infection (HKx31) and the T cell memory response with heterologous infection (PR8, H1N1). Although Atm-null mice survived primary and secondary infections, they lost more weight than WT mice during secondary infections. This enhanced morbidity to secondary infections was not attributed to failure to effectively clear virus during the primary IAV infection. Instead, Atm-null mice developed persistent peribronchial inflammation, characterized in part by clusters of B220+ B cells. Additionally, levels of select serum antibodies to hemagglutinin-specific IAV were significantly lower in Atm-null than WT mice. These findings reveal that Atm is required to mount a proper memory response to a primary IAV infection, implying that vaccination of children with A-T by itself may not be sufficiently protective against respiratory viral infections.
Collapse
Affiliation(s)
- Rachel Warren
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - William Domm
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Min Yee
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Andrew Campbell
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Jane Malone
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Terry Wright
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Margot Mayer-Pröschel
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Michael A O'Reilly
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| |
Collapse
|
46
|
Ho U, Luff J, James A, Lee CS, Quek H, Lai HC, Apte S, Lim YC, Lavin MF, Roberts TL. SMG1 heterozygosity exacerbates haematopoietic cancer development in Atm null mice by increasing persistent DNA damage and oxidative stress. J Cell Mol Med 2019; 23:8151-8160. [PMID: 31565865 PMCID: PMC6850945 DOI: 10.1111/jcmm.14685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 04/25/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022] Open
Abstract
Suppressor of morphogenesis in genitalia 1 (SMG1) and ataxia telangiectasia mutated (ATM) are members of the PI3‐kinase like–kinase (PIKK) family of proteins. ATM is a well‐established tumour suppressor. Loss of one or both alleles of ATM results in an increased risk of cancer development, particularly haematopoietic cancer and breast cancer in both humans and mouse models. In mice, total loss of SMG1 is embryonic lethal and loss of a single allele results in an increased rate of cancer development, particularly haematopoietic cancers and lung cancer. In this study, we generated mice deficient in Atm and lacking one allele of Smg1, Atm−/−Smg1gt/+ mice. These mice developed cancers more rapidly than either of the parental genotypes, and all cancers were haematopoietic in origin. The combined loss of Smg1 and Atm resulted in a higher level of basal DNA damage and oxidative stress in tissues than loss of either gene alone. Furthermore, Atm−/−Smg1gt/+ mice displayed increased cytokine levels in haematopoietic tissues compared with wild‐type animals indicating the development of low‐level inflammation and a pro‐tumour microenvironment. Overall, our data demonstrated that combined loss of Atm expression and decreased Smg1 expression increases haematopoietic cancer development.
Collapse
Affiliation(s)
- Uda Ho
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St Lucia, Qld, Australia
| | - John Luff
- UQCCR, University of Queensland, Brisbane, Qld, Australia
| | - Alexander James
- The Ingham Institute for Applied Medical Research and School of Medicine, Western Sydney University, Liverpool, NSW, Australia
| | - Cheok Soon Lee
- The Ingham Institute for Applied Medical Research and School of Medicine, Western Sydney University, Liverpool, NSW, Australia.,South West Sydney Clinical School, UNSW Sydney, Liverpool, NSW, Australia.,Department of Anatomical Pathology, Molecular Pathology Laboratory, Liverpool Hospital, Liverpool, NSW, Australia
| | - Hazel Quek
- UQCCR, University of Queensland, Brisbane, Qld, Australia.,QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Hui-Chi Lai
- The Ingham Institute for Applied Medical Research and School of Medicine, Western Sydney University, Liverpool, NSW, Australia.,South West Sydney Clinical School, UNSW Sydney, Liverpool, NSW, Australia
| | - Simon Apte
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Yi Chieh Lim
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia.,Danish Cancer Society Research Centre, Copenhagen, Denmark
| | - Martin F Lavin
- UQCCR, University of Queensland, Brisbane, Qld, Australia
| | - Tara L Roberts
- UQCCR, University of Queensland, Brisbane, Qld, Australia.,The Ingham Institute for Applied Medical Research and School of Medicine, Western Sydney University, Liverpool, NSW, Australia.,South West Sydney Clinical School, UNSW Sydney, Liverpool, NSW, Australia
| |
Collapse
|
47
|
Lanz MC, Dibitetto D, Smolka MB. DNA damage kinase signaling: checkpoint and repair at 30 years. EMBO J 2019; 38:e101801. [PMID: 31393028 PMCID: PMC6745504 DOI: 10.15252/embj.2019101801] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/03/2019] [Accepted: 07/24/2019] [Indexed: 12/27/2022] Open
Abstract
From bacteria to mammalian cells, damaged DNA is sensed and targeted by DNA repair pathways. In eukaryotes, kinases play a central role in coordinating the DNA damage response. DNA damage signaling kinases were identified over two decades ago and linked to the cell cycle checkpoint concept proposed by Weinert and Hartwell in 1988. Connections between the DNA damage signaling kinases and DNA repair were scant at first, and the initial perception was that the importance of these kinases for genome integrity was largely an indirect effect of their roles in checkpoints, DNA replication, and transcription. As more substrates of DNA damage signaling kinases were identified, it became clear that they directly regulate a wide range of DNA repair factors. Here, we review our current understanding of DNA damage signaling kinases, delineating the key substrates in budding yeast and humans. We trace the progress of the field in the last 30 years and discuss our current understanding of the major substrate regulatory mechanisms involved in checkpoint responses and DNA repair.
