1
|
Lyu SI, Fretter C, Eckel HNC, Knipper K, Schultheis AM, Büttner R, Quaas A, Klussmann JP, Simon AG. High expression of H2AX/γ-H2AX is associated with distinct biological pathway alterations and shorter survival in oropharyngeal squamous cell carcinoma. Oral Oncol 2025; 161:107171. [PMID: 39756240 DOI: 10.1016/j.oraloncology.2024.107171] [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: 11/09/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND The histone gene H2AX and its phosphorylated protein γ-H2AX play a crucial role in the DNA damage response. This study investigates the expression of H2AX mRNA and its phosphorylated γ-H2AX protein in oropharyngeal squamous cell carcinoma (OPSCC), its association with distinct biological pathway alterations and its potential as a biomarker. MATERIALS AND METHODS Expression of H2AX mRNA in 76 OPSCC from The Cancer Genome Atlas (TCGA) cohort was analyzed. Patients were stratified into H2AXhigh- and H2AXlow OPSCC based on a survival-associated cutoff. Differentially expressed genes were identified using DESeq2, followed by pathway enrichment analyses. Immunohistochemical staining of γ-H2AX protein expression was performed on an independent cohort of 209 OPSCC, followed by survival and Cox regression analyses. RESULTS High H2AX mRNA expression was a significant prognostic factor associated with shorter OS in the TCGA OPSCC cohort (HR 4.77, p = 0.04). In H2AXhigh tumors, differential gene expression analysis revealed upregulation of genes regulating DNA repair and cell cycle (CDK1, CCNB1, ZWINT). High γ-H2AX protein expression was significantly associated with HPV-negative OPSCC (p = 0.005), and remained an independent predictor of poor survival in the total OPSCC cohort (HR 2.24, p = 0.03) and particularly in HPV-negative patients (HR 3.67, p = 0.007). CONCLUSION H2AX/γ-H2AX expression is a potential prognostic biomarker in OPSCC, with elevated levels indicating poor survival, especially in HPV-negative cases. These findings suggest distinct molecular behaviors in OPSCC based on H2AX expression and highlight the need for further investigation into its therapeutic implications.
Collapse
Affiliation(s)
- Su Ir Lyu
- Institute of Pathology, University Hospital of Cologne, University Cologne, Faculty of Medicine, Kerpener Strasse 62, 50937 Cologne, Germany.
| | - Caroline Fretter
- Institute of Pathology, University Hospital of Cologne, University Cologne, Faculty of Medicine, Kerpener Strasse 62, 50937 Cologne, Germany; Department of Internal Medicine, Lee Health Hospital, Florida State University College of Medicine at Cape Coral, 636 Del Prado Boulevard, Cape Coral, FL 33990, USA.
| | - Hans Nikolaus Caspar Eckel
- Department of Otorhinolaryngology/Head and Neck Surgery, University Hospital of Cologne, University Cologne, Faculty of Medicine, Kerpener Strasse 62, 50973 Cologne, Germany.
| | - Karl Knipper
- Department of General, Visceral and Cancer Surgery, University of Cologne, University Cologne, Faculty of Medicine, Kerpener Strasse 62, 50973 Cologne, Germany.
| | - Anne Maria Schultheis
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg im Breisgau, Germany.
| | - Reinhard Büttner
- Institute of Pathology, University Hospital of Cologne, University Cologne, Faculty of Medicine, Kerpener Strasse 62, 50937 Cologne, Germany.
| | - Alexander Quaas
- Institute of Pathology, University Hospital of Cologne, University Cologne, Faculty of Medicine, Kerpener Strasse 62, 50937 Cologne, Germany.
| | - Jens Peter Klussmann
- Department of Otorhinolaryngology/Head and Neck Surgery, University Hospital of Cologne, University Cologne, Faculty of Medicine, Kerpener Strasse 62, 50973 Cologne, Germany.
| | - Adrian Georg Simon
- Institute of Pathology, University Hospital of Cologne, University Cologne, Faculty of Medicine, Kerpener Strasse 62, 50937 Cologne, Germany.
| |
Collapse
|
2
|
Delint-Ramirez I, Madabhushi R. DNA damage and its links to neuronal aging and degeneration. Neuron 2025; 113:7-28. [PMID: 39788088 PMCID: PMC11832075 DOI: 10.1016/j.neuron.2024.12.001] [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: 09/12/2024] [Revised: 11/07/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025]
Abstract
DNA damage is a major risk factor for the decline of neuronal functions with age and in neurodegenerative diseases. While how DNA damage causes neurodegeneration is still being investigated, innovations over the past decade have provided significant insights into this issue. Breakthroughs in next-generation sequencing methods have begun to reveal the characteristics of neuronal DNA damage hotspots and the causes of DNA damage. Chromosome conformation capture-based approaches have shown that, while DNA damage and the ensuing cellular response alter chromatin topology, chromatin organization at damage sites also affects DNA repair outcomes in neurons. Additionally, neuronal activity results in the formation of programmed DNA breaks, which could burden DNA repair mechanisms and promote neuronal dysfunction. Finally, emerging evidence implicates DNA damage-induced inflammation as an important contributor to the age-related decline in neuronal functions. Together, these discoveries have ushered in a new understanding of the significance of genome maintenance for neuronal function.
Collapse
Affiliation(s)
- Ilse Delint-Ramirez
- Departments of Psychiatry, Neuroscience, and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Peter O' Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ram Madabhushi
- Departments of Psychiatry, Neuroscience, and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Peter O' Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
3
|
Hidmi O, Oster S, Shatleh D, Monin J, Aqeilan RI. Precise Mapping of Physiological DSBs Using In-Suspension Break Labeling In Situ and Sequencing (sBLISS). Methods Mol Biol 2025; 2906:113-136. [PMID: 40082353 DOI: 10.1007/978-1-0716-4426-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
DNA double-strand breaks (DSBs) are a major source of genomic instability. Physiological DSBs are naturally occurring breaks that happen during normal cellular processes. Unlike DNA breaks resulting from DNA damage due to external factors like radiation or chemicals, physiological DSBs play critical roles in various normal biological functions. Some key processes involving physiological DSBs include V(D)J recombination, transcription, and replication. These breaks are typically tightly controlled and are part of the cellular machinery designed to maintain and enhance genomic integrity and diversity. However, if these breaks are misrepaired or left unrepaired, they can contribute to genomic instability, potentially leading to senescence and diseases such as cancer. Here, we outline various methods commonly employed to detect physiological DSBs and introduce a detailed, step-by-step protocol for mapping these breaks using the in-suspension break labeling in situ and sequencing (sBLISS) technique. sBLISS offers single nucleotide resolution and is versatile enough to be applied to any cell type amenable to single-cell suspension. This comprehensive approach not only enhances our understanding of DSBs but also aids in the exploration of their roles in genomic instability.
Collapse
Affiliation(s)
- Osama Hidmi
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sara Oster
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Diala Shatleh
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jonathan Monin
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rami I Aqeilan
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
4
|
Someya Y, Saito S, Takeda S, Adachi N, Kurosawa A. Quercetin exhibits cytotoxicity in cancer cells by inducing two-ended DNA double-strand breaks. Biochem Biophys Res Commun 2024; 739:150977. [PMID: 39549336 DOI: 10.1016/j.bbrc.2024.150977] [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: 08/25/2024] [Revised: 10/09/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
Quercetin, a flavonoid, is involved in the induction of DNA double-strand breaks (DSBs), in addition to its antioxidant properties. Although DNA topoisomerase II (Top2) and reactive oxygen species (ROS) have been suggested as possible mechanisms through which quercetin induces DSBs, the exact mechanism remains unclear. In this study, we examined the mechanism of DSB induction by quercetin and its repair using HeLa cells and gene-knockout cell lines generated from human Nalm-6 cells. Immunofluorescence staining for γH2AX, a DSB marker, and analysis of the frequency of random integration of foreign DNA, which correlates with the number of DSBs and DSB repair pathways, indicated that quercetin induces DSBs in a concentration-dependent manner. The sensitivity assay suggested that the factor involved in quercetin-induced DSBs was not Top2. However, ROS was found to accumulate transiently in quercetin-treated HeLa cells. Furthermore, the addition of ascorbic acid increased the survival of quercetin-treated HeLa cells, suggesting that quercetin induces a transient accumulation of ROS, which in turn induces DSBs. The resulting DSBs were repaired primarily by non-homologous end-joining and homologous recombination, similar to X-ray-induced DSBs. Taken together, quercetin, used as a radiomimetic agent, has the potential to produce effects equivalent to those of an X ray-dose at a relatively low risk.
Collapse
Affiliation(s)
- Yuduki Someya
- Graduate School of Science and Technology, Gunma University, Kiryu, 376-8515, Japan
| | - Shinta Saito
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Shigeki Takeda
- Graduate School of Science and Technology, Gunma University, Kiryu, 376-8515, Japan
| | - Noritaka Adachi
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Aya Kurosawa
- Graduate School of Science and Technology, Gunma University, Kiryu, 376-8515, Japan; Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan; Gunma University Center for Food and Science and Wellness, Gunma University, Kiryu, 376-8515, Japan.
| |
Collapse
|
5
|
Campos Gudiño R, Neudorf NM, Andromidas D, Lichtensztejn Z, McManus KJ. Loss of EMI1 compromises chromosome stability and is associated with cellular transformation in colonic epithelial cell contexts. Br J Cancer 2024; 131:1516-1528. [PMID: 39358461 PMCID: PMC11519589 DOI: 10.1038/s41416-024-02855-9] [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: 07/23/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is still a leading cause of cancer deaths worldwide. Thus, identifying the aberrant genes and proteins underlying disease pathogenesis is critical to improve early detection methods and develop novel therapeutic strategies. Chromosome instability (CIN), or ongoing changes in chromosome complements, is a predominant form of genome instability. It is a driver of genetic heterogeneity found in ~85% of CRCs. Although CIN contributes to CRC pathogenesis, the molecular determinants underlying CIN remain poorly understood. Recently, EMI1, an F-box protein, was identified as a candidate CIN gene. In this study, we sought to determine the impact reduced EMI1 expression has on CIN and cellular transformation. METHODS Coupling siRNA-based silencing and CRISPR/Cas9 knockout clones with quantitative imaging microscopy we evaluated the impact reduced EMI1 expression has on CIN and cellular transformation in four colonic epithelial cell contexts. RESULTS Quantitative imaging microscopy data revealed that reduced EMI1 expression induces increases in CIN phenotypes in both transient (siRNA) and constitutive (CRISPR/Cas9) cell models that are associated with increases in DNA damage and cellular transformation phenotypes in long-term studies. CONCLUSIONS This study determined that reduced EMI1 expression induces CIN and promotes cellular transformation, which is consistent with a role in early CRC development.
Collapse
Affiliation(s)
- Rubi Campos Gudiño
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Nicole M Neudorf
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada
| | - Demi Andromidas
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Zelda Lichtensztejn
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Kirk J McManus
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada.
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
6
|
Gu J, Iyer A, Wesley B, Taglialatela A, Leuzzi G, Hangai S, Decker A, Gu R, Klickstein N, Shuai Y, Jankovic K, Parker-Burns L, Jin Y, Zhang JY, Hong J, Niu X, Costa JA, Pezet MG, Chou J, Chen C'C, Paiva M, Snoeck HW, Landau DA, Azizi E, Chan EM, Ciccia A, Gaublomme JT. Mapping multimodal phenotypes to perturbations in cells and tissue with CRISPRmap. Nat Biotechnol 2024:10.1038/s41587-024-02386-x. [PMID: 39375448 DOI: 10.1038/s41587-024-02386-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 08/12/2024] [Indexed: 10/09/2024]
Abstract
Unlike sequencing-based methods, which require cell lysis, optical pooled genetic screens enable investigation of spatial phenotypes, including cell morphology, protein subcellular localization, cell-cell interactions and tissue organization, in response to targeted CRISPR perturbations. Here we report a multimodal optical pooled CRISPR screening method, which we call CRISPRmap. CRISPRmap combines in situ CRISPR guide-identifying barcode readout with multiplexed immunofluorescence and RNA detection. Barcodes are detected and read out through combinatorial hybridization of DNA oligos, enhancing barcode detection efficiency. CRISPRmap enables in situ barcode readout in cell types and contexts that were elusive to conventional optical pooled screening, including cultured primary cells, embryonic stem cells, induced pluripotent stem cells, derived neurons and in vivo cells in a tissue context. We conducted a screen in a breast cancer cell line of the effects of DNA damage repair gene variants on cellular responses to commonly used cancer therapies, and we show that optical phenotyping pinpoints likely pathogenic patient-derived mutations that were previously classified as variants of unknown clinical significance.
Collapse
Affiliation(s)
- Jiacheng Gu
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Abhishek Iyer
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Ben Wesley
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Angelo Taglialatela
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Giuseppe Leuzzi
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, USA
| | - Sho Hangai
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Aubrianna Decker
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Ruoyu Gu
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Naomi Klickstein
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Yuanlong Shuai
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Kristina Jankovic
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Lucy Parker-Burns
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Yinuo Jin
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jia Yi Zhang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Justin Hong
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Xiang Niu
- Genentech Research and Early Development, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Jonathon A Costa
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mikael G Pezet
- Department of Medicine, Columbia Center for Stem Cell Therapies, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Jacqueline Chou
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Cao 'Claire' Chen
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Margaret Paiva
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Hans-Willem Snoeck
- Department of Medicine, Columbia Center for Stem Cell Therapies, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Dan A Landau
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Elham Azizi
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Computer Science, Columbia University, New York, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Edmond M Chan
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
| | - Alberto Ciccia
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, USA
| | - Jellert T Gaublomme
- Department of Biological Sciences, Columbia University, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- New York Genome Center, New York, NY, USA.
