1
|
Bril M, Boesveld JN, Coelho-Rato LS, Sahlgren CM, Bouten CVC, Kurniawan NA. Dynamic substrate topographies drive actin- and vimentin-mediated nuclear mechanoprotection events in human fibroblasts. BMC Biol 2025; 23:94. [PMID: 40189524 PMCID: PMC11974106 DOI: 10.1186/s12915-025-02199-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Dynamic physical changes in the extracellular environment of living tissues present a mechanical challenge for resident cells that can lead to damage to the nucleus, genome, and DNA. Recent studies have started to uncover nuclear mechanoprotection mechanisms that prevent excessive mechanical deformations of the nucleus. Here, we hypothesized that dynamic topographical changes in the cellular environment can be mechanically transmitted to the nucleus and trigger nuclear mechanoprotection events. We tested this using a photoresponsive hydrogel whose surface topography can be reversibly changed on demand upon light illumination, allowing us to subject cells to recurring microscale topographical changes. RESULTS With each recurring topographical change, fibroblasts were found to increasingly compact and relocate their nuclei away from the dynamic regions of the hydrogel. These cell-scale reorganization events were accompanied by an increase of global histone acetylation and decreased methylation in cells on the dynamic topographies, resulting in a minimization of DNA strand breakage. We further found that these nuclear mechanoprotection events were mediated by both vimentin intermediate filaments and the actin cytoskeleton. CONCLUSIONS Together, these data reveal that fibroblasts actively protect their nuclei in the presence of dynamic topographical changes through cytoskeleton-mediated mechanisms. Broadly, these results stress the importance of gaining a deeper fundamental understanding of the cellular mechanoresponse under dynamically changing conditions.
Collapse
Affiliation(s)
- Maaike Bril
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Jules N Boesveld
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Leila S Coelho-Rato
- Faculty of Science and Engineering, Åbo Akademi University, 20520, Turku, Finland
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Faculty of Science and Engineering, Åbo Akademi University, 20520, Turku, Finland
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands.
| |
Collapse
|
2
|
Seale C, Barazas M, van Schendel R, Tijsterman M, Gonçalves JP. MUSICiAn: Genome-wide Identification of Genes Involved in DNA Repair via Control-Free Mutational Spectra Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635038. [PMID: 39975194 PMCID: PMC11838396 DOI: 10.1101/2025.01.27.635038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Motivation Understanding the factors involved in DNA double-strand break (DSB) repair is crucial for the development of targeted anti-cancer therapies, yet the roles of many genes remain unclear. Recent studies show that perturbations of certain genes can alter the distribution of sequence-specific mutations left behind after DSB repair. This suggests that genome-wide screening could reveal novel DSB repair factors by identifying genes whose perturbation causes the mutational distribution spectra observed at a given DSB site to deviate significantly from the wild-type. However, designing proper controls for a genome-wide perturbation screen could be challenging. We explore the idea that a genome-wide screen might allow us to forgo the use of traditional non-targeting controls by reframing the analysis as an outlier detection problem, assuming that most genes have minimal influence on DSB repair. Results We propose MUSICiAn (Mutational Signature Catalogue Analysis), a compositional data analysis method that ranks gene perturbation-specific mutational spectra without controls by measuring deviations from the central tendency in the distributions of all spectra. We show that MUSICiAn can effectively estimate pseudo-controls for the existing Repair-seq dataset, screening 476 genes and 60 non-targeting controls. We further apply MUSICiAn to a genome-wide dataset profiling mutational outcomes induced by CRISPR-Cas9 at three target sites across cells with individual perturbations of 18,406 genes. MUSICiAn successfully recovers known genes, highlights the spliceosome as a lesser-appreciated player in DSB repair, and reveals candidates for further investigation. Availability github.com/joanagoncalveslab/MUSICiAn.
Collapse
Affiliation(s)
- Colm Seale
- Pattern Recognition & Bioinformatics, Department of Intelligent Systems, EEMCS Faculty, Delft University of Technology, Delft, The Netherlands
- Holland Proton Therapy Center, Delft, The Netherlands
| | - Marco Barazas
- Human Genetics Department, EEMCS Faculty, Leiden University Medical Centre, Leiden, The Netherlands
| | - Robin van Schendel
- Human Genetics Department, EEMCS Faculty, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marcel Tijsterman
- Human Genetics Department, EEMCS Faculty, Leiden University Medical Centre, Leiden, The Netherlands
| | - Joana P. Gonçalves
- Pattern Recognition & Bioinformatics, Department of Intelligent Systems, EEMCS Faculty, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
3
|
Jiang T, Zhang C, Cao X, Tian Y, Cai H, Kong S, Lu J, Wang H, Lu Z. EZH1/2 plays critical roles in oocyte meiosis prophase I in mice. Biol Res 2024; 57:83. [PMID: 39511641 PMCID: PMC11545252 DOI: 10.1186/s40659-024-00564-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUD abnormalities or defects in oocyte meiosis can result in decreased oocyte quality, reduced ovarian reserve, and female diseases. However, the mechanisms of oocyte meiosis remain largely unknown, especially epigenetic regulation. Here, we explored the role of EZH1/2 (histone methyltransferase of H3K27) in mouse oocyte meiosis by inhibiting its activity and deleting its gene. RESULTS with embryonic ovary cultured in vitro, EZH1/2 was demonstrated to be essential for oocyte development during meiosis prophase I in mice. Activity inhibition or gene knockout of EZH1/2 resulted in cell apoptosis and a reduction in oocyte numbers within embryonic ovaries. By observing the expression of some meiotic marker protein (γ-H2AX, diplotene stage marker MSY2 and synapsis complex protein SCP1), we found that function deficiency of EZH1/2 resulted in failure of DNA double-strand breaks (DSBs) repair and break of meiotic progression in fetal mouse ovaries. Moreover, Ezh1/2 deficiency led to the suppression of ATM (Ataxia Telangiectasia Mutated kinase) phosphorylation and a decrease in the expression of key DNA repair proteins Hormad1, Mre11, Rad50, and Nbs1 in fetal mouse ovaries, underscoring the enzyme's pivotal role in initiating DNA repair. RNA-seq analysis revealed that Ezh1/2-deletion induced abnormal expression of multiple genes involved into several function of oocyte development in embryonic ovaries. Knockout of Ezh1/2 in ovaries also affected the levels of H3K9me3 and H4K20me2, as well as the expression of their target genes L3mbtl4 and Fbxo44. CONCLUSIONS our study demonstrated that EZH1/2 plays a role in the DSBs repair in oocyte meiosis prophase I via multiple mechanisms and offers new insights into the physiological regulatory role of histone modification in fetal oocyte guardianship and female fertility.
Collapse
Affiliation(s)
- Ting Jiang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chengxiu Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361005, China
| | - Xinjing Cao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yingpu Tian
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361005, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
- Medical College of Xiamen University, Xiamen, Fujian, 361102, China.
| | - Zhongxian Lu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361005, China.
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
- School of Pharmaceutical Sciences, Xiamen University, Zhuangjin Hall, Room 363, Xiamen, Fujian, 361102, China.
| |
Collapse
|
4
|
Bian X, Piipponen M, Liu Z, Luo L, Geara J, Chen Y, Sangsuwan T, Maselli M, Diaz C, Bain CA, Eenjes E, Genander M, Crichton M, Cash JL, Archambault L, Haghdoost S, Fradette J, Sommar P, Halle M, Xu Landén N. Epigenetic memory of radiotherapy in dermal fibroblasts impairs wound repair capacity in cancer survivors. Nat Commun 2024; 15:9286. [PMID: 39468077 PMCID: PMC11519383 DOI: 10.1038/s41467-024-53295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Radiotherapy (RT), a common cancer treatment, unintentionally harms surrounding tissues, including the skin, and hinders wound healing years after treatment. This study aims to understand the mechanisms behind these late-onset adverse effects. We compare skin biopsies from previously irradiated (RT+) and non-irradiated (RT-) sites in breast cancer survivors who underwent RT years ago. Here we show that the RT+ skin has compromised healing capacity and fibroblast functions. Using ATAC-seq, we discover altered chromatin landscapes in RT+ fibroblasts, with THBS1 identified as a crucial epigenetically primed wound repair-related gene. This is further confirmed by single-cell RNA-sequencing and spatial transcriptomic analysis of human wounds. Notably, fibroblasts in both murine and human post-radiation wound models show heightened and sustained THBS1 expression, impairing fibroblast motility and contractility. Treatment with anti-THBS1 antibodies promotes ex vivo wound closure in RT+ skin from breast cancer survivors. Our findings suggest that fibroblasts retain a long-term radiation memory in the form of epigenetic changes. Targeting this maladaptive epigenetic memory could mitigate RT's late-onset adverse effects, improving the quality of life for cancer survivors.
Collapse
Affiliation(s)
- Xiaowei Bian
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Minna Piipponen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zhuang Liu
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lihua Luo
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jennifer Geara
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Traimate Sangsuwan
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Monica Maselli
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Candice Diaz
- Centre de recherche en organogénèse expérimentale de l'Université Laval / LOEX, Québec, QC, Canada
- Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
| | - Connor A Bain
- Institute of Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Evelien Eenjes
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Maria Genander
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Michael Crichton
- Institute of Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Jenna L Cash
- Centre for Inflammation Research, Institute for Regeneration and Repair, 4-5 Little France Drive, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Louis Archambault
- Department of Physics, Université Laval/Centre de Recherche sur le Cancer, Université Laval/Centre de recherche du CHU de Québec, Québec, QC, Canada
| | - Siamak Haghdoost
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- ABTE/ToxEMAC laboratory, University of Caen Normandy, Advanced Resource Center for HADrontherapy in Europe (ARCHADE), Caen, France
| | - Julie Fradette
- Centre de recherche en organogénèse expérimentale de l'Université Laval / LOEX, Québec, QC, Canada
- Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Pehr Sommar
- Department of Plastic and Reconstructive Surgery, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Martin Halle
- Department of Plastic and Reconstructive Surgery, Karolinska University Hospital, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| | - Ning Xu Landén
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
5
|
Kim JE, Pan X, Tse KY, Chan HH, Dong C, Huen MSY. PHF8 facilitates transcription recovery following DNA double-strand break repair. Nucleic Acids Res 2024; 52:10297-10310. [PMID: 39087553 PMCID: PMC11417394 DOI: 10.1093/nar/gkae661] [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: 11/13/2023] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Transient halting of transcription activity on the damaged chromatin facilitates DNA double-strand break (DSB) repair. However, the molecular mechanisms that facilitate transcription recovery following DSB repair remain largely undefined. Notably, failure to restore gene expression in a timely manner can compromise transcriptome signatures and may impose deleterious impacts on cell identity and cell fate. Here, we report PHF8 as the major demethylase that reverses transcriptionally repressive epigenetic modification laid down by the DYRK1B-EHMT2 pathway. We found that PHF8 concentrates at laser-induced DNA damage tracks in a DYRK1B-dependent manner and promotes timely resolution of local H3K9me2 to facilitate the resumption of transcription. Moreover, PHF8 also assists in the recovery of ribosomal DNA (rDNA) transcription following the repair of nucleolar DSBs. Taken together, our findings uncover PHF8 as a key mediator that coordinates transcription activities during the recovery phase of DSB responses.