Collapse
Affiliation(s)
- Michael Charles Lanz
- Department of Molecular Biology and GeneticsWeill Institute for Cell and Molecular BiologyCornell UniversityIthacaNYUSA
| | - Diego Dibitetto
- Department of Molecular Biology and GeneticsWeill Institute for Cell and Molecular BiologyCornell UniversityIthacaNYUSA
| | - Marcus Bustamante Smolka
- Department of Molecular Biology and GeneticsWeill Institute for Cell and Molecular BiologyCornell UniversityIthacaNYUSA
| |
Collapse
|
48
|
Li Z, Chen Y, Tang M, Li Y, Zhu WG. Regulation of DNA damage-induced ATM activation by histone modifications. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s42764-019-00004-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
49
|
Xing M, Oksenych V. Genetic interaction between DNA repair factors PAXX, XLF, XRCC4 and DNA-PKcs in human cells. FEBS Open Bio 2019; 9:1315-1326. [PMID: 31141305 PMCID: PMC6609761 DOI: 10.1002/2211-5463.12681] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/03/2019] [Accepted: 05/28/2019] [Indexed: 01/26/2023] Open
Abstract
DNA double-strand breaks (DSBs) are highly cytotoxic lesions, and unrepaired or misrepaired DSBs can lead to various human diseases, including immunodeficiency, neurological abnormalities, growth retardation, and cancer. Nonhomologous end joining (NHEJ) is the major DSB repair pathway in mammals. Ku70 and Ku80 are DSB sensors that facilitate the recruitment of downstream factors, including protein kinase DNA-dependent protein kinase, catalytic subunit (DNA-PKcs), structural components [X-ray repair cross-complementing protein 4 (XRCC4), XRCC4-like factor (XLF), and paralogue of XRCC4 and XLF (PAXX)], and DNA ligase IV (LIG4), which complete DNA repair. DSBs also trigger the activation of the DNA damage response pathway, in which protein kinase ataxia-telangiectasia mutated (ATM) phosphorylates multiple substrates, including histone H2AX. Traditionally, research on NHEJ factors was performed using in vivo mouse models and murine cells. However, the current knowledge of the genetic interactions between NHEJ factors in human cells is incomplete. Here, we obtained genetically modified human HAP1 cell lines, which lacked one or two NHEJ factors, including LIG4, XRCC4, XLF, PAXX, DNA-PKcs, DNA-PKcs/XRCC4, and DNA-PKcs/PAXX. We examined the genomic instability of HAP1 cells, as well as their sensitivity to DSB-inducing agents. In addition, we determined the genetic interaction between XRCC4 paralogues (XRCC4, XLF, and PAXX) and DNA-PKcs. We found that in human cells, XLF, but not PAXX or XRCC4, genetically interacts with DNA-PKcs. Moreover, ATM possesses overlapping functions with DNA-PKcs, XLF, and XRCC4, but not with PAXX in response to DSBs. Finally, NHEJ-deficient HAP1 cells show increased chromosomal and chromatid breaks, when compared to the WT parental control. Overall, we found that HAP1 is a suitable model to study the genetic interactions in human cells.
Collapse
Affiliation(s)
- Mengtan Xing
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Valentyn Oksenych
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
50
|
ATM, DNA-PKcs and ATR: shaping development through the regulation of the DNA damage responses. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s42764-019-00003-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|