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA.
| |
Collapse
|
7
|
Escobar Marcillo DI, Guglielmi V, Privitera GF, Signore M, Simonelli V, Manganello F, Dell'Orso A, Laterza S, Parlanti E, Pulvirenti A, Marcon F, Siniscalchi E, Fertitta V, Iorio E, Varì R, Nisticò L, Valverde M, Sbraccia P, Dogliotti E, Fortini P. The dual nature of DNA damage response in obesity and bariatric surgery-induced weight loss. Cell Death Dis 2024; 15:664. [PMID: 39256343 PMCID: PMC11387396 DOI: 10.1038/s41419-024-06922-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 09/12/2024]
Abstract
This novel study applies targeted functional proteomics to examine tissues and cells obtained from a cohort of individuals with severe obesity who underwent bariatric surgery (BS), using a Reverse-Phase Protein Array (RPPA). In obese individuals, visceral adipose tissue (VAT), but not subcutaneous adipose tissue (SAT), shows activation of DNA damage response (DDR) markers including ATM, ATR, histone H2AX, KAP1, Chk1, and Chk2, alongside senescence markers p16 and p21. Additionally, stress-responsive metabolic markers, such as survivin, mTOR, and PFKFB3, are specifically elevated in VAT, suggesting both cellular stress and metabolic dysregulation. Conversely, peripheral blood mononuclear cells (PBMCs), while exhibiting elevated mTOR and JNK levels, did not present significant changes in DDR or senescence markers. Following BS, unexpected increases in phosphorylated ATM, ATR, and KAP1 levels, but not in Chk1 and Chk2 nor in senescence markers, were observed. This was accompanied by heightened levels of survivin and mTOR, along with improvement in markers of mitochondrial quality and health. This suggests that, following BS, pro-survival pathways involved in cellular adaptation to various stressors and metabolic alterations are activated in circulating PBMCs. Moreover, our findings demonstrate that the DDR has a dual nature. In the case of VAT from individuals with obesity, chronic DDR proves to be harmful, as it is associated with senescence and chronic inflammation. Conversely, after BS, the activation of DDR proteins in PBMCs is associated with a beneficial survival response. This response is characterized by metabolic redesign and improved mitochondrial biogenesis and functionality. This study reveals physiological changes associated with obesity and BS that may aid theragnostic approaches.
Collapse
Affiliation(s)
| | - Valeria Guglielmi
- Internal Medicine Unit and Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Grete Francesca Privitera
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Michele Signore
- Core Facilities, ISS, Viale Regina Elena 299, 00161, Roma, Italy
| | - Valeria Simonelli
- Dept of Environment and Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy
| | - Federico Manganello
- Dept of Environment and Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy
| | - Ambra Dell'Orso
- Dept of Environment and Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy
| | - Serena Laterza
- Internal Medicine Unit and Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Eleonora Parlanti
- Dept of Environment and Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy
| | - Alfredo Pulvirenti
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Francesca Marcon
- Dept of Environment and Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy
| | - Ester Siniscalchi
- Dept of Environment and Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy
| | - Veronica Fertitta
- Dept of Environment and Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy
| | - Egidio Iorio
- High Resolution NMR Unit-Core Facilities, ISS, Viale Regina Elena, 299, 00161, Roma, Italy
| | - Rosaria Varì
- Center for Gender-Specific Medicine, ISS, Viale Regina Elena 299, 00161, Rome, Italy
| | - Lorenza Nisticò
- Centre for Behavioral Sciences and Mental Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy
| | - Mahara Valverde
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, C.U. C.P, 04510, CDMX, México
| | - Paolo Sbraccia
- Internal Medicine Unit and Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Eugenia Dogliotti
- Dept of Environment and Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy.
| | - Paola Fortini
- Dept of Environment and Health, ISS, Viale Regina Elena 299, 00161, Roma, Italy.
| |
Collapse
|
8
|
Cohen A, Lubenski L, Mouzon A, Kupiec M, Weisman R. TORC2 is required for the accumulation of γH2A in response to DNA damage. J Biol Chem 2024; 300:107531. [PMID: 38971312 PMCID: PMC11321321 DOI: 10.1016/j.jbc.2024.107531] [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: 01/07/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/08/2024] Open
Abstract
TOR protein kinases serve as the catalytic subunit of the TORC1 and TORC2 complexes, which regulate cellular growth, proliferation, and survival. In the fission yeast, Schizosaccharomyces pombe, cells lacking TORC2 or its downstream kinase Gad8 (AKT or SGK1 in human cells) exhibit sensitivity to a wide range of stress conditions, including DNA damage stress. One of the first responses to DNA damage is the phosphorylation of C-terminal serine residues within histone H2AX in human cells (γH2AX), or histone H2A in yeast cells (γH2A). The kinases responsible for γH2A in S. pombe are the two DNA damage checkpoint kinases Rad3 and Tel1 (ATR and ATM, respectively, in human cells). Here we report that TORC2-Gad8 signaling is required for accumulation of γH2A in response to DNA damage and during quiescence. Using the TOR-specific inhibitor, Torin1, we demonstrate that the effect of TORC2 on γH2A in response to DNA damage is immediate, rather than adaptive. The lack of γH2A is restored by deletion mutations of transcription and chromatin modification factors, including loss of components of Paf1C, SAGA, Mediator, and the bromo-domain proteins Bdf1/Bdf2. Thus, we suggest that TORC2-Gad8 may affect the accumulation of γH2A by regulating chromatin structure and function.
Collapse
Affiliation(s)
- Adiel Cohen
- Department of Natural Sciences, The Open University of Israel, Ranana, Israel
| | - Lea Lubenski
- The Shmunis School of Biomedicine & Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Ava Mouzon
- The Shmunis School of Biomedicine & Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Martin Kupiec
- The Shmunis School of Biomedicine & Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Weisman
- Department of Natural Sciences, The Open University of Israel, Ranana, Israel.
| |
Collapse
|
9
|
Hidmi O, Oster S, Shatleh D, Monin J, Aqeilan RI. Protocol for mapping physiological DSBs using in-suspension break labeling in situ and sequencing. STAR Protoc 2024; 5:103059. [PMID: 38717906 PMCID: PMC11098942 DOI: 10.1016/j.xpro.2024.103059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/16/2024] [Accepted: 04/22/2024] [Indexed: 05/20/2024] Open
Abstract
Physiological double-stranded breaks (DSBs) are a major source of genomic instability. Here, we present a protocol for mapping physiological DSBs by in-suspension break labeling in situ and sequencing (sBLISS) in a single-nucleotide resolution. We describe steps for cell fixation, labeling of DSBs, DNA isolation followed by in vitro transcription (IVT), reverse transcription, and library preparation. sBLISS provides a map of DSBs over the genome and can be used to study the role of different factors in DSB formation. For complete details on the use and execution of this protocol, please refer to Hidmi et al.1.
Collapse
Affiliation(s)
- Osama Hidmi
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Sara Oster
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Diala Shatleh
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jonathan Monin
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rami I Aqeilan
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; Cyprus Cancer Research Institute (CCRI), Nicosia, Cyprus.
| |
Collapse
|
10
|
Granzotto A, El Nachef L, Restier-Verlet J, Sonzogni L, Al-Choboq J, Bourguignon M, Foray N. When Chromatin Decondensation Affects Nuclear γH2AX Foci Pattern and Kinetics and Biases the Assessment of DNA Double-Strand Breaks by Immunofluorescence. Biomolecules 2024; 14:703. [PMID: 38927105 PMCID: PMC11201768 DOI: 10.3390/biom14060703] [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: 05/26/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Immunofluorescence with antibodies against phosphorylated forms of H2AX (γH2AX) is revolutionizing our understanding of repair and signaling of DNA double-strand breaks (DSBs). Unfortunately, the pattern of γH2AX foci depends upon a number of parameters (nature of stress, number of foci, radiation dose, repair time, cell cycle phase, gene mutations, etc…) whose one of the common points is chromatin condensation/decondensation. Here, we endeavored to demonstrate how chromatin conformation affects γH2AX foci pattern and influences immunofluorescence signal. DSBs induced in non-transformed human fibroblasts were analyzed by γH2AX immunofluorescence with sodium butyrate treatment of chromatin applied after the irradiation that decondenses chromatin but does not induce DNA breaks. Our data showed that the pattern of γH2AX foci may drastically change with the experimental protocols in terms of size and brightness. Notably, some γH2AX minifoci resulting from the dispersion of the main signal due to chromatin decondensation may bias the quantification of the number of DSBs. We proposed a model called "Christmas light models" to tentatively explain this diversity of γH2AX foci pattern that may also be considered for any DNA damage marker that relocalizes as nuclear foci.
Collapse
Affiliation(s)
- Adeline Granzotto
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (A.G.); (L.E.N.); (J.R.-V.); (L.S.); (J.A.-C.); (M.B.)
| | - Laura El Nachef
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (A.G.); (L.E.N.); (J.R.-V.); (L.S.); (J.A.-C.); (M.B.)
| | - Juliette Restier-Verlet
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (A.G.); (L.E.N.); (J.R.-V.); (L.S.); (J.A.-C.); (M.B.)
| | - Laurène Sonzogni
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (A.G.); (L.E.N.); (J.R.-V.); (L.S.); (J.A.-C.); (M.B.)
| | - Joëlle Al-Choboq
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (A.G.); (L.E.N.); (J.R.-V.); (L.S.); (J.A.-C.); (M.B.)
| | - Michel Bourguignon
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (A.G.); (L.E.N.); (J.R.-V.); (L.S.); (J.A.-C.); (M.B.)
- Department of Biophysics and Nuclear Medicine, University Paris Saclay (UVSQ), 78035 Versailles, France
| | - Nicolas Foray
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (A.G.); (L.E.N.); (J.R.-V.); (L.S.); (J.A.-C.); (M.B.)
| |
Collapse
|
11
|
Contreras L, García-Gaipo L, Casar B, Gandarillas A. DNA damage signalling histone H2AX is required for tumour growth. Cell Death Discov 2024; 10:99. [PMID: 38402225 PMCID: PMC10894207 DOI: 10.1038/s41420-024-01869-9] [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: 08/14/2023] [Revised: 02/06/2024] [Accepted: 02/14/2024] [Indexed: 02/26/2024] Open
Abstract
Cancer most frequently develops in self-renewal tissues that are the target of genetic alterations due to mutagens or intrinsic DNA replication errors. Histone γH2AX has a critical role in the cellular DNA repair pathway cascade and contributes to genomic stability. However, the role of γH2AX in the ontology of cancer is unclear. We have investigated this issue in the epidermis, a self-renewal epithelium continuously exposed to genetic hazard and replication stress. Silencing H2AX caused cell cycle hyperactivation, impaired DNA repair and epidermal hyperplasia in the skin. However, mutagen-induced carcinogenesis was strikingly reduced in the absence of H2AX. KO tumours appeared significantly later than controls and were fewer, smaller and more benign. The stem cell marker Δp63 drastically diminished in the KO epidermis. We conclude that H2AX is required for tissue-making during both homoeostasis and tumourigenesis, possibly by contributing to the control and repair of stem cells. Therefore, although H2AX is thought to act as a tumour suppressor and our results show that it contributes to homeostasis, they also indicate that it is required for the development of cancer.
Collapse
Affiliation(s)
- Lizbeth Contreras
- Cell cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Lorena García-Gaipo
- Cell cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Berta Casar
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Cantabria (UC), 39011, Santander, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Alberto Gandarillas
- Cell cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain.
- Institut National de la Santé et de la Recherche Médicale, (INSERM), Délégation Occitanie, 34394, Montpellier, France.
| |
Collapse
|
12
|
Gu J, Iyer A, Wesley B, Taglialatela A, Leuzzi G, Hangai S, Decker A, Gu R, Klickstein N, Shuai Y, Jankovic K, Parker-Burns L, Jin Y, Zhang JY, Hong J, Niu S, Chou J, Landau DA, Azizi E, Chan EM, Ciccia A, Gaublomme JT. CRISPRmap: Sequencing-free optical pooled screens mapping multi-omic phenotypes in cells and tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.26.572587. [PMID: 38234835 PMCID: PMC10793456 DOI: 10.1101/2023.12.26.572587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Pooled genetic screens are powerful tools to study gene function in a high-throughput manner. Typically, sequencing-based screens require cell lysis, which limits the examination of critical phenotypes such as cell morphology, protein subcellular localization, and cell-cell/tissue interactions. In contrast, emerging optical pooled screening methods enable the investigation of these spatial phenotypes in response to targeted CRISPR perturbations. In this study, we report a multi-omic optical pooled CRISPR screening method, which we have named CRISPRmap. Our method combines a novel in situ CRISPR guide identifying barcode readout approach with concurrent multiplexed immunofluorescence and in situ RNA detection. CRISPRmap barcodes are detected and read out through combinatorial hybridization of DNA oligos, enhancing barcode detection efficiency, while reducing both dependency on third party proprietary sequencing reagents and assay cost. Notably, we conducted a multi-omic base-editing screen in a breast cancer cell line on core DNA damage repair genes involved in the homologous recombination and Fanconi anemia pathways investigating how nucleotide variants in those genes influence DNA damage signaling and cell cycle regulation following treatment with ionizing radiation or DNA damaging agents commonly used for cancer therapy. Approximately a million cells were profiled with our multi-omic approach, providing a comprehensive phenotypic assessment of the functional consequences of the studied variants. CRISPRmap enabled us to pinpoint likely-pathogenic patient-derived mutations that were previously classified as variants of unknown clinical significance. Furthermore, our approach effectively distinguished barcodes of a pooled library in tumor tissue, and we coupled it with cell-type and molecular phenotyping by cyclic immunofluorescence. Multi-omic spatial analysis of how CRISPR-perturbed cells respond to various environmental cues in the tissue context offers the potential to significantly expand our understanding of tissue biology in both health and disease.