Collapse
Affiliation(s)
- Jung Eun Kim
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
| | - Xiyue Pan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
| | - Kwan Yiu Tse
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
| | - Henry Hei Chan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
| | - Chao Dong
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
- Department of Occupational and Environmental Health, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Michael Shing Yan Huen
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R
| |
Collapse
|
6
|
Ren XG, Li W, Li WX, Yu WQ. Mechanism of Histone Arginine Methylation Dynamic Change in Cellular Stress. Int J Mol Sci 2024; 25:7562. [PMID: 39062806 PMCID: PMC11277302 DOI: 10.3390/ijms25147562] [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: 05/08/2024] [Revised: 06/03/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Histone arginine residue methylation is crucial for individual development and gene regulation. However, the dynamics of histone arginine methylation in response to cellular stress remains largely unexplored. In addition, the interplay and regulatory mechanisms between this and other histone modifications are important scientific questions that require further investigation. This study aimed to investigate the changes in histone arginine methylation in response to DNA damage. We report a global decrease in histone H3R26 symmetric dimethylation (H3R26me2s) and hypoacetylation at the H3K27 site in response to DNA damage. Notably, H3R26me2s exhibits a distribution pattern similar to that of H3K27ac across the genome, both of which are antagonistic to H3K27me3. Additionally, histone deacetylase 1 (HDAC1) may be recruited to the H3R26me2s demethylation region to mediate H3K27 deacetylation. These findings suggest crosstalk between H3R26me2s and H3K27ac in regulating gene expression.
Collapse
Affiliation(s)
| | | | | | - Wen-Qiang Yu
- Department of RNA Epigenetics, Faculty of Institute of Biomedical Sciences, Campus of Shanghai Medical College, Fudan University, Shanghai 200032, China; (X.-G.R.); (W.L.); (W.-X.L.)
| |
Collapse
|
7
|
González-Alvarez ME, Inyang I, Keating AF. Exposure to 7,12-dimethylbenz[a]anthracene impacts ovarian DNA damage sensing and repair proteins differently in lean and obese female mice and weight loss may mitigate obesity-induced ovarian dysfunction. Toxicol Appl Pharmacol 2024; 486:116930. [PMID: 38626870 DOI: 10.1016/j.taap.2024.116930] [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: 02/29/2024] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 05/21/2024]
Abstract
Obesity impairs oocyte quality, fertility, pregnancy maintenance, and is associated with offspring birth defects. The model ovotoxicant, 7,12-dimethylbenz[a]anthracene (DMBA), causes ovarian DNA damage and follicle loss. Both DMBA-induced chemical biotransformation and the DNA damage response are partially attenuated in obese relative to lean female mice but whether weight loss could improve the DNA damage response to DMBA exposure has not been explored. Thus, at six weeks of age, C57BL/6 J female mice were divided in three groups: 1) Lean (L; n = 20) fed a chow diet for 12 weeks, 2) obese (O; n = 20) fed a high fat high sugar (HFHS) diet for 12 weeks and, 3) slim-down (S; n = 20). The S group was fed with HFHS diet for 7 weeks until attaining a higher body relative to L mice on week 7.5 and switched to a chow diet for 5 weeks to achieve weight loss. Mice then received either corn oil (CT) or DMBA (D; 1 mg/kg) for 7 d via intraperitoneal injection (n = 10/treatment). Obesity increased (P < 0.05) kidney and spleen weight, and DMBA decreased uterine weight (P < 0.05). Ovarian weight was reduced (P < 0.05) in S mice, but DMBA exposure increased ovary weight in the S mice. LC-MS/MS identified 18, 64, and 7 ovarian proteins as altered (P < 0.05) by DMBA in the L, S and O groups, respectively. In S and O mice, 24 and 8 proteins differed, respectively, from L mice. These findings support weight loss as a strategy to modulate the ovarian genotoxicant response.
Collapse
Affiliation(s)
| | - Imaobong Inyang
- Department of Animal Science, Iowa State University, United States of America
| | - Aileen F Keating
- Department of Animal Science, Iowa State University, United States of America.
| |
Collapse
|
8
|
Valdez BC, Yuan B, Murray D, Ramdial JL, Popat U, Nieto Y, Andersson BS. ABT199/venetoclax synergism with thiotepa enhances the cytotoxicity of fludarabine, cladribine and busulfan in AML cells. Oncotarget 2024; 15:220-231. [PMID: 38484153 PMCID: PMC10939475 DOI: 10.18632/oncotarget.28563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
ABT199/venetoclax, an inhibitor of the pro-survival BCL-2 protein, has improved AML treatment. Its efficacy in hematopoietic stem cell transplantation (HSCT), when combined with other chemotherapeutic drugs, has not been thoroughly investigated. The present study demonstrates the synergistic cytotoxicity of ABT199/venetoclax with the DNA alkylator thiotepa (Thio) in AML cells. Cleavage of Caspase 3, PARP1 and HSP90, as well as increased Annexin V positivity, suggest potent activation of apoptosis by this two-drug combination; increased levels of γ-H2AX, P-CHK1 (S317), P-CHK2 (S19) and P-SMC1 (S957) indicate an enhanced DNA damage response. Likewise, the increased level of P-SAPK/JNK (T183/Y185) and decreased P-PI3Kp85 (Y458) suggest enhanced activation of stress signaling pathways. These molecular readouts were synergistically enhanced when ABT199/venetoclax and Thio were combined with fludarabine, cladribine and busulfan. The five-drug combination decreased the levels of BCL-2, BCL-xL and MCL-1, suggesting its potential clinical relevance in overcoming ABT199/venetoclax resistance. Moreover, this combination is active against P53-negative and FLT3-ITD-positive cell lines. Enhanced activation of apoptosis was observed in leukemia patient-derived cell samples exposed to the five-drug combination, suggesting a clinical relevance. The results provide a rationale for clinical trials using these two- and five-drug combinations as part of a conditioning regimen for AML patients undergoing HSCT.
Collapse
Affiliation(s)
- Benigno C. Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bin Yuan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David Murray
- Department of Oncology, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Jeremy L. Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Borje S. Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
9
|
Zaalberg A, Pottendorfer E, Zwart W, Bergman AM. It Takes Two to Tango: The Interplay between Prostate Cancer and Its Microenvironment from an Epigenetic Perspective. Cancers (Basel) 2024; 16:294. [PMID: 38254784 PMCID: PMC10813511 DOI: 10.3390/cancers16020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer is the second most common cancer in men worldwide and is associated with high morbidity and mortality. Consequently, there is an urgent unmet need for novel treatment avenues. In addition to somatic genetic alterations, deviations in the epigenetic landscape of cancer cells and their tumor microenvironment (TME) are critical drivers of prostate cancer initiation and progression. Unlike genomic mutations, epigenetic modifications are potentially reversible. Therefore, the inhibition of aberrant epigenetic modifications represents an attractive and exciting novel treatment strategy for castration-resistant prostate cancer patients. Moreover, drugs targeting the epigenome also exhibit synergistic interactions with conventional therapeutics by directly enhancing their anti-tumorigenic properties by "priming" the tumor and tumor microenvironment to increase drug sensitivity. This review summarizes the major epigenetic alterations in prostate cancer and its TME, and their involvement in prostate tumorigenesis, and discusses the impact of epigenome-targeted therapies.
Collapse
Affiliation(s)
- Anniek Zaalberg
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
| | - Elisabeth Pottendorfer
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
| | - Wilbert Zwart
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Oncode Institute
| | - Andries M. Bergman
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
- Division of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
10
|
Zhuang X, Yao J, Li X, Jiang Y, Zhong M, Tan J, Zhou H, Li G, Zha J, Xu B. Anlotinib suppresses the DNA damage response by disrupting SETD1A and inducing p53-dependent apoptosis in Transformed Follicular Lymphoma. Int J Med Sci 2024; 21:70-79. [PMID: 38164353 PMCID: PMC10750341 DOI: 10.7150/ijms.84952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/26/2023] [Indexed: 01/03/2024] Open
Abstract
Purpose: The high tumor mutational burden (TMB) of transformed follicular lymphoma (tFL) leads to tumor heterogeneity and poor prognosis in follicular lymphoma, in which endogenous DNA damage and epigenetic modification are the key factors. This study aims to evaluate the efficacy of anlotinib in tFL and to investigate its potential therapeutic mechanism. Methods: Cell viability and apoptosis were tested with CCK-8 and annexin V/PI staining kits, respectively. The tumorigenicity test in mice was utilized to further confirm the efficacy of anlotinib in vivo. Western blotting was utilized to explore the molecular mechanisms. Results: Anlotinib induced G2/M phase arrest in tFL cells, inhibited the proliferation of tFL cells and promoted the apoptosis of tFL cells in a dose-dependent manner. Administration of anlotinib markedly reduced tumor mass and weight in an FL xenograft mouse model. The western blot and immunohistochemistry staining results confirmed that the mechanism by which anlotinib promoted tumor cell apoptosis was DNA damage. Further results showed that anlotinib significantly downregulated the expression of SETD1A, leading to its destruction. Anlotinib administration resulted in a significant dose-dependent increase in the level of p-p53. Furthermore, anlotinib greatly downregulated the antiapoptotic proteins Mcl-1 and in parallel upregulated the proapoptotic element BAX and Bak, accompanied by caspase-3 activation and PARP degradation. Conclusion: Anlotinib has a good proapoptotic effect on tumor cells in vitro and in vivo, and its possible mechanism is related to the inhibition of the DNA damage response by disrupting SETD1A.
Collapse
Affiliation(s)
- Xinguo Zhuang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361102, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingwei Yao
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361102, China
| | - Xun Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yuelong Jiang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361102, China
| | - Mengya Zhong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361102, China
| | - Jinshui Tan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361102, China
| | - Hui Zhou
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361102, China
| | - Genhong Li
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361102, China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361102, China
| |
Collapse
|
11
|
Tayanloo-Beik A, Hamidpour SK, Nikkhah A, Arjmand R, Mafi AR, Rezaei-Tavirani M, Larijani B, Gilany K, Arjmand B. DNA Damage Responses, the Trump Card of Stem Cells in the Survival Game. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1470:165-188. [PMID: 37923882 DOI: 10.1007/5584_2023_791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2023]
Abstract
Stem cells, as a group of undifferentiated cells, are enriched with self-renewal and high proliferative capacity, which have attracted the attention of many researchers as a promising approach in the treatment of many diseases over the past years. However, from the cellular and molecular point of view, the DNA repair system is one of the biggest challenges in achieving therapeutic goals through stem cell technology. DNA repair mechanisms are an advantage for stem cells that are constantly multiplying to deal with various types of DNA damage. However, this mechanism can be considered a trump card in the game of cell survival and treatment resistance in cancer stem cells, which can hinder the curability of various types of cancer. Therefore, getting a deep insight into the DNA repair system can bring researchers one step closer to achieving major therapeutic goals. The remarkable thing about the DNA repair system is that this system is not only under the control of genetic factors, but also under the control of epigenetic factors. Therefore, it is necessary to investigate the role of the DNA repair system in maintaining the survival of cancer stem cells from both aspects.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amirabbas Nikkhah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasta Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Rezazadeh Mafi
- Department of Radiation Oncology, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Kambiz Gilany
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Réthi-Nagy Z, Ábrahám E, Sinka R, Juhász S, Lipinszki Z. Protein Phosphatase 4 Is Required for Centrobin Function in DNA Damage Repair. Cells 2023; 12:2219. [PMID: 37759442 PMCID: PMC10526779 DOI: 10.3390/cells12182219] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Genome stability in human cells relies on the efficient repair of double-stranded DNA breaks, which is mainly achieved by homologous recombination (HR). Among the regulators of various cellular functions, Protein phosphatase 4 (PP4) plays a pivotal role in coordinating cellular response to DNA damage. Meanwhile, Centrobin (CNTRB), initially recognized for its association with centrosomal function and microtubule dynamics, has sparked interest due to its potential contribution to DNA repair processes. In this study, we investigate the involvement of PP4 and its interaction with CNTRB in HR-mediated DNA repair in human cells. Employing a range of experimental strategies, we investigate the physical interaction between PP4 and CNTRB and shed light on the importance of two specific motifs in CNTRB, the PP4-binding FRVP and the ATR kinase recognition SQ sequences, in the DNA repair process. Moreover, we examine cells depleted of PP4 or CNTRB and cells harboring FRVP and SQ mutations in CNTRB, which result in similar abnormal chromosome morphologies. This phenomenon likely results from the impaired resolution of Holliday junctions, which serve as crucial intermediates in HR. Taken together, our results provide new insights into the intricate mechanisms of PP4 and CNTRB-regulated HR repair and their interrelation.
Collapse
Affiliation(s)
- Zsuzsánna Réthi-Nagy
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (Z.R.-N.); (E.Á.)
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, H-6726 Szeged, Hungary
| | - Edit Ábrahám
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (Z.R.-N.); (E.Á.)
- National Laboratory for Biotechnology, Institute of Genetics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
| | - Rita Sinka
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary;
| | - Szilvia Juhász
- Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
| | - Zoltán Lipinszki
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (Z.R.-N.); (E.Á.)
- National Laboratory for Biotechnology, Institute of Genetics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
| |
Collapse
|
13
|
Oleforuh-Okoleh VU, Sikiru AB, Kakulu II, Fakae BB, Obianwuna UE, Shoyombo AJ, Adeolu AI, Ollor OA, Emeka OC. Improving hydrocarbon toxicity tolerance in poultry: role of genes and antioxidants. Front Genet 2023; 14:1060138. [PMID: 37388938 PMCID: PMC10302211 DOI: 10.3389/fgene.2023.1060138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 05/23/2023] [Indexed: 07/01/2023] Open
Abstract
Sustenance of smallholder poultry production as an alternative source of food security and income is imperative in communities exposed to hydrocarbon pollution. Exposure to hydrocarbon pollutants causes disruption of homeostasis, thereby compromising the genetic potential of the birds. Oxidative stress-mediated dysfunction of the cellular membrane is a contributing factor in the mechanism of hydrocarbon toxicity. Epidemiological studies show that tolerance to hydrocarbon exposure may be caused by the activation of genes that control disease defense pathways like aryl hydrocarbon receptor (AhR) and nuclear factor erythroid 2p45-related factor 2 (Nrf2). Disparity in the mechanism and level of tolerance to hydrocarbon fragments among species may exist and may result in variations in gene expression within individuals of the same species upon exposure. Genomic variability is critical for adaptation and serves as a survival mechanism in response to environmental pollutants. Understanding the interplay of diverse genetic mechanisms in relation to environmental influences is important for exploiting the differences in various genetic variants. Protection against pollutant-induced physiological responses using dietary antioxidants can mitigate homeostasis disruptions. Such intervention may initiate epigenetic modulation relevant to gene expression of hydrocarbon tolerance, enhancing productivity, and possibly future development of hydrocarbon-tolerant breeds.
Collapse
Affiliation(s)
| | - Akeem B. Sikiru
- Department of Animal Science, Federal University of Agriculture, Zuru, Kebbi State, Nigeria
| | - Iyenemi I. Kakulu
- Department of Estate Management, Faculty of Environmental Sciences, Rivers State University, Port Harcourt, Nigeria
| | - Barineme B. Fakae
- Department of Animal and Environmental Biology, Rivers State University, Port Harcourt, Rivers State, Nigeria
| | | | - Ayoola J. Shoyombo
- Department of Animal Science, College of Agricultural Science, Landmark University, Omu-aran, Kwara State, Nigeria
| | - Adewale I. Adeolu
- Department of Agriculture, Animal Science Programme, Alex-Ekwueme Federal University, Ikwo, Ebonyi, Nigeria
| | - Ollor A. Ollor
- Department of Medical Laboratory Science, Faculty of Science, Rivers State University, Port Harcourt, Rivers State, Nigeria
| | - Onyinyechi C. Emeka
- Department of Animal Science, Rivers State University, Port Harcourt, Rivers State, Nigeria
| |
Collapse
|
14
|
Tzeplaeff L, Wilfling S, Requardt MV, Herdick M. Current State and Future Directions in the Therapy of ALS. Cells 2023; 12:1523. [PMID: 37296644 PMCID: PMC10252394 DOI: 10.3390/cells12111523] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disorder affecting upper and lower motor neurons, with death resulting mainly from respiratory failure three to five years after symptom onset. As the exact underlying causative pathological pathway is unclear and potentially diverse, finding a suitable therapy to slow down or possibly stop disease progression remains challenging. Varying by country Riluzole, Edaravone, and Sodium phenylbutyrate/Taurursodiol are the only drugs currently approved in ALS treatment for their moderate effect on disease progression. Even though curative treatment options, able to prevent or stop disease progression, are still unknown, recent breakthroughs, especially in the field of targeting genetic disease forms, raise hope for improved care and therapy for ALS patients. In this review, we aim to summarize the current state of ALS therapy, including medication as well as supportive therapy, and discuss the ongoing developments and prospects in the field. Furthermore, we highlight the rationale behind the intense research on biomarkers and genetic testing as a feasible way to improve the classification of ALS patients towards personalized medicine.
Collapse
Affiliation(s)
- Laura Tzeplaeff
- Department of Neurology, Rechts der Isar Hospital, Technical University of Munich, 81675 München, Germany
| | - Sibylle Wilfling
- Department of Neurology, University of Regensburg, 93053 Regensburg, Germany;
- Center for Human Genetics Regensburg, 93059 Regensburg, Germany
| | - Maria Viktoria Requardt
- Formerly: Department of Neurology with Institute of Translational Neurology, Münster University Hospital (UKM), 48149 Münster, Germany;
| | - Meret Herdick
- Precision Neurology, University of Lübeck, 23562 Luebeck, Germany
| |
Collapse
|
15
|
Wei W, Zhao Y, Chai Y, Shou S, Jin H. A novel role of DOT1L in kidney diseases. Mol Biol Rep 2023; 50:5415-5423. [PMID: 37085741 DOI: 10.1007/s11033-023-08415-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/29/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND We systematically summarized the structure and biological function of DOT1L in detail, and further discussed the role of DOT1L in kidney diseases through different mechanisms. METHODS AND RESULTS We first described the role of DOT1L in various kidney diseases including AKI, CKD, DN and kidney tumor diseases. CONCLUSIONS A better understanding of DOT1L as a histone methylase based on characteristics of regulating telomere silencing, transcriptional extension, DNA damage repair and cell cycle could lead to the development of new therapeutic targets for various kidney diseases, thereby improving the prognosis of kidney disease patients.
Collapse
Affiliation(s)
- Wei Wei
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Yibo Zhao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Yanfen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Songtao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| | - Heng Jin
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
16
|
Kim T, Nosella M, Bolik-Coulon N, Harkness R, Huang S, Kay L. Correlating histone acetylation with nucleosome core particle dynamics and function. Proc Natl Acad Sci U S A 2023; 120:e2301063120. [PMID: 37011222 PMCID: PMC10104578 DOI: 10.1073/pnas.2301063120] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/06/2023] [Indexed: 04/05/2023] Open
Abstract
Epigenetic modifications of chromatin play a critical role in regulating the fidelity of the genetic code and in controlling the translation of genetic information into the protein components of the cell. One key posttranslational modification is acetylation of histone lysine residues. Molecular dynamics simulations, and to a smaller extent experiment, have established that lysine acetylation increases the dynamics of histone tails. However, a systematic, atomic resolution experimental investigation of how this epigenetic mark, focusing on one histone at a time, influences the structural dynamics of the nucleosome beyond the tails, and how this translates into accessibility of protein factors such as ligases and nucleases, has yet to be performed. Herein, using NMR spectroscopy of nucleosome core particles (NCPs), we evaluate the effects of acetylation of each histone on tail and core dynamics. We show that for histones H2B, H3, and H4, the histone core particle dynamics are little changed, even though the tails have increased amplitude motions. In contrast, significant increases to H2A dynamics are observed upon acetylation of this histone, with the docking domain and L1 loop particularly affected, correlating with increased susceptibility of NCPs to nuclease digestion and more robust ligation of nicked DNA. Dynamic light scattering experiments establish that acetylation decreases inter-NCP interactions in a histone-dependent manner and facilitates the development of a thermodynamic model for NCP stacking. Our data show that different acetylation patterns result in nuanced changes to NCP dynamics, modulating interactions with other protein factors, and ultimately controlling biological output.
Collapse
Affiliation(s)
- Tae Hun Kim
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Chemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Michael L. Nosella
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Nicolas Bolik-Coulon
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Chemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Robert W. Harkness
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Chemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Shuya Kate Huang
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Chemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Lewis E. Kay
- Department of Molecular Genetics, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Chemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| |
Collapse
|
17
|
Das C, Adhikari S, Bhattacharya A, Chakraborty S, Mondal P, Yadav SS, Adhikary S, Hunt CR, Yadav K, Pandita S, Roy S, Tainer JA, Ahmed Z, Pandita TK. Epigenetic-Metabolic Interplay in the DNA Damage Response and Therapeutic Resistance of Breast Cancer. Cancer Res 2023; 83:657-666. [PMID: 36661847 PMCID: PMC11285093 DOI: 10.1158/0008-5472.can-22-3015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/30/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023]
Abstract
Therapy resistance is imposing a daunting challenge on effective clinical management of breast cancer. Although the development of resistance to drugs is multifaceted, reprogramming of energy metabolism pathways is emerging as a central but heterogenous regulator of this therapeutic challenge. Metabolic heterogeneity in cancer cells is intricately associated with alterations of different signaling networks and activation of DNA damage response pathways. Here we consider how the dynamic metabolic milieu of cancer cells regulates their DNA damage repair ability to ultimately contribute to development of therapy resistance. Diverse epigenetic regulators are crucial in remodeling the metabolic landscape of cancer. This epigenetic-metabolic interplay profoundly affects genomic stability of the cancer cells as well as their resistance to genotoxic therapies. These observations identify defining mechanisms of cancer epigenetics-metabolism-DNA repair axis that can be critical for devising novel, targeted therapeutic approaches that could sensitize cancer cells to conventional treatment strategies.