Collapse
Affiliation(s)
- Jiacheng Gu
- Department of Biological Sciences, Columbia University, NY, USA
| | - Abhishek Iyer
- Department of Biological Sciences, Columbia University, NY, USA
| | - Ben Wesley
- Department of Biological Sciences, Columbia University, NY, USA
| | - Angelo Taglialatela
- Department of Genetics and Development, Columbia University Irving Medical Center, NY, USA
| | - Giuseppe Leuzzi
- Department of Genetics and Development, Columbia University Irving Medical Center, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, NY, USA
- Institute for Cancer Genetics, Columbia University Irving Medical Center, NY, USA
| | - Sho Hangai
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, NY, USA
| | | | - Ruoyu Gu
- Department of Biological Sciences, Columbia University, NY, USA
| | | | - Yuanlong Shuai
- Department of Biological Sciences, Columbia University, NY, USA
| | - Kristina Jankovic
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, NY, USA
| | - Lucy Parker-Burns
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, NY, USA
| | - Yinuo Jin
- Department of Biomedical Engineering, Columbia University, NY, USA
| | - Jia Yi Zhang
- Department of Biomedical Engineering, Columbia University, NY, USA
| | - Justin Hong
- Department of Computer Science, Columbia University, NY, USA
| | - Steve Niu
- Weill Cornell Medicine, NY, USA
- Genentech Research and Early Development, CA, USA
| | - Jacqueline Chou
- Department of Biological Sciences, Columbia University, NY, USA
- Weill Cornell Medicine, NY, USA
| | - Dan A. Landau
- Weill Cornell Medicine, NY, USA
- New York Genome Center, NY, USA
| | - Elham Azizi
- Department of Biomedical Engineering, Columbia University, NY, USA
- Department of Computer Science, Columbia University, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, NY, USA
| | - Edmond M. Chan
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, NY, USA
- New York Genome Center, NY, USA
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, NY, USA
| | - Alberto Ciccia
- Department of Genetics and Development, Columbia University Irving Medical Center, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, NY, USA
- Institute for Cancer Genetics, Columbia University Irving Medical Center, NY, USA
| | - Jellert T. Gaublomme
- Department of Biological Sciences, Columbia University, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, NY, USA
- New York Genome Center, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, NY, USA
| |
Collapse
|
13
|
Xu K, Qiao JY, Zhao BW, Dong MZ, Lei WL, Li YY, Ju Z, Schatten H, Wang ZB, Liu K, Sun QY. Maternal SMC2 is essential for embryonic development via participating chromosome condensation in mice. J Cell Physiol 2023; 238:2535-2545. [PMID: 37642322 DOI: 10.1002/jcp.31102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/16/2023] [Accepted: 08/07/2023] [Indexed: 08/31/2023]
Abstract
During the oocyte growth, maturation and zygote development, chromatin structure keeps changing to regulate different nuclear activities. Here, we reported the role of SMC2, a core component of condensin complex, in oocyte and embryo development. Oocyte-specific conditional knockout of SMC2 caused female infertility. In the absence of SMC2, oocyte meiotic maturation and ovulation occurred normally, but chromosome condensation showed defects and DNA damages were accumulated in oocytes. The pronuclei were abnormally organized and micronuclei were frequently observed in fertilized eggs, their activity was impaired, and embryo development was arrested at the one-cell stage, suggesting that maternal SMC2 is essential for embryonic development.
Collapse
Affiliation(s)
- Ke Xu
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Guangdong, China
- Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jing-Yi Qiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Bin-Wang Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Ming-Zhe Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wen-Long Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yuan-Yuan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Kui Liu
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Guangdong, China
- Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qing-Yuan Sun
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
14
|
Li F, Peng X, Zheng L, Liu Y, Liu Q, Zhang B, Shi Y, Wu H, Xu C. YAP nuclear translocation facilitates radiation resistance in nasopharyngeal carcinoma cells. Biochem Biophys Res Commun 2023; 670:109-116. [PMID: 37290285 DOI: 10.1016/j.bbrc.2023.05.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023]
Abstract
OBJECTIVES Investigate the role of the Hippo-YAP signaling pathway in radioresistant Nasopharyngeal Carcinoma (NPC). METHODS Establishment of radioresistant CNE-1 cells (CNE-1-RR) by gradually increasing ionizing radiation (IR) doses, and identifying the apoptosis of CNE-1-RR by flow cytometry. We employed immunoblot and immunofluorescence staining to detect the expression of YAP in both CNE-1-RR and control group cells. Moreover, we validated the role of YAP in CNE-1-RR by inhibiting its nuclear translocation. RESULTS In contrast to the control group, radioresistant NPC cells demonstrated significant YAP dephosphorylation and nuclear translocation. CNE-1-RR cells exhibited enhanced activation of γ-H2AX (Ser139) upon exposure to IR and greater recruitment of double-strand breaks (DSBs) repair-related proteins. Additionally, inhibiting YAP nuclear translocation in radioresistant CNE-1-RR cells significantly increased their sensitivity to radiotherapy. CONCLUSIONS The present investigation has unveiled the intricate mechanisms and physiological roles of YAP in CNE-1-RR cells exhibiting resistance to IR. Based on our findings, it can be inferred that a combinational therapeutic strategy involving radiotherapy and inhibitors that impede the nuclear translocation of YAP holds promising potential for treating radioresistant NPC.
Collapse
Affiliation(s)
- Feifei Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China; Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Xinhao Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610047, China
| | - Linlin Zheng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610047, China
| | - Yiqiang Liu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610047, China
| | - Qianshi Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China; Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Biqin Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610047, China
| | - Ying Shi
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610047, China
| | - Hong Wu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610047, China.
| | - Chuan Xu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China; Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610047, China.
| |
Collapse
|
15
|
Xing X, Liang Y, Li Y, Zhao Y, Zhang Y, Li Z, Li Z, Wu Z. Fisetin Delays Postovulatory Oocyte Aging by Regulating Oxidative Stress and Mitochondrial Function through Sirt1 Pathway. Molecules 2023; 28:5533. [PMID: 37513404 PMCID: PMC10384696 DOI: 10.3390/molecules28145533] [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: 06/13/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The quality of oocytes determines the development potential of an embryo and is dependent on their timely fertilization after ovulation. Postovulatory oocyte aging is an inevitable factor during some assisted reproduction technology procedures, which results in poor fertilization rates and impairs embryo development. We found that fisetin, a bioactive flavonol contained in fruits and vegetables, delayed postovulatory oocyte aging in mice. Fisetin improved the development of aged oocytes after fertilization and inhibited the Sirt1 reduction in aged oocytes. Fisetin increased the GSH level and Sod2 transcription level to inhibit ROS accumulation in aged oocytes. Meanwhile, fisetin attenuated aging-induced spindle abnormalities, mitochondrial dysfunction, and apoptosis. At the molecular level, fisetin decreased aging-induced aberrant expression of H3K9me3. In addition, fisetin increased the expression levels of the mitochondrial transcription factor Tfam and the mitochondrial genes Co2 and Atp8 by upregulating Sirt1 in aged oocytes. Finally, inhibition of Sirt1 reversed the anti-aging effects of fisetin. Taken together, fisetin delayed postovulatory oocyte aging by upregulating Sirt1.
Collapse
Affiliation(s)
- Xupeng Xing
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yalin Liang
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yanan Li
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yaolu Zhao
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yuxing Zhang
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zheng Li
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
16
|
Mauro S, Bolognesi MM, Villa N, Capitoli G, Furia L, Mascadri F, Zucchini N, Totis M, Faretta M, Galimberti S, Bovo G, Cattoretti G. A DNA damage response-like phenotype defines a third of colon cancers at onset. FASEB J 2023; 37:e23020. [PMID: 37342943 DOI: 10.1096/fj.202300132r] [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: 01/25/2023] [Revised: 04/12/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023]
Abstract
Colon adenocarcinoma (COAD) has a limited range of diversified, personalized therapeutic opportunities, besides DNA hypermutating cases; thus, both new targets or broadening existing strategies for personalized intervention are of interest. Routinely processed material from 246 untreated COADs with clinical follow-up was probed for evidence of DNA damage response (DDR), that is, the gathering of DDR-associated molecules at discrete nuclear spots, by multiplex immunofluorescence and immunohistochemical staining for DDR complex proteins (γH2AX, pCHK2, and pNBS1). We also tested the cases for type I interferon response, T-lymphocyte infiltration (TILs), and mutation mismatch repair defects (MMRd), known to be associated with defects of DNA repair. FISH analysis for chromosome 20q copy number variations was obtained. A total of 33.7% of COAD display a coordinated DDR on quiescent, non-senescent, non-apoptotic glands, irrespective of TP53 status, chromosome 20q abnormalities, and type I IFN response. Clinicopathological parameters did not differentiate DDR+ cases from the other cases. TILs were equally present in DDR and non-DDR cases. DDR+ MMRd cases were preferentially retaining wild-type MLH1. The outcome after 5FU-based chemotherapy was not different in the two groups. DDR+ COAD represents a subgroup not aligned with known diagnostic, prognostic, or therapeutic categories, with potential new targeted treatment opportunities, exploiting the DNA damage repair pathways.
Collapse
Affiliation(s)
- Stefania Mauro
- Pathology, Vimercate Hospital, ASST-Brianza, Vimercate, Italy
| | - Maddalena M Bolognesi
- Pathology, Department of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
| | - Nicoletta Villa
- Genetics, Fondazione IRCCS San Gerardo dei Tintori Monza, Monza, Italy
| | - Giulia Capitoli
- Bicocca Bioinformatics Biostatistics and Bioimaging B4 Center, School of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
| | - Laura Furia
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Francesco Mascadri
- Pathology, Department of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
| | - Nicola Zucchini
- Pathology, Fondazione IRCCS San Gerardo dei Tintori Monza, Monza, Italy
| | - Mauro Totis
- GI Surgery, Fondazione IRCCS San Gerardo dei Tintori Monza, Monza, Italy
| | - Mario Faretta
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Stefania Galimberti
- Bicocca Bioinformatics Biostatistics and Bioimaging B4 Center, School of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
| | - Giorgio Bovo
- Pathology, Vimercate Hospital, ASST-Brianza, Vimercate, Italy
| | - Giorgio Cattoretti
- Pathology, Department of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
- Pathology, Fondazione IRCCS San Gerardo dei Tintori Monza, Monza, Italy
| |
Collapse
|
17
|
Factors to Consider for the Correct Use of γH2AX in the Evaluation of DNA Double-Strand Breaks Damage Caused by Ionizing Radiation. Cancers (Basel) 2022; 14:cancers14246204. [PMID: 36551689 PMCID: PMC9776434 DOI: 10.3390/cancers14246204] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
People exposed to ionizing radiation (IR) both for diagnostic and therapeutic purposes is constantly increasing. Since the use of IR involves a risk of harmful effects, such as the DNA DSB induction, an accurate determination of this induced DNA damage and a correct evaluation of the risk-benefit ratio in the clinical field are of key relevance. γH2AX (the phosphorylated form of the histone variant H2AX) is a very early marker of DSBs that can be induced both in physiological conditions, such as in the absence of specific external agents, and by external factors such as smoking, heat, background environmental radiation, and drugs. All these internal and external conditions result in a basal level of γH2AX which must be considered for the correct assessment of the DSBs after IR exposure. In this review we analyze the most common conditions that induce H2AX phosphorylation, including specific exogenous stimuli, cellular states, basic environmental factors, and lifestyles. Moreover, we discuss the most widely used methods for γH2AX determination and describe the principal applications of γH2AX scoring, paying particular attention to clinical studies. This knowledge will help us optimize the use of available methods in order to discern the specific γH2AX following IR-induced DSBs from the basal level of γH2AX in the cells.
Collapse
|
18
|
Characterizing and exploiting the many roles of aberrant H2B monoubiquitination in cancer pathogenesis. Semin Cancer Biol 2022; 86:782-798. [PMID: 34953650 DOI: 10.1016/j.semcancer.2021.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 12/08/2021] [Accepted: 12/19/2021] [Indexed: 01/27/2023]
Abstract
Monoubiquitination of histone H2B on lysine 120 (H2Bub1) is implicated in the control of multiple essential processes, including transcription, DNA damage repair and mitotic chromosome segregation. Accordingly, aberrant regulation of H2Bub1 can induce transcriptional reprogramming and genome instability that may promote oncogenesis. Remarkably, alterations of the ubiquitin ligases and deubiquitinating enzymes regulating H2Bub1 are emerging as ubiquitous features in cancer, further supporting the possibility that the misregulation of H2Bub1 is an underlying mechanism contributing to cancer pathogenesis. To date, aberrant H2Bub1 dynamics have been reported in multiple cancer types and are associated with transcriptional changes that promote oncogenesis in a cancer type-specific manner. Owing to the multi-functional nature of H2Bub1, misregulation of its writers and erasers may drive disease initiation and progression through additional synergistic processes. Accordingly, understanding the molecular determinants and pathogenic impacts associated with aberrant H2Bub1 regulation may reveal novel drug targets and therapeutic vulnerabilities that can be exploited to develop innovative precision medicine strategies that better combat cancer. In this review, we present the normal functions of H2Bub1 in the control of DNA-associated processes and describe the pathogenic implications associated with its misregulation in cancer. We further discuss the challenges coupled with the development of therapeutic strategies targeting H2Bub1 misregulation and expose the potential benefits of designing treatments that synergistically exploit the multiple functionalities of H2Bub1 to improve treatment selectivity and efficacy.
Collapse
|
19
|
Porcine Enteric Coronavirus PEDV Induces the ROS-ATM and Caspase7-CAD-γH2AX Signaling Pathways to Foster Its Replication. Viruses 2022; 14:v14081782. [PMID: 36016404 PMCID: PMC9413700 DOI: 10.3390/v14081782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/12/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
DNA damage response (DDR) is an evolutionarily conserved mechanism by which eukaryotic cells sense DNA lesions caused by intrinsic and extrinsic stimuli, including virus infection. Although interactions between DNA viruses and DDR have been extensively studied, how RNA viruses, especially coronaviruses, regulate DDR remains unknown. A previous study showed that the porcine epidemic diarrhea virus (PEDV), a member of the genus Alphacoronavirus in the Coronaviridae family, induces DDR in infected cells. However, the underlying mechanism was unclear. This study showed that PEDV activates the ATM-Chk2 signaling, while inhibition of ATM or Chk2 dampens the early stage of PEDV infection. Additionally, we found that PEDV-activated ATM signaling correlates with intracellular ROS production. Interestingly, we showed that, unlike the typical γH2AX foci, PEDV infection leads to a unique γH2AX staining pattern, including phase I (nuclear ring staining), II (pan-nuclear staining), and III (co-staining with apoptotic bodies), which highly resembles the apoptosis process. Furthermore, we demonstrated that PEDV-induced H2AX phosphorylation depends on the activation of caspase-7 and caspase-activated DNAse (CAD), but not ATM-Chk2. Finally, we showed that the knockdown of H2AX attenuates PEDV replication. Taken together, we conclude that PEDV induces DDR through the ROS-ATM and caspase7-CAD-γH2AX signaling pathways to foster its early replication.