Collapse
Affiliation(s)
- Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Swagata Adhikari
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Apoorva Bhattacharya
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | | | - Payel Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Shalini S. Yadav
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Clayton R Hunt
- Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Kamlesh Yadav
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, 77030, USA
| | - Shruti Pandita
- University of Texas Health San Antonio MD Anderson Cancer Center, San Antonio, Texas, 78229, USA
| | - Siddhartha Roy
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - John A Tainer
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zamal Ahmed
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tej K. Pandita
- Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, 77030, USA
| |
Collapse
|
18
|
Liu D, Zheng Z, Zhang S, Zhu C, Zhang H, Zhou Y. Analysis of risk factors related to acute radiation dermatitis in breast cancer patients during radiotherapy. J Cancer Res Ther 2022; 18:1903-1909. [PMID: 36647948 DOI: 10.4103/jcrt.jcrt_1203_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Aims To investigate the incidence and influencing factors of acute radiation dermatitis (ARD) induced by radiotherapy in postoperative patients with breast cancer. Methods and Materials A retrospective analysis was conducted on 598 patients with breast cancer who received postoperative radiotherapy from November 18, 2014 to September 14, 2019. The radiotherapy technology included two-dimensional radiotherapy, three-dimensional conformal radiotherapy, intensity-modulated radiotherapy, and tomotherapy. The occurrence of ARD in patients was then followed up and recorded. The independent risk factors for radiation-induced dermatitis were analyzed by using an orderly logistic regression model. Results Of the 598 patients, 431 had mild skin reactions, including pigmentation and dry desquamation (grade 1), 151 developed wet desquamation and tender erythema (grade 2), and 16 had severe skin reactions, including flaky wet scaling and erosion (grade 3). There were no grade 4 skin reactions. The severity of ARD was independent of the following factors: Age, diabetes, allergy, quadrant, pathological type, the clinical stage, the tumor stage, triple-negative breast cancer, ki-67 expression, adjuvant chemotherapy, endocrine therapy, targeted therapy, radiotherapy area, and boost irradiation. However, it was found to be dependent on the body mass index, surgery type, radiotherapy technique, node stage, and the prophylactic use of topical agents. Conclusions ARD in response to postoperative radiotherapy in patients with breast cancer is common and mild. Clinicians and patients need to cultivate awareness of the potential risk factors involved and then intervene to alleviate skin reactions and improve the quality of life.
Collapse
Affiliation(s)
- Di Liu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province; Hubei Key Laboratory of Tumour Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhewen Zheng
- Hubei Key Laboratory of Tumour Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Shuyuan Zhang
- Hubei Key Laboratory of Tumour Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Chunmei Zhu
- Hubei Key Laboratory of Tumour Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Hongyan Zhang
- Department of Radiation and Medical Oncology; Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yunfeng Zhou
- Department of Radiation and Medical Oncology; Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
19
|
Biological Mechanisms to Reduce Radioresistance and Increase the Efficacy of Radiotherapy: State of the Art. Int J Mol Sci 2022; 23:ijms231810211. [PMID: 36142122 PMCID: PMC9499172 DOI: 10.3390/ijms231810211] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Cancer treatment with ionizing radiation (IR) is a well-established and effective clinical method to fight different types of tumors and is a palliative treatment to cure metastatic stages. Approximately half of all cancer patients undergo radiotherapy (RT) according to clinical protocols that employ two types of ionizing radiation: sparsely IR (i.e., X-rays) and densely IR (i.e., protons). Most cancer cells irradiated with therapeutic doses exhibit radio-induced cytotoxicity in terms of cell proliferation arrest and cell death by apoptosis. Nevertheless, despite the more tailored advances in RT protocols in the last few years, several tumors show a relatively high percentage of RT failure and tumor relapse due to their radioresistance. To counteract this extremely complex phenomenon and improve clinical protocols, several factors associated with radioresistance, of both a molecular and cellular nature, must be considered. Tumor genetics/epigenetics, tumor microenvironment, tumor metabolism, and the presence of non-malignant cells (i.e., fibroblast-associated cancer cells, macrophage-associated cancer cells, tumor-infiltrating lymphocytes, endothelial cells, cancer stem cells) are the main factors important in determining the tumor response to IR. Here, we attempt to provide an overview of how such factors can be taken advantage of in clinical strategies targeting radioresistant tumors.
Collapse
|
20
|
Liu Q, Liu P, Ji T, Zheng L, Shen C, Ran S, Liu J, Zhao Y, Niu Y, Wang T, Dong J. The histone methyltransferase SUVR2 promotes DSB repair via chromatin remodeling and liquid-liquid phase separation. MOLECULAR PLANT 2022; 15:1157-1175. [PMID: 35610973 DOI: 10.1016/j.molp.2022.05.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/15/2022] [Accepted: 05/18/2022] [Indexed: 06/15/2023]
Abstract
Maintaining genomic integrity and stability is particularly important for stem cells, which are at the top of the cell lineage origin. Here, we discovered that the plant-specific histone methyltransferase SUVR2 maintains the genome integrity of the root tip stem cells through chromatin remodeling and liquid-liquid phase separation (LLPS) when facing DNA double-strand breaks (DSBs). The histone methyltransferase SUVR2 (MtSUVR2) has histone methyltransferase activity and catalyzes the conversion of histone H3 lysine 9 monomethylation (H3K9me1) to H3K9me2/3 in vitro and in Medicago truncatula. Under DNA damage, the proportion of heterochromatin decreased and the level of DSB damage marker γ-H2AX increased in suvr2 mutants, indicating that MtSUVR2 promotes the compaction of the chromatin structure through H3K9 methylation modification to protect DNA from damage. Interestingly, MtSUVR2 was induced by DSBs to phase separate and form droplets to localize at the damage sites, and this was confirmed by immunofluorescence and fluorescence recovery after photobleaching experiments. The IDR1 and low-complexity domain regions of MtSUVR2 determined its phase separation in the nucleus, whereas the IDR2 region determined the interaction with the homologous recombinase MtRAD51. Furthermore, we found that MtSUVR2 drove the phase separation of MtRAD51 to form "DNA repair bodies," which could enhance the stability of MtRAD51 proteins to facilitate error-free homologous recombination repair of stem cells. Taken together, our study reveals that chromatin remodeling-associated proteins participate in DNA repair through LLPS.
Collapse
Affiliation(s)
- Qianwen Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Peng Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Tuo Ji
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Lihua Zheng
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Chen Shen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shasha Ran
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jinling Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yafei Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yiding Niu
- Key Laboratory of Forage and Endemic Crop Biology, Ministry of Education, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Tao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Jiangli Dong
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
21
|
De S, Jose J, Pal A, Roy Choudhury S, Roy S. Exposure to Low UV-B Dose Induces DNA Double-Strand Breaks Mediated Onset of Endoreduplication in Vigna radiata (L.) R. Wilczek Seedlings. PLANT & CELL PHYSIOLOGY 2022; 63:463-483. [PMID: 35134223 DOI: 10.1093/pcp/pcac012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Multiple lines of evidence indicate that solar UV-B light acts as an important environmental signal in plants, regulating various cellular and metabolic activities, gene expression, growth and development. Here, we show that low levels of UV-B (4.0 kJ m-2) significantly influence plant response during early seedling development in the tropical legume crop Vigna radiata (L.) R. Wilczek. Exposure to low doses of UV-B showed relatively less growth inhibition yet remarkably enhanced lateral root formation in seedlings. Both low and high (8.0 kJ m-2) doses of UV-B treatment induced DNA double-strand breaks and activated the SOG1-related ATM-ATR-mediated DNA damage response pathway. These effects led to G2-M-phase arrest with a compromised expression of the key cell cycle regulators, including CDKB1;1, CDKB2;1 and CYCB1;1, respectively. However, along with these effects, imbibitional exposure of seeds to a low UV-B dose resulted in enhanced accumulation of FZR1/CCS52A, E2Fa and WEE1 kinase and prominent induction of endoreduplication in 7-day-old seedlings. Low dose of UV-B mediated phenotypical responses, while the onset of endoreduplication appeared to be regulated at least in part via UV-B induced reactive oxygen species accumulation. Transcriptome analyses further revealed a network of co-regulated genes associated with DNA repair, cell cycle regulation and oxidative stress response pathways that are activated upon exposure to low doses of UV-B.
Collapse
Affiliation(s)
- Sayanti De
- Department of Botany, UGC Center for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan, West Bengal 713104, India
| | - Jismon Jose
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh 517507, India
| | - Amita Pal
- Division of Plant Biology, Bose Institute, Kolkata, West Bengal 700054, India
| | - Swarup Roy Choudhury
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh 517507, India
| | - Sujit Roy
- Department of Botany, UGC Center for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan, West Bengal 713104, India
| |
Collapse
|
22
|
Chakraborty S, Singh M, Pandita RK, Singh V, Lo CS, Leonard F, Horikoshi N, Moros EG, Guha D, Hunt CR, Chau E, Ahmed KM, Sethi P, Charaka V, Godin B, Makhijani K, Scherthan H, Deck J, Hausmann M, Mushtaq A, Altaf M, Ramos KS, Bhat KM, Taneja N, Das C, Pandita TK. Heat-induced SIRT1-mediated H4K16ac deacetylation impairs resection and SMARCAD1 recruitment to double strand breaks. iScience 2022; 25:104142. [PMID: 35434547 PMCID: PMC9010620 DOI: 10.1016/j.isci.2022.104142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/16/2022] [Accepted: 03/21/2022] [Indexed: 12/17/2022] Open
Abstract
Hyperthermia inhibits DNA double-strand break (DSB) repair that utilizes homologous recombination (HR) pathway by a poorly defined mechanism(s); however, the mechanisms for this inhibition remain unclear. Here we report that hyperthermia decreases H4K16 acetylation (H4K16ac), an epigenetic modification essential for genome stability and transcription. Heat-induced reduction in H4K16ac was detected in humans, Drosophila, and yeast, indicating that this is a highly conserved response. The examination of histone deacetylase recruitment to chromatin after heat-shock identified SIRT1 as the major deacetylase subsequently enriched at gene-rich regions. Heat-induced SIRT1 recruitment was antagonized by chromatin remodeler SMARCAD1 depletion and, like hyperthermia, the depletion of the SMARCAD1 or combination of the two impaired DNA end resection and increased replication stress. Altered repair protein recruitment was associated with heat-shock-induced γ-H2AX chromatin changes and DSB repair processing. These results support a novel mechanism whereby hyperthermia impacts chromatin organization owing to H4K16ac deacetylation, negatively affecting the HR-dependent DSB repair.
Collapse
Affiliation(s)
- Sharmistha Chakraborty
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Mayank Singh
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Raj K. Pandita
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
| | - Vipin Singh
- Biophysics & Structural Genomics Division Saha Institute of Nuclear Physics, Bidhan Nagar, Kolkata, West Bengal 700064, India
- Homi Bhaba National Institute, Mumbai, India
| | - Calvin S.C. Lo
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 Rotterdam, CA, the Netherlands
| | - Fransisca Leonard
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Nobuo Horikoshi
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
| | - Eduardo G. Moros
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
- Departments of Radiation Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Deblina Guha
- Biophysics & Structural Genomics Division Saha Institute of Nuclear Physics, Bidhan Nagar, Kolkata, West Bengal 700064, India
| | - Clayton R. Hunt
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
| | - Eric Chau
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Kazi M. Ahmed
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Prayas Sethi
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Vijaya Charaka
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Biana Godin
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Kalpana Makhijani
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Harry Scherthan
- Bundeswehr Institute of Radiobiology Affiliated to the University of Ulm, Neuherbergstr. 11, 80937 Munich, Germany
| | - Jeanette Deck
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| | - Michael Hausmann
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| | - Arjamand Mushtaq
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Mohammad Altaf
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Kenneth S. Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, USA
| | - Krishna M. Bhat
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Nitika Taneja
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 Rotterdam, CA, the Netherlands
| | - Chandrima Das
- Biophysics & Structural Genomics Division Saha Institute of Nuclear Physics, Bidhan Nagar, Kolkata, West Bengal 700064, India
- Homi Bhaba National Institute, Mumbai, India
| | - Tej K. Pandita
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, USA
| |
Collapse
|
23
|
Gautam Y, Johansson E, Mersha TB. Multi-Omics Profiling Approach to Asthma: An Evolving Paradigm. J Pers Med 2022; 12:jpm12010066. [PMID: 35055381 PMCID: PMC8778153 DOI: 10.3390/jpm12010066] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
Asthma is a complex multifactorial and heterogeneous respiratory disease. Although genetics is a strong risk factor of asthma, external and internal exposures and their interactions with genetic factors also play important roles in the pathophysiology of asthma. Over the past decades, the application of high-throughput omics approaches has emerged and been applied to the field of asthma research for screening biomarkers such as genes, transcript, proteins, and metabolites in an unbiased fashion. Leveraging large-scale studies representative of diverse population-based omics data and integrating with clinical data has led to better profiling of asthma risk. Yet, to date, no omic-driven endotypes have been translated into clinical practice and management of asthma. In this article, we provide an overview of the current status of omics studies of asthma, namely, genomics, transcriptomics, epigenomics, proteomics, exposomics, and metabolomics. The current development of the multi-omics integrations of asthma is also briefly discussed. Biomarker discovery following multi-omics profiling could be challenging but useful for better disease phenotyping and endotyping that can translate into advances in asthma management and clinical care, ultimately leading to successful precision medicine approaches.
Collapse
|
24
|
Pandita TK, Hunt CR, Singh V, Adhikary S, Pandita S, Roy S, Ramos K, Das C. Role of the Histone Acetyl Transferase MOF and the Histone Deacetylase Sirtuins in Regulation of H4K16ac During DNA Damage Repair and Metabolic Programming: Implications in Cancer and Aging. Subcell Biochem 2022; 100:115-141. [PMID: 36301493 DOI: 10.1007/978-3-031-07634-3_4] [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: 06/16/2023]
Abstract
The accurate repair of genomic damage mediated by ionizing radiation (IR), chemo- or radiomimetic drugs, or other exogenous agents, is necessary for maintenance of genome integrity, preservation of cellular viability and prevention of oncogenic transformation. Eukaryotes have conserved mechanisms designed to perceive and repair the damaged DNA quite efficiently. Among the different types of DNA damage, double strand breaks (DSB) are the most detrimental. The cellular DNA DSB response is a hierarchical signaling network that integrates damage sensing and repair with chromatin structural changes that involve a range of pre-existing and induced covalent modifications. Recent studies have revealed that pre-existing histone modifications are important contributors within this signaling/repair network. This chapter discusses the role of a critical histone acetyl transferase (HAT) known as MOF (males absent on the first) and the histone deacetylases (HDACs) Sirtuins on histone H4K16 acetylation (H4K16ac) and DNA damage repair. We also discuss the role of this important histone modification in light of metabolic rewiring and its role in regulating human pathophysiologic states.
Collapse
Affiliation(s)
- Tej K Pandita
- The Houston Methodist Research Institute, Houston, TX, USA.
- Department of Cellular and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, USA.
| | - Clayton R Hunt
- The Houston Methodist Research Institute, Houston, TX, USA
| | - Vipin Singh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhaba National Institute, Mumbai, India
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Shruti Pandita
- Department of Internal Medicine, Division of Hematology, Oncology and Cellular Therapy, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Siddhartha Roy
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Kenneth Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, USA
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
25
|
Mir US, Bhat A, Mushtaq A, Pandita S, Altaf M, Pandita TK. Role of histone acetyltransferases MOF and Tip60 in genome stability. DNA Repair (Amst) 2021; 107:103205. [PMID: 34399315 DOI: 10.1016/j.dnarep.2021.103205] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/01/2021] [Accepted: 08/05/2021] [Indexed: 01/23/2023]
Abstract
The accurate repair of DNA damage specifically the chromosomal double-strand breaks (DSBs) arising from exposure to physical or chemical agents, such as ionizing radiation (IR) and radiomimetic drugs is critical in maintaining genomic integrity. The DNA DSB response and repair is facilitated by hierarchical signaling networks that orchestrate chromatin structural changes specifically histone modifications which impact cell-cycle checkpoints through enzymatic activities to repair the broken DNA ends. Various histone posttranslational modifications such as phosphorylation, acetylation, methylation and ubiquitylation have been shown to play a role in DNA damage repair. Recent studies have provided important insights into the role of histone-specific modifications in sensing DNA damage and facilitating the DNA repair. Histone modifications have been shown to determine the pathway choice for repair of DNA DSBs. This review will summarize the role of important histone acetyltransferases MOF and Tip60 mediated acetylation in repair of DNA DSBs in eukaryotic cells.
Collapse
Affiliation(s)
- Ulfat Syed Mir
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, 190006, India
| | - Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, 181143, India
| | - Arjamand Mushtaq
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, 190006, India
| | - Shruti Pandita
- Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Mohammad Altaf
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, 190006, India; Centre for Interdisciplinary Research and Innovations, University of Kashmir, Srinagar, Jammu and Kashmir, 190006, India.
| | - Tej K Pandita
- Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
26
|
González‐Arzola K, Guerra‐Castellano A, Rivero‐Rodríguez F, Casado‐Combreras MÁ, Pérez‐Mejías G, Díaz‐Quintana A, Díaz‐Moreno I, De la Rosa MA. Mitochondrial cytochrome c shot towards histone chaperone condensates in the nucleus. FEBS Open Bio 2021; 11:2418-2440. [PMID: 33938164 PMCID: PMC8409293 DOI: 10.1002/2211-5463.13176] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Despite mitochondria being key for the control of cell homeostasis and fate, their role in DNA damage response is usually just regarded as an apoptotic trigger. However, growing evidence points to mitochondrial factors modulating nuclear functions. Remarkably, after DNA damage, cytochrome c (Cc) interacts in the cell nucleus with a variety of well-known histone chaperones, whose activity is competitively inhibited by the haem protein. As nuclear Cc inhibits the nucleosome assembly/disassembly activity of histone chaperones, it might indeed affect chromatin dynamics and histone deposition on DNA. Several histone chaperones actually interact with Cc Lys residues through their acidic regions, which are also involved in heterotypic interactions leading to liquid-liquid phase transitions responsible for the assembly of nuclear condensates, including heterochromatin. This relies on dynamic histone-DNA interactions that can be modulated by acetylation of specific histone Lys residues. Thus, Cc may have a major regulatory role in DNA repair by fine-tuning nucleosome assembly activity and likely nuclear condensate formation.
Collapse
Affiliation(s)
- Katiuska González‐Arzola
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Alejandra Guerra‐Castellano
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Francisco Rivero‐Rodríguez
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Miguel Á. Casado‐Combreras
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Gonzalo Pérez‐Mejías
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Antonio Díaz‐Quintana
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Irene Díaz‐Moreno
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Miguel A. De la Rosa
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| |
Collapse
|
27
|
Banerjee S, Roy S. An insight into understanding the coupling between homologous recombination mediated DNA repair and chromatin remodeling mechanisms in plant genome: an update. Cell Cycle 2021; 20:1760-1784. [PMID: 34437813 DOI: 10.1080/15384101.2021.1966584] [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] [Indexed: 10/20/2022] Open
Abstract
Plants, with their obligatory immobility, are vastly exposed to a wide range of environmental agents and also various endogenous processes, which frequently cause damage to DNA and impose genotoxic stress. These factors subsequently increase genome instability, thus affecting plant growth and productivity. Therefore, to survive under frequent and extreme environmental stress conditions, plants have developed highly efficient and powerful defense mechanisms to repair the damages in the genome for maintaining genome stability. Such multi-dimensional signaling response, activated in presence of damage in the DNA, is collectively known as DNA Damage Response (DDR). DDR plays a crucial role in the remarkably efficient detection, signaling, and repair of damages in the genome for maintaining plant genome stability and normal growth responses. Like other highly advanced eukaryotic systems, chromatin dynamics play a key role in regulating cell cycle progression in plants through remarkable orchestration of environmental and developmental signals. The regulation of chromatin architecture and nucleosomal organization in DDR is mainly modulated by the ATP dependent chromatin remodelers (ACRs), chromatin modifiers, and histone chaperones. ACRs are mainly responsible for transcriptional regulation of several homologous recombination (HR) repair genes in plants under genotoxic stress. The HR-based repair of DNA damage has been considered as the most error-free mechanism of repair and represents one of the essential sources of genetic diversity and new allelic combinations in plants. The initiation of DDR signaling and DNA damage repair pathway requires recruitment of epigenetic modifiers for remodeling of the damaged chromatin while accumulating evidence has shown that chromatin remodeling and DDR share part of the similar signaling pathway through the altered epigenetic status of the associated chromatin region. In this review, we have integrated information to provide an overview on the association between chromatin remodeling mediated regulation of chromatin structure stability and DDR signaling in plants, with emphasis on the scope of the utilization of the available knowledge for the improvement of plant health and productivity.Abbreviation: ADH: Alcohol Dehydrogenase; AGO2: Argonaute 2; ARP: Actin-Related Protein; ASF:1- Anti-Silencing Function-1; ATM: Ataxia Telangiectasia Mutated; ATR: ATM and Rad3- Related; AtSWI3c: Arabidopsis thaliana Switch 3c; ATXR5: Arabidopsis Trithorax-Related5; ATXR6: Arabidopsis Trithorax-Related6; BER: Base Excision Repair; BRCA1: Breast Cancer Associated 1; BRM: BRAHMA; BRU1: BRUSHY1; CAF:1- Chromatin Assembly Factor-1; CHD: Chromodomain Helicase DNA; CHR5: Chromatin Remodeling Protein 5; CHR11/17: Chromatin Remodeling Protein 11/17; CIPK11- CBL- Interacting Protein Kinase 11; CLF: Curly Leaf; CMT3: Chromomethylase 3; COR15A: Cold Regulated 15A; COR47: Cold Regulated 47; CRISPR: Clustered Regulatory Interspaced Short Palindromic Repeats; DDM1: Decreased DNA Methylation1; DRR: DNA Repair and Recombination; DSBs: Double-Strand Breaks; DDR: DNA Damage Response; EXO1: Exonuclease 1; FAS1/2: Fasciata1/2; FACT: Facilitates Chromatin Transcription; FT: Flowering Locus T; GMI1: Gamma-Irradiation And Mitomycin C Induced 1; HAC1: Histone Acetyltransferase of the CBP Family 1; HAM1: Histone Acetyltransferase of the MYST Family 1; HAM2: Histone Acetyltransferase of the MYST Family 2; HAF1: Histone Acetyltransferase of the TAF Family 1; HAT: Histone Acetyl Transferase; HDA1: Histone Deacetylase 1; HDA6: Histone Deacetylase 6; HIRA: Histone Regulatory Homolog A; HR- Homologous recombination; HAS: Helicase SANT Associated; HSS: HAND-SLANT-SLIDE; ICE1: Inducer of CBF Expression 1; INO80: Inositol Requiring Mutant 80; ISW1: Imitation Switch 1; KIN1/2: Kinase 1 /2; MET1: Methyltransferase 1; MET2: Methyltransferase 2; MINU: MINUSCULE; MMS: Methyl Methane Sulfonate; MMS21: Methyl Methane Sulfonate Sensitivity 21; MRN: MRE11, RAD50 and NBS1; MSI1: Multicopy Suppressor Of Ira1; NAP1: Nucleosome Assembly Protein 1; NRP1/NRP2: NAP1-Related Protein; NER: Nucleotide Excision Repair; NHEJ: Non-Homologous End Joining; PARP1: Poly-ADP Ribose Polymerase; PIE1: Photoperiod Independent Early Flowering 1; PIKK: Phosphoinositide 3-Kinase-Like Kinase; PKL: PICKLE; PKR1/2: PICKLE Related 1/2; RAD: Radiation Sensitive Mutant; RD22: Responsive To Desiccation 22; RD29A: Responsive To Desiccation 29A; ROS: Reactive Oxygen Species; ROS1: Repressor of Silencing 1; RPA1E: Replication Protein A 1E; SANT: Swi3, Ada2, N-Cor and TFIIIB; SEP3: SEPALLATA3; SCC3: Sister Chromatid Cohesion Protein 3; SMC1: Structural Maintenance of Chromosomes Protein 1; SMC3: Structural Maintenance of Chromosomes Protein 3; SOG1: Suppressor of Gamma Response 1; SWC6: SWR1 Complex Subunit 6; SWR1: SWI2/SNF2-Related 1; SYD: SPLAYED; SMC5: Structural Maintenance of Chromosome 5; SWI/SNF: Switch/Sucrose Non-Fermentable; TALENs: Transcription Activators Like Effector Nucleases; TRRAP: Transformation/Transactivation Domain-Associated Protein; ZFNs: Zinc Finger Nucleases.