Collapse
|
20
|
Dong MZ, Ouyang YC, Gao SC, Ma XS, Hou Y, Schatten H, Wang ZB, Sun QY. PPP4C facilitates homologous recombination DNA repair by dephosphorylating PLK1 during early embryo development. Development 2022; 149:dev200351. [PMID: 35546066 DOI: 10.1242/dev.200351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/24/2022] [Indexed: 12/17/2023]
Abstract
Mammalian early embryo cells have complex DNA repair mechanisms to maintain genomic integrity, and homologous recombination (HR) plays the main role in response to double-strand DNA breaks (DSBs) in these cells. Polo-like kinase 1 (PLK1) participates in the HR process and its overexpression has been shown to occur in a variety of human cancers. Nevertheless, the regulatory mechanism of PLK1 remains poorly understood, especially during the S and G2 phase. Here, we show that protein phosphatase 4 catalytic subunit (PPP4C) deletion causes severe female subfertility due to accumulation of DNA damage in oocytes and early embryos. PPP4C dephosphorylated PLK1 at the S137 site, negatively regulating its activity in the DSB response in early embryonic cells. Depletion of PPP4C induced sustained activity of PLK1 when cells exhibited DNA lesions that inhibited CHK2 and upregulated the activation of CDK1, resulting in inefficient loading of the essential HR factor RAD51. On the other hand, when inhibiting PLK1 in the S phase, DNA end resection was restricted. These results demonstrate that PPP4C orchestrates the switch between high-PLK1 and low-PLK1 periods, which couple the checkpoint to HR.
Collapse
Affiliation(s)
- Ming-Zhe Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shi-Cai Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xue-Shan Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| |
Collapse
|
21
|
Herchenröther A, Wunderlich TM, Lan J, Hake SB. Spotlight on histone H2A variants: From B to X to Z. Semin Cell Dev Biol 2022; 135:3-12. [PMID: 35365397 DOI: 10.1016/j.semcdb.2022.03.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 12/30/2022]
Abstract
Chromatin, the functional organization of DNA with histone proteins in eukaryotic nuclei, is the tightly-regulated template for several biological processes, such as transcription, replication, DNA damage repair, chromosome stability and sister chromatid segregation. In order to achieve a reversible control of local chromatin structure and DNA accessibility, various interconnected mechanisms have evolved. One of such processes includes the deposition of functionally-diverse variants of histone proteins into nucleosomes, the building blocks of chromatin. Among core histones, the family of H2A histone variants exhibits the largest number of members and highest sequence-divergence. In this short review, we report and discuss recent discoveries concerning the biological functions of the animal histone variants H2A.B, H2A.X and H2A.Z and how dysregulation or mutation of the latter impacts the development of disease.
Collapse
Affiliation(s)
| | - Tim M Wunderlich
- Institute for Genetics, Justus Liebig University, 35390 Giessen, Germany
| | - Jie Lan
- Institute for Genetics, Justus Liebig University, 35390 Giessen, Germany.
| | - Sandra B Hake
- Institute for Genetics, Justus Liebig University, 35390 Giessen, Germany.
| |
Collapse
|
22
|
Merighi A, Gionchiglia N, Granato A, Lossi L. The Phosphorylated Form of the Histone H2AX (γH2AX) in the Brain from Embryonic Life to Old Age. Molecules 2021; 26:7198. [PMID: 34885784 PMCID: PMC8659122 DOI: 10.3390/molecules26237198] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
The γ phosphorylated form of the histone H2AX (γH2AX) was described more than 40 years ago and it was demonstrated that phosphorylation of H2AX was one of the first cellular responses to DNA damage. Since then, γH2AX has been implicated in diverse cellular functions in normal and pathological cells. In the first part of this review, we will briefly describe the intervention of H2AX in the DNA damage response (DDR) and its role in some pivotal cellular events, such as regulation of cell cycle checkpoints, genomic instability, cell growth, mitosis, embryogenesis, and apoptosis. Then, in the main part of this contribution, we will discuss the involvement of γH2AX in the normal and pathological central nervous system, with particular attention to the differences in the DDR between immature and mature neurons, and to the significance of H2AX phosphorylation in neurogenesis and neuronal cell death. The emerging picture is that H2AX is a pleiotropic molecule with an array of yet not fully understood functions in the brain, from embryonic life to old age.
Collapse
Affiliation(s)
| | | | | | - Laura Lossi
- Department of Veterinary Sciences, University of Turin, I-10095 Grugliasco, Italy; (A.M.); (N.G.); (A.G.)
| |
Collapse
|
23
|
Aki T, Uemura K. Cell Death and Survival Pathways Involving ATM Protein Kinase. Genes (Basel) 2021; 12:1581. [PMID: 34680975 PMCID: PMC8535589 DOI: 10.3390/genes12101581] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
Cell death is the ultimate form of cellular dysfunction, and is induced by a wide range of stresses including genotoxic stresses. During genotoxic stress, two opposite cellular reactions, cellular protection through DNA repair and elimination of damaged cells by the induction of cell death, can occur in both separate and simultaneous manners. ATM (ataxia telangiectasia mutated) kinase (hereafter referred to as ATM) is a protein kinase that plays central roles in the induction of cell death during genotoxic stresses. It has long been considered that ATM mediates DNA damage-induced cell death through inducing apoptosis. However, recent research progress in cell death modality is now revealing ATM-dependent cell death pathways that consist of not only apoptosis but also necroptosis, ferroptosis, and dysfunction of autophagy, a cellular survival mechanism. In this short review, we intend to provide a brief outline of cell death mechanisms in which ATM is involved, with emphasis on pathways other than apoptosis.
Collapse
Affiliation(s)
- Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan;
| | | |
Collapse
|
24
|
Dicks N, Gutierrez K, Currin L, de Macedo MP, Glanzner WG, Mondadori RG, Michalak M, Agellon LB, Bordignon V. Tauroursodeoxycholic acid/TGR5 signaling promotes survival and early development of glucose-stressed porcine embryos†. Biol Reprod 2021; 105:76-86. [PMID: 33889948 PMCID: PMC8256098 DOI: 10.1093/biolre/ioab072] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/24/2021] [Accepted: 04/12/2021] [Indexed: 01/21/2023] Open
Abstract
Conditions of impaired energy and nutrient homeostasis, such as diabetes and obesity, are associated with infertility. Hyperglycemia increases endoplasmic reticulum stress as well as oxidative stress and reduces embryo development and quality. Oxidative stress also causes deoxyribonucleic acid damage, which impairs embryo quality and development. The natural bile acid tauroursodeoxycholic acid reduces endoplasmic reticulum stress and rescues developmentally incompetent late-cleaving embryos, as well as embryos subjected to nuclear stress, suggesting the endoplasmic reticulum stress response, or unfolded protein response, and the genome damage response are linked. Tauroursodeoxycholic acid acts via the Takeda-G-protein-receptor-5 to alleviate nuclear stress in embryos. To evaluate the role of tauroursodeoxycholic acid/Takeda-G-protein-receptor-5 signaling in embryo unfolded protein response, we used a model of glucose-induced endoplasmic reticulum stress. Embryo development was impaired by direct injection of tauroursodeoxycholic acid into parthenogenetically activated oocytes, whereas it was improved when tauroursodeoxycholic acid was added to the culture medium. Attenuation of the Takeda-G-protein-receptor-5 precluded the positive effect of tauroursodeoxycholic acid supplementation on development of parthenogenetically activated and fertilized embryos cultured under standard conditions and parthenogenetically activated embryos cultured with excess glucose. Moreover, attenuation of tauroursodeoxycholic acid/Takeda-G-protein-receptor-5 signaling induced endoplasmic reticulum stress, oxidative stress and cell survival genes, but decreased expression of pluripotency genes in parthenogenetically activated embryos cultured under excess glucose conditions. These data suggest that Takeda-G-protein-receptor-5 signaling pathways link the unfolded protein response and genome damage response. Furthermore, this study identifies Takeda-G-protein-receptor-5 signaling as a potential target for mitigating fertility issues caused by nutrient excess-associated blastomere stress and embryo death.
Collapse
Affiliation(s)
- Naomi Dicks
- Department of Animal Science, McGill University, Quebec, Canada
| | | | - Luke Currin
- Department of Animal Science, McGill University, Quebec, Canada
| | | | | | - Rafael G Mondadori
- Department of Animal Science, McGill University, Quebec, Canada
- ReproPel, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Quebec, Canada
| | | |
Collapse
|
25
|
Tatin X, Muggiolu G, Sauvaigo S, Breton J. Evaluation of DNA double-strand break repair capacity in human cells: Critical overview of current functional methods. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2021; 788:108388. [PMID: 34893153 DOI: 10.1016/j.mrrev.2021.108388] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 02/05/2023]
Abstract
DNA double-strand breaks (DSBs) are highly deleterious lesions, responsible for mutagenesis, chromosomal translocation or cell death. DSB repair (DSBR) is therefore a critical part of the DNA damage response (DDR) to restore molecular and genomic integrity. In humans, this process is achieved through different pathways with various outcomes. The balance between DSB repair activities varies depending on cell types, tissues or individuals. Over the years, several methods have been developed to study variations in DSBR capacity. Here, we mainly focus on functional techniques, which provide dynamic information regarding global DSB repair proficiency or the activity of specific pathways. These methods rely on two kinds of approaches. Indirect techniques, such as pulse field gel electrophoresis (PFGE), the comet assay and immunofluorescence (IF), measure DSB repair capacity by quantifying the time-dependent decrease in DSB levels after exposure to a DNA-damaging agent. On the other hand, cell-free assays and reporter-based methods directly track the repair of an artificial DNA substrate. Each approach has intrinsic advantages and limitations and despite considerable efforts, there is currently no ideal method to quantify DSBR capacity. All techniques provide different information and can be regarded as complementary, but some studies report conflicting results. Parameters such as the type of biological material, the required equipment or the cost of analysis may also limit available options. Improving currently available methods measuring DSBR capacity would be a major step forward and we present direct applications in mechanistic studies, drug development, human biomonitoring and personalized medicine, where DSBR analysis may improve the identification of patients eligible for chemo- and radiotherapy.
Collapse
Affiliation(s)
- Xavier Tatin
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, 38000 Grenoble, France; LXRepair, 5 Avenue du Grand Sablon, 38700 La Tronche, France
| | | | - Sylvie Sauvaigo
- LXRepair, 5 Avenue du Grand Sablon, 38700 La Tronche, France
| | - Jean Breton
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, 38000 Grenoble, France.
| |
Collapse
|
26
|
Jeusset LM, Guppy BJ, Lichtensztejn Z, McDonald D, McManus KJ. Reduced USP22 Expression Impairs Mitotic Removal of H2B Monoubiquitination, Alters Chromatin Compaction and Induces Chromosome Instability That May Promote Oncogenesis. Cancers (Basel) 2021; 13:cancers13051043. [PMID: 33801331 PMCID: PMC7958346 DOI: 10.3390/cancers13051043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
Chromosome instability (CIN) is an enabling feature of oncogenesis associated with poor patient outcomes, whose genetic determinants remain largely unknown. As mitotic chromatin compaction defects can compromise the accuracy of chromosome segregation into daughter cells and drive CIN, characterizing the molecular mechanisms ensuring accurate chromatin compaction may identify novel CIN genes. In vitro, histone H2B monoubiquitination at lysine 120 (H2Bub1) impairs chromatin compaction, while in vivo H2Bub1 is rapidly depleted from chromatin upon entry into mitosis, suggesting that H2Bub1 removal may be a pre-requisite for mitotic fidelity. The deubiquitinating enzyme USP22 catalyzes H2Bub1 removal in interphase and may also be required for H2Bub1 removal in early mitosis to maintain chromosome stability. In this study, we demonstrate that siRNA-mediated USP22 depletion increases H2Bub1 levels in early mitosis and induces CIN phenotypes associated with mitotic chromatin compaction defects revealed by super-resolution microscopy. Moreover, USP22-knockout models exhibit continuously changing chromosome complements over time. These data identify mitotic removal of H2Bub1 as a critical determinant of chromatin compaction and faithful chromosome segregation. We further demonstrate that USP22 is a CIN gene, indicating that USP22 deletions, which are frequent in many tumor types, may drive genetic heterogeneity and contribute to cancer pathogenesis.
Collapse
Affiliation(s)
- Lucile M. Jeusset
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, MB R3E0V9, Canada; (L.M.J.); (B.J.G.); (Z.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E0J9, Canada
| | - Brent J. Guppy
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, MB R3E0V9, Canada; (L.M.J.); (B.J.G.); (Z.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E0J9, Canada
| | - Zelda Lichtensztejn
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, MB R3E0V9, Canada; (L.M.J.); (B.J.G.); (Z.L.)
| | - Darin McDonald
- Department of Oncology, University of Alberta, Edmonton, AB T6G2H7, Canada;
| | - Kirk J. McManus
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, MB R3E0V9, Canada; (L.M.J.); (B.J.G.); (Z.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E0J9, Canada
- Correspondence: ; Tel.: +1-(204)-787-2833
| |
Collapse
|
27
|
Orlando L, Tanasijevic B, Nakanishi M, Reid JC, García-Rodríguez JL, Chauhan KD, Porras DP, Aslostovar L, Lu JD, Shapovalova Z, Mitchell RR, Boyd AL, Bhatia M. Phosphorylation state of the histone variant H2A.X controls human stem and progenitor cell fate decisions. Cell Rep 2021; 34:108818. [PMID: 33691101 DOI: 10.1016/j.celrep.2021.108818] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/28/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Histone variants (HVs) are a subfamily of epigenetic regulators implicated in embryonic development, but their role in human stem cell fate remains unclear. Here, we reveal that the phosphorylation state of the HV H2A.X (γH2A.X) regulates self-renewal and differentiation of human pluripotent stem cells (hPSCs) and leukemic progenitors. As demonstrated by CRISPR-Cas deletion, H2A.X is essential in maintaining normal hPSC behavior. However, reduced levels of γH2A.X enhances hPSC differentiation toward the hematopoietic lineage with concomitant inhibition of neural development. In contrast, activation and sustained levels of phosphorylated H2A.X enhance hPSC neural fate while suppressing hematopoiesis. This controlled lineage bias correlates to occupancy of γH2A.X at genomic loci associated with ectoderm versus mesoderm specification. Finally, drug modulation of H2A.X phosphorylation overcomes differentiation block of patient-derived leukemic progenitors. Our study demonstrates HVs may serve to regulate pluripotent cell fate and that this biology could be extended to somatic cancer stem cell control.
Collapse
Affiliation(s)
- Luca Orlando
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Borko Tanasijevic
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Mio Nakanishi
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Jennifer C Reid
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Juan L García-Rodríguez
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Kapil Dev Chauhan
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Deanna P Porras
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Lili Aslostovar
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Justin D Lu
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Zoya Shapovalova
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Ryan R Mitchell
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Allison L Boyd
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Mickie Bhatia
- McMaster University, Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
28
|
Zhang JQJ, Saravanabavan S, Chandra AN, Munt A, Wong ATY, Harris PC, Harris DCH, McKenzie P, Wang Y, Rangan GK. Up-Regulation of DNA Damage Response Signaling in Autosomal Dominant Polycystic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:902-920. [PMID: 33549515 DOI: 10.1016/j.ajpath.2021.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 12/19/2022]
Abstract
DNA damage and alterations in DNA damage response (DDR) signaling could be one of the molecular mechanisms mediating focal kidney cyst formation in autosomal dominant polycystic kidney disease (ADPKD). The aim of this study was to test the hypothesis that markers of DNA damage and DDR signaling are increased in human and experimental ADPKD. In the human ADPKD transcriptome, the number of up-regulated DDR-related genes was increased by 16.6-fold compared with that in normal kidney, and by 2.5-fold in cystic compared with that in minimally cystic tissue (P < 0.0001). In end-stage human ADPKD tissue, γ-H2A histone family member X (H2AX), phosphorylated ataxia telangiectasia and radiation-sensitive mutant 3 (Rad3)-related (pATR), and phosphorylated ataxia telangiectasia mutated (pATM) localized to cystic kidney epithelial cells. In vitro, pATR and pATM were also constitutively increased in human ADPKD tubular cells (WT 9-7 and 9-12) compared with control (HK-2). In addition, extrinsic oxidative DNA damage by hydrogen peroxide augmented γ-H2AX and cell survival in human ADPKD cells, and exacerbated cyst growth in the three-dimensional Madin-Darby canine kidney cyst model. In contrast, DDR-related gene expression was only transiently increased on postnatal day 0 in Pkd1RC/RC mice, and not altered at later time points up to 12 months of age. In conclusion, DDR signaling is dysregulated in human ADPKD and during the early phases of murine ADPKD. The constitutive expression of the DDR pathway in ADPKD may promote survival of PKD1-mutated cells and contribute to kidney cyst growth.
Collapse
Affiliation(s)
- Jennifer Q J Zhang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Sayanthooran Saravanabavan
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Ashley N Chandra
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Alexandra Munt
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Annette T Y Wong
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Peter C Harris
- Mayo Translational Polycystic Kidney Disease Center, Mayo Clinic, Rochester, Minnesota
| | - David C H Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Paul McKenzie
- Department of Tissue Pathology, NSW Health Pathology, Royal Prince Alfred Hospital, The University of Sydney, Sydney, New South Wales, Australia
| | - Yiping Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Gopala K Rangan
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia.
| |
Collapse
|
29
|
Biernacka A, Skrzypczak M, Zhu Y, Pasero P, Rowicka M, Ginalski K. High-resolution, ultrasensitive and quantitative DNA double-strand break labeling in eukaryotic cells using i-BLESS. Nat Protoc 2021; 16:1034-1061. [PMID: 33349705 PMCID: PMC8088906 DOI: 10.1038/s41596-020-00448-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 10/09/2020] [Indexed: 11/09/2022]
Abstract
DNA double-strand breaks (DSBs) are implicated in various physiological processes, such as class-switch recombination or crossing-over during meiosis, but also present a threat to genome stability. Extensive evidence shows that DSBs are a primary source of chromosome translocations or deletions, making them a major cause of genomic instability, a driving force of many diseases of civilization, such as cancer. Therefore, there is a great need for a precise, sensitive, and universal method for DSB detection, to enable both the study of their mechanisms of formation and repair as well as to explore their therapeutic potential. We provide a detailed protocol for our recently developed ultrasensitive and genome-wide DSB detection method: immobilized direct in situ breaks labeling, enrichment on streptavidin and next-generation sequencing (i-BLESS), which relies on the encapsulation of cells in agarose beads and labeling breaks directly and specifically with biotinylated linkers. i-BLESS labels DSBs with single-nucleotide resolution, allows detection of ultrarare breaks, takes 5 d to complete, and can be applied to samples from any organism, as long as a sufficient amount of starting material can be obtained. We also describe how to combine i-BLESS with our qDSB-Seq approach to enable the measurement of absolute DSB frequencies per cell and their precise genomic coordinates at the same time. Such normalization using qDSB-Seq is especially useful for the evaluation of spontaneous DSB levels and the estimation of DNA damage induced rather uniformly in the genome (e.g., by irradiation or radiomimetic chemotherapeutics).
Collapse
Affiliation(s)
- Anna Biernacka
- Laboratory of Bioinformatics and Systems Biology, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Magdalena Skrzypczak
- Laboratory of Bioinformatics and Systems Biology, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Yingjie Zhu
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Philippe Pasero
- Institut de Génétique Humaine, CNRS et Université de Montpellier, Montpellier, France
| | - Maga Rowicka
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Institute for Translational Sciences, University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Krzysztof Ginalski
- Laboratory of Bioinformatics and Systems Biology, Centre of New Technologies, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
30
|
Molinuevo R, Freije A, Contreras L, Sanz JR, Gandarillas A. The DNA damage response links human squamous proliferation with differentiation. J Cell Biol 2020; 219:152154. [PMID: 33007086 PMCID: PMC7534927 DOI: 10.1083/jcb.202001063] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/08/2020] [Accepted: 08/14/2020] [Indexed: 12/26/2022] Open
Abstract
How rapid cell multiplication leads to cell differentiation in developing tissues is still enigmatic. This question is central to morphogenesis, cell number control, and homeostasis. Self-renewal epidermoid epithelia are continuously exposed to mutagens and are the most common target of cancer. Unknown mechanisms commit rapidly proliferating cells to post-mitotic terminal differentiation. We have over-activated or inhibited the endogenous DNA damage response (DDR) pathways by combinations of activating TopBP1 protein, specific shRNAs, or chemical inhibitors for ATR, ATM, and/or DNA-PK. The results dissect and demonstrate that these signals control keratinocyte differentiation in proliferating cells independently of actual DNA damage. The DDR limits keratinocyte multiplication upon hyperproliferative stimuli. Moreover, knocking down H2AX, a common target of the DDR pathways, inhibits the epidermoid phenotype. The results altogether show that the DDR is required to maintain the balance proliferation differentiation and suggest that is part of the squamous program. We propose a homeostatic model where genetic damage is automatically and continuously cleansed by cell-autonomous mechanisms.
Collapse
Affiliation(s)
- Rut Molinuevo
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla, Santander, Spain
| | - Ana Freije
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla, Santander, Spain
| | - Lizbeth Contreras
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla, Santander, Spain
| | - Juan R Sanz
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla, Santander, Spain.,Plastic Surgery Service, Hospital Universitario Marqués de Valdecilla, Santander, Spain.,Plastic Surgery Department, Universidad de Cantabria, Santander, Spain
| | - Alberto Gandarillas
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla, Santander, Spain.,Institut National de la Santé et de la Recherche Médicale, Languedoc-Roussillon, Montpellier, France
| |
Collapse
|
31
|
Ethyl-2-amino-pyrrole-3-carboxylates are active against imatinib-resistant gastrointestinal stromal tumors in vitro and in vivo. Anticancer Drugs 2020; 30:475-484. [PMID: 30986804 DOI: 10.1097/cad.0000000000000753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We showed recently that ethyl-2-amino-pyrrole-3-carboxylates (EAPCs) exhibit potent antiproliferative activities against a broad spectrum of soft tissue sarcoma and gastrointestinal stromal tumor (GIST) cell lines in vitro. The molecular mechanism of action was owing to inhibition of tubulin polymerization and induction of a robust G2/M cell-cycle arrest, leading to the accumulation of tumor cells in the M-phase and induction of apoptosis. Given that more than 50% of the patients with GISTs develop resistance to imatinib (IM) over the 2 years of IM-based therapy, we examined whether EAPCs exhibit activity against IM-resistant GISTs in vitro and in vivo. A real-time antiproliferation assay illustrated the potent antiproliferative activities of EAPCs against IM-sensitive and IM-resistant GISTs. This was in agreement with the colony formation assay, which revealed potent antiproliferative activities of EAPCs against IM-resistant GISTs, being much stronger when compared with IM and doxorubicin, a topoisomerase II inhibitor. Next, we tested the efficacy of EAPCs in the xenograft model of GISTs, exhibiting secondary IM resistance owing to RTK switch (loss of c-KIT/gain of FGFR2α). A total of 30 5- to 8-week-old female nu/nu mice were subcutaneously inoculated into the flank areas with IM-resistant GIST-T1-R cells (100 μl of 1×10 GIST T-1R cells/ml suspension, in Dulbecco's PBS). Mice were randomized as control (untreated), IM (50 mg/kg), EAPC-20 (10 mg/kg) or EAPC-24 (10 mg/kg) and were treated orally for 10 days. IM has a minor inhibitory effect on tumor size, thus revealing GIST resistance to IM. In contrast, both of EAPCs effectively reduced the tumor size. This was associated with an increased intratumoral apoptosis as detected by immunohistochemical staining for cleaved caspase-3 on day 5 of the treatment. Furthermore, both EAPCs significantly reduced the proliferative activity of tumor cells in the central zones of tumors as measured by positivity for Ki-67 staining. More importantly, in EAPC-24-treated GISTs, the histological response was mainly characterized by the induction of necrosis, whereas EAPC-20 induced the signs of intratumoral fibrosis and myxoid degeneration. Collectively, our data suggest that EAPC-20 and EAPC-24 are the perspective antitumor agents that exhibit antiproliferative and cytotoxic activity against GISTs exhibiting secondary resistance to IM.
Collapse
|
32
|
Osrodek M, Rozanski M, Czyz M. Insulin Reduces the Efficacy of Vemurafenib and Trametinib in Melanoma Cells. Cancer Manag Res 2020; 12:7231-7250. [PMID: 32982400 PMCID: PMC7501594 DOI: 10.2147/cmar.s263767] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Despite the progress made in the clinical management of metastatic melanoma, a patient's response to treatment cannot be fully predicted, and intrinsic or acquired resistance that is developed in most melanoma patients warrants further research efforts. In addition to genetic factors, microenvironmental input should be considered to explain the diversity of response to treatment among melanoma patients. In this study, we evaluated the impact of insulin on patient-derived BRAFV600E melanoma cells, either untreated or treated with vemurafenib or trametinib, inhibitors of BRAFV600 and MEK1/2, respectively. METHODS Cells were cultured in serum-free conditions, either with or without insulin. The activity of the MAPK/ERK and PI3K/AKT pathways was assessed by Western blotting, cell viability, and percentages of Ki-67- and NGFR-positive cells by flow cytometry. Transcript levels were analyzed using qRT-PCR, and γ-H2AX levels by immunoblotting and confocal microscopy. A luminescence-based assay was used to measure glutathione content. RESULTS While insulin did not influence the MAPK/ERK pathway activity, it had a strong influence on melanoma cells, in which this pathway was suppressed by either vemurafenib or trametinib. In the presence of insulin, both drugs were much less efficient in 1) inhibiting proliferation and reducing the percentage of Ki-67-positive cells, and 2) inducing apoptosis and phosphorylation of histone H2AX in melanoma cells. Changes induced by vemurafenib and trametinib in glutathione homeostasis and DNA repair gene expression were also attenuated by insulin. Moreover, insulin impaired the combined effects of targeted drugs and doxorubicin in melanoma cells. In addition to insulin-induced PI3K/AKT activity, which was either transient or sustainable depending on the cell line, an insulin-triggered increase in the percentage of cells expressing NGFR, a marker of neural crest stem-like cells, may contribute to the reduced drug efficacy. CONCLUSION Our results demonstrate the role of insulin in reducing the efficacy of vemurafenib and trametinib. This needs clinical assessment.
Collapse
Affiliation(s)
- Marta Osrodek
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
| | - Michal Rozanski
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
33
|
Liddle P, Jara-Wilde J, Lafon-Hughes L, Castro I, Härtel S, Folle G. dSTORM microscopy evidences in HeLa cells clustered and scattered γH2AX nanofoci sensitive to ATM, DNA-PK, and ATR kinase inhibitors. Mol Cell Biochem 2020; 473:77-91. [PMID: 32638256 DOI: 10.1007/s11010-020-03809-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/18/2020] [Indexed: 12/13/2022]
Abstract
In response to DNA double-strand breaks (DSB), histone H2AX is phosphorylated around the lesion by a feed forward signal amplification loop, originating γH2AX foci detectable by immunofluorescence and confocal microscopy as elliptical areas of uniform intensity. We exploited the significant increase in resolution (~ × 10) provided by single-molecule localization microscopy (SMLM) to investigate at nanometer scale the distribution of γH2AX signals either endogenous (controls) or induced by the radiomimetic bleomycin (BLEO) in HeLa cells. In both conditions, clustered substructures (nanofoci) confined to γH2AX foci and scattered nanofoci throughout the remnant nuclear area were detected. SR-Tesseler software (Voronoï tessellation-based segmentation) was combined with a custom Python script to first separate clustered nanofoci inside γH2AX foci from scattered nanofoci, and then to perform a cluster analysis upon each nanofoci type. Compared to controls, γH2AX foci in BLEO-treated nuclei presented on average larger areas (0.41 versus 0.19 µm2), more nanofoci per focus (22.7 versus 13.2) and comparable nanofoci densities (~ 60 nanofoci/µm2). Scattered γH2AX nanofoci were equally present (~ 3 nanofoci/µm2), suggesting an endogenous origin. BLEO-treated cells were challenged with specific inhibitors of canonical H2AX kinases, namely: KU-55933, VE-821 and NU-7026 for ATM, ATR and DNA-PK, respectively. Under treatment with pooled inhibitors, clustered nanofoci vanished from super-resolution images while scattered nanofoci decreased (~ 50%) in density. Residual scattered nanofoci could reflect, among other alternatives, H2AX phosphorylation mediated by VRK1, a recently described non-canonical H2AX kinase. In addition to H2AX findings, an analytical approach to quantify clusters of highly differing density from SMLM data is put forward.