Collapse
Affiliation(s)
- Samrat Banerjee
- Department of Botany, UGC Centre for Advanced Studies, the University of Burdwan, Golapbag Campus, Burdwan, West Bengal, India
| | - Sujit Roy
- Department of Botany, UGC Centre for Advanced Studies, the University of Burdwan, Golapbag Campus, Burdwan, West Bengal, India
| |
Collapse
|
28
|
Banday S, Pandita RK, Mushtaq A, Bacolla A, Mir US, Singh DK, Jan S, Bhat KP, Hunt CR, Rao G, Charaka VK, Tainer JA, Pandita TK, Altaf M. Autism-Associated Vigilin Depletion Impairs DNA Damage Repair. Mol Cell Biol 2021; 41:e0008221. [PMID: 33941620 PMCID: PMC8224237 DOI: 10.1128/mcb.00082-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/17/2021] [Accepted: 04/28/2021] [Indexed: 12/24/2022] Open
Abstract
Vigilin (Vgl1) is essential for heterochromatin formation, chromosome segregation, and mRNA stability and is associated with autism spectrum disorders and cancer: vigilin, for example, can suppress proto-oncogene c-fms expression in breast cancer. Conserved from yeast to humans, vigilin is an RNA-binding protein with 14 tandemly arranged nonidentical hnRNP K-type homology (KH) domains. Here, we report that vigilin depletion increased cell sensitivity to cisplatin- or ionizing radiation (IR)-induced cell death and genomic instability due to defective DNA repair. Vigilin depletion delayed dephosphorylation of IR-induced γ-H2AX and elevated levels of residual 53BP1 and RIF1 foci, while reducing Rad51 and BRCA1 focus formation, DNA end resection, and double-strand break (DSB) repair. We show that vigilin interacts with the DNA damage response (DDR) proteins RAD51 and BRCA1, and vigilin depletion impairs their recruitment to DSB sites. Transient hydroxyurea (HU)-induced replicative stress in vigilin-depleted cells increased replication fork stalling and blocked restart of DNA synthesis. Furthermore, histone acetylation promoted vigilin recruitment to DSBs preferentially in the transcriptionally active genome. These findings uncover a novel vigilin role in DNA damage repair with implications for autism and cancer-related disorders.
Collapse
Affiliation(s)
- Shahid Banday
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Raj K. Pandita
- Houston Methodist Research Institute, Houston, Texas, USA
- Baylor College of Medicine, Houston, Texas, USA
| | - Arjamand Mushtaq
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Albino Bacolla
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Ulfat Syed Mir
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | | | - Sadaf Jan
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Krishna P. Bhat
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | | | - Ganesh Rao
- Baylor College of Medicine, Houston, Texas, USA
| | | | - John A. Tainer
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Tej K. Pandita
- Houston Methodist Research Institute, Houston, Texas, USA
- Baylor College of Medicine, Houston, Texas, USA
| | - Mohammad Altaf
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
- Centre for Interdisciplinary Research and Innovations, University of Kashmir, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
29
|
Kaur E, Nair J, Ghorai A, Mishra SV, Achareker A, Ketkar M, Sarkar D, Salunkhe S, Rajendra J, Gardi N, Desai S, Iyer P, Thorat R, Dutt A, Moiyadi A, Dutt S. Inhibition of SETMAR-H3K36me2-NHEJ repair axis in residual disease cells prevents glioblastoma recurrence. Neuro Oncol 2020; 22:1785-1796. [PMID: 32458986 PMCID: PMC7746947 DOI: 10.1093/neuonc/noaa128] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Residual disease of glioblastoma (GBM) causes recurrence. However, targeting residual cells has failed, due to their inaccessibility and our lack of understanding of their survival mechanisms to radiation therapy. Here we deciphered a residual cell-specific survival mechanism essential for GBM relapse. METHODS Therapy resistant residual (RR) cells were captured from primary patient samples and cell line models mimicking clinical scenario of radiation resistance. Molecular signaling of resistance in RR cells was identified using RNA sequencing, genetic and pharmacological perturbations, overexpression systems, and molecular and biochemical assays. Findings were validated in patient samples and an orthotopic mouse model. RESULTS RR cells form more aggressive tumors than the parental cells in an orthotopic mouse model. Upon radiation-induced damage, RR cells preferentially activated a nonhomologous end joining (NHEJ) repair pathway, upregulating Ku80 and Artemis while downregulating meiotic recombination 11 (Mre11) at protein but not RNA levels. Mechanistically, RR cells upregulate the Su(var)3-9/enhancer-of-zeste/trithorax (SET) domain and mariner transposase fusion gene (SETMAR), mediating high levels of H3K36me2 and global euchromatization. High H3K36me2 leads to efficiently recruiting NHEJ proteins. Conditional knockdown of SETMAR in RR cells induced irreversible senescence partly mediated by reduced H3K36me2. RR cells expressing mutant H3K36A could not retain Ku80 at double-strand breaks, thus compromising NHEJ repair, leading to apoptosis and abrogation of tumorigenicity in vitro and in vivo. Pharmacological inhibition of the NHEJ pathway phenocopied H3K36 mutation effect, confirming dependency of RR cells on the NHEJ pathway for their survival. CONCLUSIONS We demonstrate that the SETMAR-NHEJ regulatory axis is essential for the survival of clinically relevant radiation RR cells, abrogation of which prevents recurrence in GBM.
Collapse
Affiliation(s)
- Ekjot Kaur
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Jyothi Nair
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Atanu Ghorai
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Saket V Mishra
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Anagha Achareker
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Madhura Ketkar
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Debashmita Sarkar
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Sameer Salunkhe
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Jacinth Rajendra
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Nilesh Gardi
- Integrated Genomics Laboratory, ACTREC, Kharghar, Navi Mumbai, India
| | - Sanket Desai
- Integrated Genomics Laboratory, ACTREC, Kharghar, Navi Mumbai, India
| | - Prajish Iyer
- Integrated Genomics Laboratory, ACTREC, Kharghar, Navi Mumbai, India
| | - Rahul Thorat
- Laboratory Animal Facility, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Amit Dutt
- Integrated Genomics Laboratory, ACTREC, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Aliasgar Moiyadi
- Department of Neurosurgery, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Shilpee Dutt
- Integrated Genomics Laboratory, ACTREC, Kharghar, Navi Mumbai, India
| |
Collapse
|
30
|
Alkhalil A, Clifford JL, Ball R, Day A, Chan R, Carney BC, Miller SA, Campbell R, Kumar R, Gautam A, Hammamieh R, Moffatt LT, Shupp JW. Blood RNA Integrity is a Direct and Simple Reporter of Radiation Exposure and Prognosis: A Pilot Study. Radiat Res 2020; 193:543-551. [PMID: 32282289 DOI: 10.1667/rr15527.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/09/2020] [Indexed: 11/03/2022]
Abstract
In the event of a mass casualty radiation scenario, rapid assessment of patients' health and triage is required for optimal resource utilization. Identifying the level and extent of exposure as well as prioritization of care is extremely challenging under such disaster conditions. Blood-based biomarkers, such as RNA integrity numbers (RIN), could help healthcare personnel quickly and efficiently determine the extent and effect of multiple injuries on patients' health. Evaluation of the effect of different radiation doses, alone or in combination with burn injury, on total RNA integrity over multiple time points was performed. Total RNA integrity was tallied in blood samples for potential application as a marker of radiation exposure and survival. Groups of aged mice (3-6 mice/group, 13-18 months old) received 0.5, 1, 5, 10 or 20 Gy ionizing radiation. Two additional mouse groups received low-dose irradiation (0.5 or 1 Gy) with a 15% total body surface area (TBSA) burn injury. Animals were euthanized at 2 or 12 h and at day 1, 2, 3, 7 or 14 postirradiation, or when injury-mediated mortality occurred. Total RNA was isolated from blood. The quality of RNA was evaluated and RNA RIN were obtained. Analysis of RIN indicated that blood showed the clearest radiation effect. There was a time- and radiation-dose-dependent reduction in RIN that was first detectable at 12 h postirradiation for all doses in animals receiving irradiation alone. This effect was reversible in lower-dose groups (i.e., 0.5, 1 and 5 Gy) that survived to the end of the study (14 days). In contrast, the effect persisted for 10 and 20 Gy groups, which showed suppression of RIN values <4.5 with high mortalities. Radiation doses of 20 Gy were lethal and required euthanasia by day 6. A low RIN (<2.5) at any time point was associated with 100% mortality. Combined radiation-burn injury produced significantly increased mortality such that no dually-injured animals survived beyond day 3, and no radiation dose >1 Gy resulted in survival past day 1. More modest suppression of RIN was observed in the surviving dually challenged mice, and no statistically significant changes were identified in RIN values of burn-only mice at any time point. In this study of an animal model, a proof of concept is presented for a simple and accurate method of assessing radiation dose exposure in blood which potentially predicts lethality. RIN assessment of blood-derived RNA could form the basis for a clinical decision-support tool to guide healthcare providers under the strenuous conditions of a radiation-based mass casualty event.