Collapse
Affiliation(s)
- Pablo Liddle
- Departamento de Genética, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay.
| | - Jorge Jara-Wilde
- SCIAN-Lab, Biomedical Neuroscience Institute (BNI), Santiago, Chile.,Departamento de Ciencias de la Computación, Universidad de Chile, Santiago, Chile
| | - Laura Lafon-Hughes
- Departamento de Genética, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Iván Castro
- SCIAN-Lab, Biomedical Neuroscience Institute (BNI), Santiago, Chile
| | - Steffen Härtel
- SCIAN-Lab, Biomedical Neuroscience Institute (BNI), Santiago, Chile.,Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gustavo Folle
- Departamento de Genética, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
34
|
Gerashchenko BI, Salmina K, Krigerts J, Erenpreisa J, Babsky AM. INDUCED POLYPLOIDY AND SORTING OF DAMAGED DNA BY MICRONUCLEATION IN RADIORESISTANT RAT LIVER EPITHELIAL STEM-LIKE CELLS EXPOSED TO X-RAYS. PROBLEMY RADIAT︠S︡IĬNOÏ MEDYT︠S︡YNY TA RADIOBIOLOHIÏ 2020; 24:220-234. [PMID: 31841469 DOI: 10.33145/2304-8336-2019-24-220-234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Rat liver stem-like epithelial cells (WB-F344) that under certain conditions may differentiate into hepa- tocyte and biliary lineages were subjected to acute X-irradiation with the aim to examine cell cycle peculiarities dur- ing the course of survival. MATERIALS AND METHODS Suspensions of WB-F344 cells that grew as a monolayer and reached sub-confluence were irradiated with 1, 5, and 10 Gy of X-rays (2 Gy/min). As an intact control, sham-irradiated cells were used. After irra- diation, cells were plated into 25-cm2 tissue culture flasks to culture them for over several days without reaching contact inhibition. On days 1, 2, 3, and 5 post-irradiation, cells were harvested and examined for nuclear morpholo- gy and DNA ploidy by stoichiometric toluidine blue reaction and image cytometry. On days 7 and 9 post-irradiation, only heavily irradiated (10 Gy) cells were examined. Also, 10 Gy-irradiated cells were chosen for immunofluorescence staining to monitor persistence of DNA lesions (γ-H2AX), cell proliferation (Ki-67), and self-renewal factors charac- teristic for stem cells (OCT4 and NANOG). RESULTS Radioresistance of WB-F344 cells was evidenced by the findings that they do not undergo rapid and mas- sive cell death that in fact was weakly manifested as apoptotic even in heavily irradiated cells. Instead, there was cell cycle progression delay accompanied by polyploidization (via Ki-67-positive mitotic slippage or via impaired cytokinesis) and micronucleation in a dose-dependent manner, although micronucleation to some extent went ahead of polyploidization. Polyploid cells amenable for recovering from DNA damage can mitotically depolyploidize. Many micronuclei contained γ-H2AX clusters, suggesting isolation of severely damaged DNA fragments. Both factors, OCT4 and NANOG, were expressed in the intact control, but became enhanced after irradiation. CONCLUSIONS Although the fact of micronucleation is indicative of genotoxic effect, WB-F344 cells can probably escape cell death via sorting of damaged DNA by micronuclei. Induction of polyploidy in these cells can be adaptive to promote cell survival and tissue regeneration with possible involvement of self-renewal mechanism.
Collapse
Affiliation(s)
- B I Gerashchenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, 45 Vasylkivska St., Kyiv, 03022, Ukraine
| | - K Salmina
- Latvian Biomedical Research and Study Centre, 1 Ratsupites St., Riga, LV-1067, Latvia
| | - J Krigerts
- Latvian Biomedical Research and Study Centre, 1 Ratsupites St., Riga, LV-1067, Latvia
| | - J Erenpreisa
- Latvian Biomedical Research and Study Centre, 1 Ratsupites St., Riga, LV-1067, Latvia
| | - A M Babsky
- Ivan Franko National University of Lviv, Faculty of Biology, 4 Mykhaila Hrushevskoho St., Lviv, 79005, Ukraine
| |
Collapse
|
35
|
Schmitz ML, Higgins JMG, Seibert M. Priming chromatin for segregation: functional roles of mitotic histone modifications. Cell Cycle 2020; 19:625-641. [PMID: 31992120 DOI: 10.1080/15384101.2020.1719585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Posttranslational modifications (PTMs) of histone proteins are important for various cellular processes including regulation of gene expression and chromatin structure, DNA damage response and chromosome segregation. Here we comprehensively review mitotic histone PTMs, in particular phosphorylations, and discuss their interplay and functions in the control of dynamic protein-protein interactions as well as their contribution to centromere and chromosome structure and function during cell division. Histone phosphorylations can create binding sites for mitotic regulators such as the chromosomal passenger complex, which is required for correction of erroneous spindle attachments and chromosome bi-orientation. Other histone PTMs can alter the structural properties of nucleosomes and the accessibility of chromatin. Epigenetic marks such as lysine methylations are maintained during mitosis and may also be important for mitotic transcription as well as bookmarking of transcriptional states to ensure the transmission of gene expression programs through cell division. Additionally, histone phosphorylation can dissociate readers of methylated histones without losing epigenetic information. Through all of these processes, mitotic histone PTMs play a functional role in priming the chromatin for faithful chromosome segregation and preventing genetic instability, one of the characteristic hallmarks of cancer cells.
Collapse
Affiliation(s)
- M Lienhard Schmitz
- Institute of Biochemistry, Medical Faculty, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Jonathan M G Higgins
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Markus Seibert
- Institute of Biochemistry, Medical Faculty, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
36
|
Dicks N, Gutierrez K, Currin L, Priotto de Macedo M, Glanzner W, Michalak M, Agellon LB, Bordignon V. Tauroursodeoxycholic acid acts via TGR5 receptor to facilitate DNA damage repair and improve early porcine embryo development. Mol Reprod Dev 2019; 87:161-173. [PMID: 31793725 DOI: 10.1002/mrd.23305] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/22/2019] [Indexed: 01/23/2023]
Abstract
DNA damage associated with assisted reproductive technologies is an important factor affecting gamete fertility and embryo development. Activation of the TGR5 receptor by tauroursodeoxycholic acid (TUDCA) has been shown to reduce endoplasmic reticulum (ER) stress in embryos; however, its effect on genome damage responses (GDR) activation to facilitate DNA damage repair has not been examined. This study aimed to investigate the effect of TUDCA on DNA damage repair and embryo development. In a porcine model of ultraviolet light (UV)-induced nuclear stress, TUDCA reduced DNA damage and ER stress in developing embryos, as measured by γH2AX and glucose-regulated protein 78 immunofluorescence, respectively. TUDCA was equally able to rescue early embryo development. No difference in total cell number, DNA damage, or percentage of apoptotic cells, measured by cleaved caspase 3 immunofluorescence, was noted in embryos that reached the blastocyst stage. Interestingly, Dicer-substrate short interfering RNA-mediated disruption of TGR5 signaling abrogated the beneficial effects of TUDCA on UV-treated embryos. Quantitative PCR analysis revealed activation of the GDR, through increased messenger RNA abundance of DNAPK, 53BP1, and DNA ligase IV, as well as the ER stress response, through increased spliced XBP1 and X-linked inhibitor of apoptosis. Results from this study demonstrated that TUDCA activates TGR5-mediated signaling to reduce DNA damage and improve embryo development after UV exposure.
Collapse
Affiliation(s)
- Naomi Dicks
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Karina Gutierrez
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Luke Currin
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | | | - Werner Glanzner
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| |
Collapse
|
37
|
Transcriptional Modulation by Idelalisib Synergizes with Bendamustine in Chronic Lymphocytic Leukemia. Cancers (Basel) 2019; 11:cancers11101519. [PMID: 31601046 PMCID: PMC6826782 DOI: 10.3390/cancers11101519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/19/2019] [Accepted: 10/02/2019] [Indexed: 11/17/2022] Open
Abstract
The phosphatidyl-inositol 3 kinase (PI3K) δ inhibitor, idelalisib (IDE), is a potent inhibitor of the B-cell receptor pathway and a novel and highly effective agent for the treatment of chronic lymphocytic leukemia (CLL). We evaluated the activities of IDE in comparison to bendamusine (BEN), a commonly used alkylating agent, in primary CLL cells ex vivo. In contrast to BEN, IDE was cytotoxic to cells from extensively-treated patients, including those with a deletion (del)17p. Cross-resistance was not observed between BEN and IDE, confirming their different modes of cytotoxicity. Marked synergy was seen between BEN and IDE, even in cases that were resistant to BEN or IDE individually, and those with deletion (del) 17p. CD40L/interleukin 4 (IL4) co-treatment mimicking the CLL microenvironment increased resistance to IDE, but synergy was retained. PI3Kδ-deficient murine splenic B cells were more resistant to IDE and showed reduced synergy with BEN, thus confirming the importance of functional PI3Kδ protein. Although IDE was observed to induce γH2AX, IDE did not enhance activation of the DNA damage response nor DNA repair activity. Interestingly, IDE decreased global RNA synthesis and was antagonistic with 5,6-Dichlorobenzimidazole 1-b-D-ribofuranoside (DRB), an inhibitor of transcription. These findings add to the increasingly complex cellular effects of IDE, and B cell receptor (BCR) inhibitors in general, in CLL.
Collapse
|
38
|
Pich D, Mrozek-Gorska P, Bouvet M, Sugimoto A, Akidil E, Grundhoff A, Hamperl S, Ling PD, Hammerschmidt W. First Days in the Life of Naive Human B Lymphocytes Infected with Epstein-Barr Virus. mBio 2019; 10:e01723-19. [PMID: 31530670 PMCID: PMC6751056 DOI: 10.1128/mbio.01723-19] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 08/16/2019] [Indexed: 12/14/2022] Open
Abstract
Epstein-Barr virus (EBV) infects and activates resting human B lymphocytes, reprograms them, induces their proliferation, and establishes a latent infection in them. In established EBV-infected cell lines, many viral latent genes are expressed. Their roles in supporting the continuous proliferation of EBV-infected B cells in vitro are known, but their functions in the early, prelatent phase of infection have not been investigated systematically. In studies during the first 8 days of infection using derivatives of EBV with mutations in single genes of EBVs, we found only Epstein-Barr nuclear antigen 2 (EBNA2) to be essential for activating naive human B lymphocytes, inducing their growth in cell volume, driving them into rapid cell divisions, and preventing cell death in a subset of infected cells. EBNA-LP, latent membrane protein 2A (LMP2A), and the viral microRNAs have supportive, auxiliary functions, but mutants of LMP1, EBNA3A, EBNA3C, and the noncoding Epstein-Barr virus with small RNA (EBERs) had no discernible phenotype compared with wild-type EBV. B cells infected with a double mutant of EBNA3A and 3C had an unexpected proliferative advantage and did not regulate the DNA damage response (DDR) of the infected host cell in the prelatent phase. Even EBNA1, which has very critical long-term functions in maintaining and replicating the viral genomic DNA in established cell lines, was dispensable for the early activation of infected cells. Our findings document that the virus dose is a decisive parameter and indicate that EBNA2 governs the infected cells initially and implements a strictly controlled temporal program independent of other viral latent genes. It thus appears that EBNA2 is sufficient to control all requirements for clonal cellular expansion and to reprogram human B lymphocytes from energetically quiescent to activated cells.IMPORTANCE The preferred target of Epstein-Barr virus (EBV) is human resting B lymphocytes. We found that their infection induces a well-coordinated, time-driven program that starts with a substantial increase in cell volume, followed by cellular DNA synthesis after 3 days and subsequent rapid rounds of cell divisions on the next day accompanied by some DNA replication stress (DRS). Two to 3 days later, the cells decelerate and turn into stably proliferating lymphoblast cell lines. With the aid of 16 different recombinant EBV strains, we investigated the individual contributions of EBV's multiple latent genes during early B-cell infection and found that many do not exert a detectable phenotype or contribute little to EBV's prelatent phase. The exception is EBNA2 that is essential in governing all aspects of B-cell reprogramming. EBV relies on EBNA2 to turn the infected B lymphocytes into proliferating lymphoblasts preparing the infected host cell for the ensuing stable, latent phase of viral infection. In the early steps of B-cell reprogramming, viral latent genes other than EBNA2 are dispensable, but some, EBNA-LP, for example, support the viral program and presumably stabilize the infected cells once viral latency is established.
Collapse
Affiliation(s)
- Dagmar Pich
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health and German Center for Infection Research (DZIF), Munich, Germany
| | - Paulina Mrozek-Gorska
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health and German Center for Infection Research (DZIF), Munich, Germany
| | - Mickaël Bouvet
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health and German Center for Infection Research (DZIF), Munich, Germany
| | - Atsuko Sugimoto
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health and German Center for Infection Research (DZIF), Munich, Germany
| | - Ezgi Akidil
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health and German Center for Infection Research (DZIF), Munich, Germany
| | - Adam Grundhoff
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Stephan Hamperl
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Paul D Ling
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Wolfgang Hammerschmidt
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health and German Center for Infection Research (DZIF), Munich, Germany
| |
Collapse
|
39
|
Shanbhag NM, Evans MD, Mao W, Nana AL, Seeley WW, Adame A, Rissman RA, Masliah E, Mucke L. Early neuronal accumulation of DNA double strand breaks in Alzheimer's disease. Acta Neuropathol Commun 2019; 7:77. [PMID: 31101070 PMCID: PMC6524256 DOI: 10.1186/s40478-019-0723-5] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 02/06/2023] Open
Abstract
The maintenance of genomic integrity is essential for normal cellular functions. However, it is difficult to maintain over a lifetime in postmitotic cells such as neurons, in which DNA damage increases with age and is exacerbated by multiple neurological disorders, including Alzheimer's disease (AD). Here we used immunohistochemical staining to detect DNA double strand breaks (DSBs), the most severe form of DNA damage, in postmortem brain tissues from patients with mild cognitive impairment (MCI) or AD and from cognitively unimpaired controls. Immunostaining for γH2AX-a post-translational histone modification that is widely used as a marker of DSBs-revealed increased proportions of γH2AX-labeled neurons and astrocytes in the hippocampus and frontal cortex of MCI and AD patients, as compared to age-matched controls. In contrast to the focal pattern associated with DSBs, some neurons and glia in humans and mice showed diffuse pan-nuclear patterns of γH2AX immunoreactivity. In mouse brains and primary neuronal cultures, such pan-nuclear γH2AX labeling could be elicited by increasing neuronal activity. To assess whether pan-nuclear γH2AX represents DSBs, we used a recently developed technology, DNA damage in situ ligation followed by proximity ligation assay, to detect close associations between γH2AX sites and free DSB ends. This assay revealed no evidence of DSBs in neurons or astrocytes with prominent pan-nuclear γH2AX labeling. These findings suggest that focal, but not pan-nuclear, increases in γH2AX immunoreactivity are associated with DSBs in brain tissue and that these distinct patterns of γH2AX formation may have different causes and consequences. We conclude that AD is associated with an accumulation of DSBs in vulnerable neuronal and glial cell populations from early stages onward. Because of the severe adverse effects this type of DNA damage can have on gene expression, chromatin stability and cellular functions, DSBs could be an important causal driver of neurodegeneration and cognitive decline in this disease.
Collapse
Affiliation(s)
- Niraj M Shanbhag
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Mark D Evans
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - Wenjie Mao
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - Alissa L Nana
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Anthony Adame
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, 92093, USA
- Present address: Division of Neuroscience, National Institute on Aging, Bethesda, MD, 20892, USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
40
|
Ebmeyer J, Braeuning A, Glatt H, These A, Hessel-Pras S, Lampen A. Human CYP3A4-mediated toxification of the pyrrolizidine alkaloid lasiocarpine. Food Chem Toxicol 2019; 130:79-88. [PMID: 31103741 DOI: 10.1016/j.fct.2019.05.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 01/05/2023]
Abstract
Pyrrolizidine alkaloids (PA) are widely distributed phytotoxins contaminating food and feed. Hepatic enzymes are considered to bioactivate PA. Previous studies showed differences in the metabolism rate in liver homogenates of different species. Thus, uncertainty remains with respect to the relevance of human metabolism. Our study aimed to analyze whether the PA representative lasiocarpine is toxified by human cytochrome P450 (CYP) enzymes. We compared the metabolic elimination of lasiocarpine in the presence of rat and human S9 fractions and liver microsomes. Experiments with the potent CYP3A/Cyp3a inhibitor ketoconazole and supersomes containing individual human and rat CYPs revealed that enzymes of the CYP3A/Cyp3a family of both species are of major relevance for lasiocarpine metabolism. To assess if metabolism by human CYP3A4 results in a toxification of lasiocarpine we performed experiments with V79 cells. γH2AX and micronucleus formation were analyzed as endpoints for genotoxicity. No effects were observed in the wildtype cells, which lack CYP activity. By contrast, a V79 clone engineered for expression of human CYP3A4 showed concentration-dependent γH2AX and micronucleus formation. Concluding, our results showed the CYP3A4-dependent formation of genotoxic metabolites of lasiocarpine. The results confirm previous data indicating the need to include metabolism of PA for human risk assessment.
Collapse
Affiliation(s)
- Johanna Ebmeyer
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Hansruedi Glatt
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Anja These
- German Federal Institute for Risk Assessment, Department Safety in the Food Chain, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Stefanie Hessel-Pras
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| | - Alfonso Lampen
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| |
Collapse
|
41
|
Luo Y, Wu J, Zou J, Cao Y, He Y, Ling H, Zeng T. BCL10 in cell survival after DNA damage. Clin Chim Acta 2019; 495:301-308. [PMID: 31047877 DOI: 10.1016/j.cca.2019.04.077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 01/01/2023]
Abstract
The complex defense mechanism of the DNA damage response (DDR) developed by cells during long-term evolution is an important mechanism for maintaining the stability of the genome. Defects in the DDR pathway can lead to the occurrence of various diseases, including tumor development. Most cancer treatments cause DNA damage and apoptosis. However, cancer cells have the natural ability to repair this damage and inhibit apoptosis, ultimately leading to the development of drug resistance. Therefore, investigating the mechanism of DNA damage may contribute markedly to the future treatment of cancer. The CARMA-BCL10-MALT1 (CBM) complex formed by B cell lymphoma/leukemia 10 (BCL10) regulates apoptosis by activating NF-κB signaling. BCL10 is involved in the formation of complexes that antagonize apoptosis and contribute to cell survival after DNA damage, with cytoplasmic BCL10 entering the nucleus to promote DNA damage repair, including histone ubiquitination and the recruitment of homologous recombination (HR) repair factors. This article reviews the role of BCL10 in cell survival following DNA damage.
Collapse
Affiliation(s)
- Yichen Luo
- Key Laboratory of Tumor Cellular & Molecular Pathology, College of Hunan Province, Cancer Research Institute, University of South China,Hengyang, Hunan 421001, China; Hunan Provincial Education Department document (Approval number: 2014-405], Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Jing Wu
- Key Laboratory of Tumor Cellular & Molecular Pathology, College of Hunan Province, Cancer Research Institute, University of South China,Hengyang, Hunan 421001, China; Hunan Provincial Education Department document (Approval number: 2014-405], Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Juan Zou
- Key Laboratory of Tumor Cellular & Molecular Pathology, College of Hunan Province, Cancer Research Institute, University of South China,Hengyang, Hunan 421001, China; Hunan Provincial Education Department document (Approval number: 2014-405], Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Yijing Cao
- Key Laboratory of Tumor Cellular & Molecular Pathology, College of Hunan Province, Cancer Research Institute, University of South China,Hengyang, Hunan 421001, China; Hunan Provincial Education Department document (Approval number: 2014-405], Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Yan He
- Key Laboratory of Tumor Cellular & Molecular Pathology, College of Hunan Province, Cancer Research Institute, University of South China,Hengyang, Hunan 421001, China; Department of Pathology, Longgang Central Hospital, Shenzhen, Guangdong 518000, China
| | - Hui Ling
- Key Laboratory of Tumor Cellular & Molecular Pathology, College of Hunan Province, Cancer Research Institute, University of South China,Hengyang, Hunan 421001, China; Hunan Provincial Education Department document (Approval number: 2014-405], Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| | - Tiebing Zeng
- Hunan Provincial Education Department document (Approval number: 2014-405], Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China; Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
42
|
Yu F, Ye K, Hu Y, Li J, An Y, Qu D. Exposure to polycyclic aromatic hydrocarbons derived from vehicle exhaust gas induces premature senescence in mouse lung fibroblast cells. Mol Med Rep 2019; 19:4326-4334. [PMID: 30942392 PMCID: PMC6471379 DOI: 10.3892/mmr.2019.10086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 02/19/2019] [Indexed: 11/06/2022] Open
Abstract
Long‑term exposure to vehicle exhaust gas may lead to various age‑associated disorders, including cardiovascular disease and cancer. Polycyclic aromatic hydrocarbons (PAHs) belong to an important class of carcinogens, which are released into the environment by vehicles and are detectable at high levels in Chinese urban areas. However, whether vehicle exhaust gas (EG), and in particular the PAHs derived from EG, are able to induce cell senescence remains unclear. In the present study, vehicle EG and pure PAHs were used as pollution sources to investigate the effects of long‑term exposure to PAH on the cellular processes occurring in mouse lung fibroblast cells (mLFCs). Using cell proliferation and apoptosis assays, it was demonstrated that benzopyrene (BaP) suppressed the proliferation of mLFCs, and benzanthracene (BaA) and BaP induced cell apoptosis. Molecular analysis suggested that long‑term exposure to BaA and BaP was able to increase the protein expression levels of p53, p21 and the apoptotic factors involved in the caspase cascade, including caspase‑3 and ‑9. Notably, the present study suggested that PAH exposure was able to promote cell senescence in mLFCs by activating the ATM serine/threonine kinase/H2A histone family member X pathway. The present study may provide novel insights into the underlying mechanism of vehicle EG and PAHs in promoting the development of age‑associated diseases.
Collapse
Affiliation(s)
- Feng Yu
- State Key Laboratory of Automotive Simulation and Control, Jilin University, Changchun, Jilin 130011, P.R. China
| | - Ke Ye
- State Key Laboratory of Automotive Simulation and Control, Jilin University, Changchun, Jilin 130011, P.R. China
| | - Yunfeng Hu
- State Key Laboratory of Automotive Simulation and Control, Jilin University, Changchun, Jilin 130011, P.R. China
| | - Jincheng Li
- Department of Gastrointestinal Colorectal Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yonglei An
- Key Laboratory of Groundwater Resources and Environment, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dawei Qu
- State Key Laboratory of Automotive Simulation and Control, Jilin University, Changchun, Jilin 130011, P.R. China
| |
Collapse
|
43
|
Dworak N, Makosa D, Chatterjee M, Jividen K, Yang CS, Snow C, Simke WC, Johnson IG, Kelley JB, Paschal BM. A nuclear lamina-chromatin-Ran GTPase axis modulates nuclear import and DNA damage signaling. Aging Cell 2019; 18:e12851. [PMID: 30565836 PMCID: PMC6351833 DOI: 10.1111/acel.12851] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 08/16/2018] [Accepted: 09/09/2018] [Indexed: 12/25/2022] Open
Abstract
The Ran GTPase regulates nuclear import and export by controlling the assembly state of transport complexes. This involves the direct action of RanGTP, which is generated in the nucleus by the chromatin‐associated nucleotide exchange factor, RCC1. Ran interactions with RCC1 contribute to formation of a nuclear:cytoplasmic (N:C) Ran protein gradient in interphase cells. In previous work, we showed that the Ran protein gradient is disrupted in fibroblasts from Hutchinson–Gilford progeria syndrome (HGPS) patients. The Ran gradient disruption in these cells is caused by nuclear membrane association of a mutant form of Lamin A, which induces a global reduction in heterochromatin marked with Histone H3K9me3 and Histone H3K27me3. Here, we have tested the hypothesis that heterochromatin controls the Ran gradient. Chemical inhibition and depletion of the histone methyltransferases (HMTs) G9a and GLP in normal human fibroblasts reduced heterochromatin levels and caused disruption of the Ran gradient, comparable to that observed previously in HGPS fibroblasts. HMT inhibition caused a defect in nuclear localization of TPR, a high molecular weight protein that, owing to its large size, displays a Ran‐dependent import defect in HGPS. We reasoned that pathways dependent on nuclear import of large proteins might be compromised in HGPS. We found that nuclear import of ATM requires the Ran gradient, and disruption of the Ran gradient in HGPS causes a defect in generating nuclear γ‐H2AX in response to ionizing radiation. Our data suggest a lamina–chromatin–Ran axis is important for nuclear transport regulation and contributes to the DNA damage response.
Collapse
Affiliation(s)
- Natalia Dworak
- Center for Cell Signaling; University of Virginia; Charlottesville Virginia
| | - Dawid Makosa
- Center for Cell Signaling; University of Virginia; Charlottesville Virginia
| | - Mandovi Chatterjee
- Center for Cell Signaling; University of Virginia; Charlottesville Virginia
| | - Kasey Jividen
- Center for Cell Signaling; University of Virginia; Charlottesville Virginia
| | - Chun-Song Yang
- Center for Cell Signaling; University of Virginia; Charlottesville Virginia
| | - Chelsi Snow
- Center for Cell Signaling; University of Virginia; Charlottesville Virginia
- Department of Biochemistry and Molecular Genetics; University of Virginia; Charlottesville Virginia
| | - William C. Simke
- Department of Molecular and Biomedical Sciences; University of Maine; Orono Maine
| | - Isaac G. Johnson
- Department of Molecular and Biomedical Sciences; University of Maine; Orono Maine
| | - Joshua B. Kelley
- Department of Molecular and Biomedical Sciences; University of Maine; Orono Maine
| | - Bryce M. Paschal
- Center for Cell Signaling; University of Virginia; Charlottesville Virginia
- Department of Biochemistry and Molecular Genetics; University of Virginia; Charlottesville Virginia
| |
Collapse
|
44
|
Hao S, Song H, Zhang W, Seldomridge A, Jung J, Giles AJ, Hutchinson MK, Cao X, Colwell N, Lita A, Larion M, Maric D, Abu-Asab M, Quezado M, Kramp T, Camphausen K, Zhuang Z, Gilbert MR, Park DM. Protein phosphatase 2A inhibition enhances radiation sensitivity and reduces tumor growth in chordoma. Neuro Oncol 2019; 20:799-809. [PMID: 29294092 DOI: 10.1093/neuonc/nox241] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Standard therapy for chordoma consists of surgical resection followed by high-dose irradiation. Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase involved in signal transduction, cell cycle progression, cell differentiation, and DNA repair. LB100 is a small-molecule inhibitor of PP2A designed to sensitize cancer cells to DNA damage from irradiation and chemotherapy. A recently completed phase I trial of LB100 in solid tumors demonstrated its safety. Here, we show the therapeutic potential of LB100 in chordoma. Methods Three patient-derived chordoma cell lines were used: U-CH1, JHC7, and UM-Chor1. Cell proliferation was determined with LB100 alone and in combination with irradiation. Cell cycle progression was assessed by flow cytometry. Quantitative γ-H2AX immunofluorescence and immunoblot evaluated the effect of LB100 on radiation-induced DNA damage. Ultrastructural evidence for nuclear damage was investigated using Raman imaging and transmission electron microscopy. A xenograft model was established to determine potential clinical utility of adding LB100 to irradiation. Results PP2A inhibition in concert with irradiation demonstrated in vitro growth inhibition. The combination of LB100 and radiation also induced accumulation at the G2/M phase of the cell cycle, the stage most sensitive to radiation-induced damage. LB100 enhanced radiation-induced DNA double-strand breaks. Animals implanted with chordoma cells and treated with the combination of LB100 and radiation demonstrated tumor growth delay. Conclusions Combining LB100 and radiation enhanced DNA damage-induced cell death and delayed tumor growth in an animal model of chordoma. PP2A inhibition by LB100 treatment may improve the effectiveness of radiation therapy for chordoma.