Collapse
Affiliation(s)
- Abdulnaser Alkhalil
- Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC 20010
| | - John L Clifford
- Integrative Systems Biology Program, U.S. Army Center for Environmental Health Research, Fort Detrick, Maryland 21702
| | - Robert Ball
- Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC 20010.,The Burn Center, MedStar Washington Hospital Center, Washington, DC 20010
| | - Anna Day
- Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC 20010
| | - Rosanna Chan
- Department of Radiology, MedStar Washington Hospital Center, Washington, DC 20010
| | - Bonnie C Carney
- Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC 20010.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University School of Medicine, Washington, DC 20010
| | - Stacy Ann Miller
- Integrative Systems Biology, The Oak Ridge Institute for Science and Education, Fort Detrick, Maryland 21702-5000
| | - Ross Campbell
- Integrative Systems Biology Program, U.S. Army Center for Environmental Health Research, Fort Detrick, Maryland 21702.,Advanced Biomedical Computational Science, Frederick National Lab for Cancer Research/Advanced Biomedical Computational, Frederick, Maryland, 21702
| | - Raina Kumar
- Integrative Systems Biology Program, U.S. Army Center for Environmental Health Research, Fort Detrick, Maryland 21702.,Advanced Biomedical Computational Science, Frederick National Lab for Cancer Research/Advanced Biomedical Computational, Frederick, Maryland, 21702
| | - Aarti Gautam
- Integrative Systems Biology Program, U.S. Army Center for Environmental Health Research, Fort Detrick, Maryland 21702
| | - Rasha Hammamieh
- Integrative Systems Biology Program, U.S. Army Center for Environmental Health Research, Fort Detrick, Maryland 21702
| | - Lauren T Moffatt
- Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC 20010.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University School of Medicine, Washington, DC 20010.,Department of Surgery, Georgetown University School of Medicine, Washington, DC 20010
| | - Jeffrey W Shupp
- Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, DC 20010.,The Burn Center, MedStar Washington Hospital Center, Washington, DC 20010.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University School of Medicine, Washington, DC 20010.,Department of Surgery, Georgetown University School of Medicine, Washington, DC 20010
| |
Collapse
|
31
|
Singh M, Bacolla A, Chaudhary S, Hunt CR, Pandita S, Chauhan R, Gupta A, Tainer JA, Pandita TK. Histone Acetyltransferase MOF Orchestrates Outcomes at the Crossroad of Oncogenesis, DNA Damage Response, Proliferation, and Stem Cell Development. Mol Cell Biol 2020; 40:e00232-20. [PMID: 32661120 PMCID: PMC7459263 DOI: 10.1128/mcb.00232-20] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The DNA and protein complex known as chromatin is subject to posttranslational modifications (PTMs) that regulate cellular functions such that PTM dysregulation can lead to disease, including cancer. One critical PTM is acetylation/deacetylation, which is being investigated as a means to develop targeted cancer therapies. The histone acetyltransferase (HAT) family of proteins performs histone acetylation. In humans, MOF (hMOF), a member of the MYST family of HATs, acetylates histone H4 at lysine 16 (H4K16ac). MOF-mediated acetylation plays a critical role in the DNA damage response (DDR) and embryonic stem cell development. Functionally, MOF is found in two distinct complexes: NSL (nonspecific lethal) in humans and MSL (male-specific lethal) in flies. The NSL complex is also able to acetylate additional histone H4 sites. Dysregulation of MOF activity occurs in multiple cancers, including ovarian cancer, medulloblastoma, breast cancer, colorectal cancer, and lung cancer. Bioinformatics analysis of KAT8, the gene encoding hMOF, indicated that it is highly overexpressed in kidney tumors as part of a concerted gene coexpression program that can support high levels of chromosome segregation and cell proliferation. The linkage between MOF and tumor proliferation suggests that there are additional functions of MOF that remain to be discovered.
Collapse
Affiliation(s)
- Mayank Singh
- Department of Medical Oncology, BRAIRCH, All India Institute of Medical Sciences Delhi, New Delhi, India
| | - Albino Bacolla
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Shilpi Chaudhary
- Department of Medical Oncology, BRAIRCH, All India Institute of Medical Sciences Delhi, New Delhi, India
| | - Clayton R Hunt
- The Houston Methodist Research Institute, Houston, Texas, USA
| | - Shruti Pandita
- Department of Internal Medicine, Division of Hematology, Oncology and Cellular Therapy, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Ravi Chauhan
- Department of Medical Oncology, BRAIRCH, All India Institute of Medical Sciences Delhi, New Delhi, India
| | - Ashna Gupta
- Department of Medical Oncology, BRAIRCH, All India Institute of Medical Sciences Delhi, New Delhi, India
| | - John A Tainer
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Tej K Pandita
- The Houston Methodist Research Institute, Houston, Texas, USA
- Department of Cellular and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
32
|
Sambyal V, Kaur S, Manjari M, Uppal MS, Singh NR, Sudan M, Guleria K. Linkage disequilibrium and haplotypes of five TP53 polymorphisms in oesophageal cancer patients. J Genet 2020. [DOI: 10.1007/s12041-020-01224-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
A role of the 53BP1 protein in genome protection: structural and functional characteristics of 53BP1-dependent DNA repair. Aging (Albany NY) 2020; 11:2488-2511. [PMID: 30996128 PMCID: PMC6519998 DOI: 10.18632/aging.101917] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022]
Abstract
Nuclear architecture plays a significant role in DNA repair mechanisms. It is evident that proteins involved in DNA repair are compartmentalized in not only spontaneously occurring DNA lesions or ionizing radiation-induced foci (IRIF), but a specific clustering of these proteins can also be observed within the whole cell nucleus. For example, 53BP1-positive and BRCA1-positive DNA repair foci decorate chromocenters and can appear close to nuclear speckles. Both 53BP1 and BRCA1 are well-described factors that play an essential role in double-strand break (DSB) repair. These proteins are members of two protein complexes: 53BP1-RIF1-PTIP and BRCA1-CtIP, which make a “decision” determining whether canonical nonhomologous end joining (NHEJ) or homology-directed repair (HDR) is activated. It is generally accepted that 53BP1 mediates the NHEJ mechanism, while HDR is activated via a BRCA1-dependent signaling pathway. Interestingly, the 53BP1 protein appears relatively quickly at DSB sites, while BRCA1 is functional at later stages of DNA repair, as soon as the Mre11-Rad50-Nbs1 complex is recruited to the DNA lesions. A function of the 53BP1 protein is also linked to a specific histone signature, including phosphorylation of histone H2AX (γH2AX) or methylation of histone H4 at the lysine 20 position (H4K20me); therefore, we also discuss an epigenetic landscape of 53BP1-positive DNA lesions.
Collapse
|
34
|
Mahapatra K, Ghosh AK, De S, Ghosh N, Sadhukhan P, Chatterjee S, Ghosh R, Sil PC, Roy S. Assessment of cytotoxic and genotoxic potentials of a mononuclear Fe(II) Schiff base complex with photocatalytic activity in Trigonella. Biochim Biophys Acta Gen Subj 2020; 1864:129503. [PMID: 31816347 DOI: 10.1016/j.bbagen.2019.129503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND In recent times, coordination complexes of iron in various oxidation states along with variety of ligand systems have been designed and developed for effective treatment of cancer cells without adversely affecting the normal cell and tissues of various organs. METHODS In this study, we have evaluated the mechanism of action of a Fe(II) Schiff base complex in the crop plant Trigonella foenum-graecum L. (Fenugreek) as the screening system by using morphological, cytological, biochemical and molecular approaches. Further functional characterization was performed using MCF-7 cell line and solid tumour model for the assessment of anti-tumour activity of the complex. RESULTS Our results indicate efficiency of the Fe(II) Schiff base complex in the induction of double strand breaks in DNA. Complex treatment clearly induced cytotoxic and genotoxic damage in Trigonella seedlings. The Fe-complex treatment caused cell cycle arrest via the activation of ATM-ATR kinase mediated DNA damage response pathway with the compromised expression of CDK1, CDK2 and CyclinB1 protein in Trigonella seedlings. In cultured MCF-7 cells, the complex induces cytotoxicity and DNA fragmentation through intracellular ROS generation. Fe-complex treatment inhibited tumour growth in solid tumour model with no additional side effects. CONCLUSION The growth inhibitory and cytotoxic effects of the complex result from activation of DNA damage response along with oxidative stress and cell cycle arrest. GENERAL SIGNIFICANCE Overall, our results have provided comprehensive information on the mechanism of action and efficacy of a Fe(II) Schiff base complex in higher eukaryotic genomes and indicated its future implications as potential therapeutic agent.
Collapse
Affiliation(s)
- Kalyan Mahapatra
- Department of Botany, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India
| | - Ayon Kanti Ghosh
- Department of Chemistry, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India
| | - Sayanti De
- Department of Botany, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India
| | - Noyel Ghosh
- Division of Molecular Medicine, Bose Institute, Centenary Campus, P-1/12 C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Pritam Sadhukhan
- Division of Molecular Medicine, Bose Institute, Centenary Campus, P-1/12 C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Sharmistha Chatterjee
- Division of Molecular Medicine, Bose Institute, Centenary Campus, P-1/12 C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Rajarshi Ghosh
- Department of Chemistry, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Centenary Campus, P-1/12 C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Sujit Roy
- Department of Botany, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India.
| |
Collapse
|
35
|
Ahmed AA, Adam Essa ME. Epigenetic alterations in female urogenital organs cancer: Premise, properties, and perspectives. SCIENTIFIC AFRICAN 2020. [DOI: 10.1016/j.sciaf.2020.e00318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
36
|
Pu J, Li T, Liu N, Luo C, Quan Z, Li L, Wu X. PLCε knockdown enhances the radiosensitivity of castration‑resistant prostate cancer via the AR/PARP1/DNA‑PKcs axis. Oncol Rep 2020; 43:1397-1412. [PMID: 32323799 PMCID: PMC7108056 DOI: 10.3892/or.2020.7520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/14/2020] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy (RT) has been used as a therapeutic option for treatment of prostate cancer (PCa) for a number of years; however, patients frequently develop RT resistance, particularly in castration-resistant PCa (CRPC), although the underlying mechanisms remain unknown. Understanding the underlying mechanism of RT resistance in CRPC may potentially highlight novel targets to improve therapeutic options for patients with PCa. In the present study, the expression levels of phospholipase Cε (PLCε), androgen receptor (AR) and DNA-dependent protein kinase catalytic subunit (PKcs) were examined in PCa tissue samples and PCa cells, and the effects of PLCε knockdown on AR and DNA damage repair (DDR)-related molecules were determined. The association between PLCε, AR and Poly (ADP-ribose) polymerase 1 (PARP1), as well as their respective roles in radiation resistance, were assessed using gene knockdown and pharmaceutical inhibitors or activators. A chromatin immunoprecipitation assay was used to determine the epigenetic regulatory effects of PLCε on PARP1. Animal experiments were performed to assess whether the mechanisms observed in vitro could be replicated in vivo. The expression levels of PLCε, AR and DNA-PKcs were significantly upregulated in PCa, particularly in CRPC. PLCε knockdown reduced the viability and increased apoptosis of cells subjected to radiation. Additionally, PLCε deficiency suppressed DDR progression by downregulating an AR and PARP1 positive feedback loop and the associated downstream molecules following radiation. PLCε depletion also increased the presence of histone H3 lysine 27 trimethylation in the PARP1 promoter region, suggesting increased methylation of the PARP1 gene and thus resulting in reduced expression of PARP1. In vivo, PLCε knockdown significantly potentiated the effects of radiation on tumor growth. Taken together, the results of the present study demonstrated that PLCε knockdown enhanced the radiosensitivity of CRPC by downregulating the AR/PARP1/DNA-PKcs axis.