Collapse
Affiliation(s)
- Shuyu Hao
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hua Song
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Wei Zhang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Ashlee Seldomridge
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jinkyu Jung
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Amber J Giles
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Marsha-Kay Hutchinson
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Xiaoyu Cao
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Nicole Colwell
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Adrian Lita
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Mioara Larion
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Dragan Maric
- Flow Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Mones Abu-Asab
- Ultrastructural Pathology Section, National Eye Institute, Bethesda, Maryland, USA
| | - Martha Quezado
- Neuropathology Section, Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | - Tamalee Kramp
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kevin Camphausen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Zhengping Zhuang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Deric M Park
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
45
|
γH2AX prefers late replicating metaphase chromosome regions. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 836:114-121. [PMID: 30442336 DOI: 10.1016/j.mrgentox.2018.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 04/20/2018] [Accepted: 06/01/2018] [Indexed: 11/23/2022]
Abstract
DNA damage response (DDR) constitutes a protein pathway to handle eukaryotic DNA lesions in the context of chromatin. DDR engages the recruitment of signaling, transducer, effector, chromatin modifiers and remodeling proteins, allowing cell cycle delay, DNA repair or induction of senescence or apoptosis. An early DDR-event includes the epigenetic phosphorylation of the histone variant H2AX on serine 139 of the C-termini, so-called gammaH2AX. GammaH2AX foci detected by immunolabeling on interphase nuclei have been largely studied; nonetheless gammaH2AX signals on mitotic chromosomes are less understood. The CHO9 cell line is a subclone of CHO (Chinese hamster ovary) cells with original and rearranged Z chromosomes originated during cell line transformation. As a result, homologous chromosome regions have been relocated in different Z-chromosomes. In a first quantitative analysis of gammaH2AX signals on immunolabeled mitotic chromosomes of cytocentrifuged metaphase spreads, we reported that gammaH2AX139 signals of both control and bleomycin-exposed cultures showed statistically equal distribution between CHO9 homologous chromosome regions, suggesting a possible dependence on the structure/function of chromatin. We have also demonstrated that bleomycin-induced gammaH2AX foci map preferentially to DNA replicating domains in CHO9 interphase nuclei. With the aim of understanding the role of gammaH2AX signals on metaphase chromosomes, the relation between 5-ethynyl-2'-deoxyuridine (EdU) labeled replicating chromosome regions and gammaH2AX signals in immunolabeled cytocentrifuged metaphase spreads from control and bleomycin-treated CHO9 cultures was analyzed in the present work. A quantitative analysis of colocalization between EdU and gammaH2AX signals based on the calculation of the Replication Related Damage Distribution Index (RDDI) on confocal metaphase images was performed. RDDI revealed a colocalization between EdU and gammaH2AX signals both in control and bleomycin-treated CHO9 metaphases, suggesting that replication may be involved in H2AX phosphorylation. The possible mechanisms implicated are discussed.
Collapse
|
46
|
Shigeta M, Kanazawa H, Yokoyama T. Tubular cell loss in early inv/nphp2 mutant kidneys represents a possible homeostatic mechanism in cortical tubular formation. PLoS One 2018; 13:e0198580. [PMID: 29889867 PMCID: PMC5995398 DOI: 10.1371/journal.pone.0198580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/22/2018] [Indexed: 11/25/2022] Open
Abstract
Inversion of embryonic turning (inv) cystic mice develop multiple renal cysts and are a model for type II nephronophthisis (NPHP2). The defect of planar cell polarity (PCP) by oriented cell division was proposed as the underlying cellular phenotype, while abnormal cell proliferation and apoptosis occur in some polycystic kidney disease models. However, how these cystogenic phenotypes are linked and what is most critical for cystogenesis remain largely unknown. In particular, in early cortical cytogenesis in the inv mutant cystic model, it remains uncertain whether the increased proliferation index results from changes in cell cycle length or cell fate determination. To address tubular cell kinetics, doubling time and total number of tubular cells, as well as amount of genomic DNA (gDNA), were measured in mutant and normal control kidneys. Despite a significantly higher bromodeoxyuridine (BrdU)-proliferation index in the mutant, total tubular cell number and doubling time were unaffected. Unexpectedly, the mutant had tubular cell loss, characterized by a temporal decrease in tubular cells incorporating 5-ethynyl-2´-deoxyuridine (EdU) and significantly increased nuclear debris. Based on current data we established a new multi-population shift model in postnatal renal development, indicating that a few restricted tubular cell populations contribute to cortical tubular formation. As in the inv mutant phenotype, the model simulation revealed a large population of tubular cells with rapid cell cycling and tubular cell loss. The proposed cellular kinetics suggest not only the underlying mechanism of the inv mutant phenotype but also a possible renal homeostatic mechanism for tubule formation.
Collapse
Affiliation(s)
- Masaki Shigeta
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto Prefectural of Medicine, Kyoto, Japan
- * E-mail:
| | - Hirotaka Kanazawa
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto Prefectural of Medicine, Kyoto, Japan
| | - Takahiko Yokoyama
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto Prefectural of Medicine, Kyoto, Japan
| |
Collapse
|
47
|
Sisario D, Memmel S, Doose S, Neubauer J, Zimmermann H, Flentje M, Djuzenova CS, Sauer M, Sukhorukov VL. Nanostructure of DNA repair foci revealed by superresolution microscopy. FASEB J 2018; 32:fj201701435. [PMID: 29894665 DOI: 10.1096/fj.201701435] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Induction of DNA double-strand breaks (DSBs) by ionizing radiation leads to formation of micrometer-sized DNA-repair foci, whose organization on the nanometer-scale remains unknown because of the diffraction limit (∼200 nm) of conventional microscopy. Here, we applied diffraction-unlimited, direct stochastic optical-reconstruction microscopy ( dSTORM) with a lateral resolution of ∼20 nm to analyze the focal nanostructure of the DSB marker histone γH2AX and the DNA-repair protein kinase (DNA-PK) in irradiated glioblastoma multiforme cells. Although standard confocal microscopy revealed substantial colocalization of immunostained γH2AX and DNA-PK, in our dSTORM images, the 2 proteins showed very little (if any) colocalization despite their close spatial proximity. We also found that γH2AX foci consisted of distinct circular subunits ("nanofoci") with a diameter of ∼45 nm, whereas DNA-PK displayed a diffuse, intrafocal distribution. We conclude that γH2AX nanofoci represent the elementary, structural units of DSB repair foci, that is, individual γH2AX-containing nucleosomes. dSTORM-based γH2AX nanofoci counting and distance measurements between nanofoci provided quantitative information on the total amount of chromatin involved in DSB repair as well as on the number and longitudinal distribution of γH2AX-containing nucleosomes in a chromatin fiber. We thus estimate that a single focus involves between ∼0.6 and ∼1.1 Mbp of chromatin, depending on radiation treatment. Because of their ability to unravel the nanostructure of DSB-repair foci, dSTORM and related single-molecule localization nanoscopy methods will likely emerge as powerful tools in biology and medicine to elucidate the effects of DNA damaging agents in cells.-Sisario, D., Memmel, S., Doose, S., Neubauer, J., Zimmermann, H., Flentje, M., Djuzenova, C. S., Sauer, M., Sukhorukov, V. L. Nanostructure of DNA repair foci revealed by superresolution microscopy.
Collapse
Affiliation(s)
- Dmitri Sisario
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| | - Simon Memmel
- Department of Radiation Oncology, University Hospital of Würzburg, Würzburg, Germany
| | - Sören Doose
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| | - Julia Neubauer
- Fraunhofer Institute for Biomedical Engineering (IBMT), Sulzbach, Germany
| | - Heiko Zimmermann
- Fraunhofer Institute for Biomedical Engineering (IBMT), Sulzbach, Germany
- Molekulare und Zelluläre Biotechnologie/Nanotechnologie, Universität des Saarlandes, Saarbrücken, Germany
- Marine Sciences, Universidad Católica del Norte, Antafogasta/Coquimbo, Chile
| | - Michael Flentje
- Department of Radiation Oncology, University Hospital of Würzburg, Würzburg, Germany
| | - Cholpon S Djuzenova
- Department of Radiation Oncology, University Hospital of Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| | - Vladimir L Sukhorukov
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| |
Collapse
|
48
|
Jiang H, Gao Q, Zheng W, Yin S, Wang L, Zhong L, Ali A, Khan T, Hao Q, Fang H, Sun X, Xu P, Pandita TK, Jiang X, Shi Q. MOF influences meiotic expansion of H2AX phosphorylation and spermatogenesis in mice. PLoS Genet 2018; 14:e1007300. [PMID: 29795555 PMCID: PMC6019819 DOI: 10.1371/journal.pgen.1007300] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 06/26/2018] [Accepted: 03/07/2018] [Indexed: 12/11/2022] Open
Abstract
Three waves of H2AX phosphorylation (γH2AX) have been observed in male meiotic prophase I: the first is ATM-dependent and occurs at leptonema, while the second and third are ATR-dependent, occuring at zygonema and pachynema, respectively. The third wave of H2AX phosphorylation marks and silences unsynapsed chromosomes. Little is known about H2AX phosphorylation expands to chromatin-wide regions in spermatocytes. Here, we report that histone acetyltransferase (HAT) MOF is involved in all three waves of H2AX phosphorylation expansion. Germ cell-specific deletion of Mof in spermatocytes by Stra8-Cre (Mof cKO) caused global loss of H4K16ac. In leptotene and zygotene spermatocytes of cKO mice, the γH2AX signals were observed only along the chromosomal axes, and chromatin-wide H2AX phosphorylation was lost. In almost 40% of early-mid pachytene spermatocytes from Mof cKO mice, γH2AX and MDC1 were detected along the unsynapsed axes of the sex chromosomes, but failed to expand, which consequently caused meiotic sex chromosome inactivation (MSCI) failure. Furthermore, though RAD51 was proficiently recruited to double-strand break (DSB) sites, defects in DSB repair and crossover formation were observed in Mof cKO spermatocytes, indicating that MOF facilitates meiotic DSB repair after RAD51 recruitment. We propose that MOF regulates male meiosis and is involved in the expansion of all three waves of H2AX phosphorylation from the leptotene to pachytene stages, initiated by ATM and ATR, respectively.
Collapse
Affiliation(s)
- Hanwei Jiang
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Qian Gao
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Wei Zheng
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Shi Yin
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Liu Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Liangwen Zhong
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Asim Ali
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Teka Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Qiaomei Hao
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Hui Fang
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Xiaoling Sun
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Peng Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Tej K. Pandita
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX, United States
| | - Xiaohua Jiang
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| | - Qinghua Shi
- Hefei National Laboratory for Physical Sciences at the Microscale, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China (USTC), Collaborative Innovation Center of Genetics and Development, Hefei, Anhui, China
| |
Collapse
|
49
|
Lee JYJ, Miller JA, Basu S, Kee TZV, Loo LH. Building predictive in vitro pulmonary toxicity assays using high-throughput imaging and artificial intelligence. Arch Toxicol 2018; 92:2055-2075. [PMID: 29705884 PMCID: PMC6002469 DOI: 10.1007/s00204-018-2213-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/25/2018] [Indexed: 01/22/2023]
Abstract
Human lungs are susceptible to the toxicity induced by soluble xenobiotics. However, the direct cellular effects of many pulmonotoxic chemicals are not always clear, and thus, a general in vitro assay for testing pulmonotoxicity applicable to a wide variety of chemicals is not currently available. Here, we report a study that uses high-throughput imaging and artificial intelligence to build an in vitro pulmonotoxicity assay by automatically comparing and selecting human lung-cell lines and their associated quantitative phenotypic features most predictive of in vivo pulmonotoxicity. This approach is called “High-throughput In vitro Phenotypic Profiling for Toxicity Prediction” (HIPPTox). We found that the resulting assay based on two phenotypic features of a human bronchial epithelial cell line, BEAS-2B, can accurately classify 33 reference chemicals with human pulmonotoxicity information (88.8% balance accuracy, 84.6% sensitivity, and 93.0% specificity). In comparison, the predictivity of a standard cell-viability assay on the same set of chemicals is much lower (77.1% balanced accuracy, 84.6% sensitivity, and 69.5% specificity). We also used the assay to evaluate 17 additional test chemicals with unknown/unclear human pulmonotoxicity, and experimentally confirmed that many of the pulmonotoxic reference and predicted-positive test chemicals induce DNA strand breaks and/or activation of the DNA-damage response (DDR) pathway. Therefore, HIPPTox helps us to uncover these common modes-of-action of pulmonotoxic chemicals. HIPPTox may also be applied to other cell types or models, and accelerate the development of predictive in vitro assays for other cell-type- or organ-specific toxicities.
Collapse
Affiliation(s)
- Jia-Ying Joey Lee
- Bioinformatics Institute, Agency for Science, Technology, and Research, 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore
| | - James Alastair Miller
- Bioinformatics Institute, Agency for Science, Technology, and Research, 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore
| | - Sreetama Basu
- Bioinformatics Institute, Agency for Science, Technology, and Research, 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore
| | - Ting-Zhen Vanessa Kee
- Bioinformatics Institute, Agency for Science, Technology, and Research, 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore
| | - Lit-Hsin Loo
- Bioinformatics Institute, Agency for Science, Technology, and Research, 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore.
| |
Collapse
|
50
|
Liang S, Guo J, Choi JW, Shin KT, Wang HY, Jo YJ, Kim NH, Cui XS. Protein phosphatase 2A regulatory subunit B55α functions in mouse oocyte maturation and early embryonic development. Oncotarget 2018; 8:26979-26991. [PMID: 28439046 PMCID: PMC5432312 DOI: 10.18632/oncotarget.15927] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/17/2017] [Indexed: 12/20/2022] Open
Abstract
Protein phosphatase 2A regulatory subunit B55α (PP2A-B55α) has been studied in mitosis. However, its functions in mammalian meiosis and early embryonic development remain unknown. Here, we report that PP2A-B55α is critical for mouse oocyte meiosis and preimplantation embryo development. Knockdown of PP2A-B55α in oocytes led to abnormal asymmetric division, disordered spindle dynamics, defects in chromosome congression, an increase in aneuploidy, and induction of the DNA damage response. Moreover, knockdown of PP2A-B55α in fertilized mouse zygotes impaired development to the blastocyst stage. The impairment of embryonic development might have been due to induction of sustained DNA damage in embryos, which caused apoptosis and inhibited cell proliferation and outgrowth potential at the blastocyst stage. Overall, these results provide a novel insight into the role of PP2A-B55α as a novel meiotic and embryonic competence factor at the onset of life.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Jing Guo
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Jeong-Woo Choi
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Kyung-Tae Shin
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Hai-Yang Wang
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Yu-Jin Jo
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Nam-Hyung Kim
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Xiang-Shun Cui
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| |
Collapse
|