Collapse
Affiliation(s)
- Jun Pu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ting Li
- Key Laboratory of Diagnostics Medicine Designated by The Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Nanjing Liu
- Key Laboratory of Diagnostics Medicine Designated by The Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Chunli Luo
- Key Laboratory of Diagnostics Medicine Designated by The Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhen Quan
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Luo Li
- Key Laboratory of Diagnostics Medicine Designated by The Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaohou Wu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
37
|
Singh A, Choudhuri P, Chandradoss KR, Lal M, Mishra SK, Sandhu KS. Does genome surveillance explain the global discrepancy between binding and effect of chromatin factors? FEBS Lett 2020; 594:1339-1353. [PMID: 31930486 DOI: 10.1002/1873-3468.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 11/11/2022]
Abstract
Knocking out a chromatin factor often does not alter the transcription of its binding targets. What explains the observed disconnect between binding and effect? We hypothesize that this discrepancy could be associated with the role of chromatin factors in maintaining genetic and epigenetic integrity at promoters, and not necessarily with transcription. Through re-analysis of published datasets, we present several lines of evidence that support our hypothesis and deflate the popular assumptions. We also tested the hypothesis through mutation accumulation assays on yeast knockouts of chromatin factors. Altogether, the proposed hypothesis presents a simple explanation for the global discord between chromatin factor binding and effect. Future work in this direction might fortify the hypothesis and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Arashdeep Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER)-Mohali, India
| | - Poulami Choudhuri
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER)-Mohali, India
| | | | - Mohan Lal
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER)-Mohali, India
| | - Shravan Kumar Mishra
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER)-Mohali, India
| | - Kuljeet Singh Sandhu
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER)-Mohali, India
| |
Collapse
|
38
|
Wang Y, Xu C, Zhong B, Zhan D, Liu M, Gao D, Wang Y, Qin J. Comparative Proteomic Analysis of Histone Modifications upon Acridone Derivative 8a-Induced CCRF-CEM Cells by Data Independent Acquisition. J Proteome Res 2020; 19:819-831. [PMID: 31887055 DOI: 10.1021/acs.jproteome.9b00650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The lead compound acridone derivative 8a showed potent antiproliferative activity by inducing DNA damage through direct stacking with DNA bases and triggering ROS in CCRF-CEM cells. To define the chromatin alterations during DNA damage sensing and repair, a detailed quantitative map of single and coexisting histone post-translational modifications (PTMs) in CCRF-CEM cells affected by 8a was performed by the Data Independent Acquisition (DIA) method on QE-plus. A total of 79 distinct and 164 coexisting histone PTMs were quantified, of which 16 distinct histone PTMs were significantly altered when comparing 8a-treated cells with vehicle control cells. The changes in histone PTMs were confirmed by Western blotting analysis for three H3 and one H4 histone markers. The up-regulated dimethylation on H3K9, H3K36, and H4K20 implied that CCRF-CEM cells might accelerate DNA damage repair to counteract the DNA lesion induced by 8a, which was verified by an increment in the 53BP1 foci localization at the damaged DNA. Most of the significantly altered PTMs were involved in transcriptional regulation, including down-regulated acetylation on H3K18, H3K27, and H3K122, and up-regulated di- and trimethylation on H3K9 and H3K27. This transcription-silencing phenomenon was associated with G2/M cell cycle arrest after 8a treatment by flow cytometry. This study shows that the DIA proteomics strategy provides a sensitive and accurate way to characterize the coexisting histone PTMs changes and their cross-talk in CCRF-CEM cells after 8a treatment. Specifically, histone PTMs rearrange transcription-silencing, and cell cycle arrest DNA damage repair may contribute to the mechanism of epigenetic response affected by 8a.
Collapse
Affiliation(s)
- Yini Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center , National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics , Beijing 102206 , China
| | - Caixia Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center , National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics , Beijing 102206 , China
| | - Bowen Zhong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center , National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics , Beijing 102206 , China
| | - Dongdong Zhan
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences , East China Normal University , Shanghai 200241 , China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center , National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics , Beijing 102206 , China
| | - Dan Gao
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology , Graduate School at Shenzhen, Tsinghua University , Shenzhen 518055 , China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center , National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics , Beijing 102206 , China.,Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology , Baylor College of Medicine , Houston , Texas 77030 , United States
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center , National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics , Beijing 102206 , China.,Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology , Baylor College of Medicine , Houston , Texas 77030 , United States
| |
Collapse
|
39
|
Structural Alterations of Histone Proteins in DNA-Damaged Cells Revealed by Synchrotron Radiation Circular Dichroism Spectroscopy: A New Piece of the DNA-Damage-Response Puzzle. QUANTUM BEAM SCIENCE 2019. [DOI: 10.3390/qubs3040023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Double-strand breaks of DNA may lead to discontinuous DNA and consequent loss of genetic information, which may result in mutations or, ultimately, carcinogenesis. To avoid such potentially serious situations, cells have evolved efficient DNA damage repair systems. It is thought that DNA-repair processes involve drastic alterations of chromatin and histone structures, but detection of these altered structures in DNA-damaged cells remains rare in the literature. Recently, synchrotron radiation circular dichroism (SRCD) spectroscopy, which can provide secondary structural information of proteins in solution, has identified structural alterations of histone proteins induced by DNA damage responses. In this review, these results and experimental procedures are discussed with the aim of facilitating further studies of the chromatin remodeling and DNA damage repair pathways using SRCD spectroscopy.
Collapse
|
40
|
House NC, Polleys EJ, Quasem I, De la Rosa Mejia M, Joyce CE, Takacsi-Nagy O, Krebs JE, Fuchs SM, Freudenreich CH. Distinct roles for S. cerevisiae H2A copies in recombination and repeat stability, with a role for H2A.1 threonine 126. eLife 2019; 8:53362. [PMID: 31804179 PMCID: PMC6927750 DOI: 10.7554/elife.53362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/26/2019] [Indexed: 01/14/2023] Open
Abstract
CAG/CTG trinuncleotide repeats are fragile sequences that when expanded form DNA secondary structures and cause human disease. We evaluated CAG/CTG repeat stability and repair outcomes in histone H2 mutants in S. cerevisiae. Although the two copies of H2A are nearly identical in amino acid sequence, CAG repeat stability depends on H2A copy 1 (H2A.1) but not copy 2 (H2A.2). H2A.1 promotes high-fidelity homologous recombination, sister chromatid recombination (SCR), and break-induced replication whereas H2A.2 does not share these functions. Both decreased SCR and the increase in CAG expansions were due to the unique Thr126 residue in H2A.1 and hta1Δ or hta1-T126A mutants were epistatic to deletion of the Polδ subunit Pol32, suggesting a role for H2A.1 in D-loop extension. We conclude that H2A.1 plays a greater repair-specific role compared to H2A.2 and may be a first step towards evolution of a repair-specific function for H2AX compared to H2A in mammalian cells.
Collapse
Affiliation(s)
- Nealia Cm House
- Department of Biology, Tufts University, Medford, United States
| | - Erica J Polleys
- Department of Biology, Tufts University, Medford, United States
| | | | | | - Cailin E Joyce
- Department of Biology, Tufts University, Medford, United States
| | | | - Jocelyn E Krebs
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, United States
| | - Stephen M Fuchs
- Department of Biology, Tufts University, Medford, United States
| | - Catherine H Freudenreich
- Department of Biology, Tufts University, Medford, United States.,Program in Genetics, Graduate School of Biomedical Sciences, Tufts University, Boston, United States
| |
Collapse
|
41
|
Proquin H, Jonkhout MCM, Jetten MJ, van Loveren H, de Kok TM, Briedé JJ. Transcriptome changes in undifferentiated Caco-2 cells exposed to food-grade titanium dioxide (E171): contribution of the nano- and micro- sized particles. Sci Rep 2019; 9:18287. [PMID: 31797963 PMCID: PMC6893026 DOI: 10.1038/s41598-019-54675-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
The food additive titanium dioxide (TiO2), or E171, is a white food colorant. Recent studies showed after E171 ingestion a significantly increased number of colorectal tumours in a colorectal cancer mouse model as well as inflammatory responses and dysregulation of the immune system in the intestine of rats. In the mouse colon, E171 induced gene expression changes related to oxidative stress, impairment of the immune system, activation of signalling and cancer-related processes. E171 comprises nanoparticles (NPs) and microparticles (MPs). Previous in vitro studies showed that E171, NPs and MPs induced oxidative stress responses, DNA damage and micronuclei formation. This study aimed to investigate the relative contribution of the NPs and MPs to effects of E171 at the transcriptome level in undifferentiated Caco-2 cells by genome wide microarray analysis. The results showed that E171, NPs, and MPs induce gene expression changes related to signalling, inflammation, immune system, transport and cancer. At the pathway level, metabolism of proteins with the insulin processing pathway and haemostasis were specific to E171 exposure. The gene expression changes associated with the immune system and inflammation induced by E171, MPs, and NPs suggest the creation of a favourable environment for colon cancer development.
Collapse
Affiliation(s)
- Héloïse Proquin
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Marloes C M Jonkhout
- Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, box 901 3000, Leuven, Belgium
| | - Marlon J Jetten
- Complex Tissue Regeneration (CTR), Institute for Technology-Inspired Regenerative Medicine (MERLN), Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Henk van Loveren
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Theo M de Kok
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Jacob J Briedé
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands.
| |
Collapse
|
42
|
Bayo J, Tran TA, Wang L, Peña-Llopis S, Das AK, Martinez ED. Jumonji Inhibitors Overcome Radioresistance in Cancer through Changes in H3K4 Methylation at Double-Strand Breaks. Cell Rep 2019; 25:1040-1050.e5. [PMID: 30355483 PMCID: PMC6245670 DOI: 10.1016/j.celrep.2018.09.081] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 03/17/2018] [Accepted: 09/25/2018] [Indexed: 01/10/2023] Open
Abstract
We have uncovered a role for Jumonji inhibitors in overcoming radioresistance through KDM5B inhibition. Pharmacological blockade of Jumonji demethy-lases with JIB-04 leads to specific accumulation of H3K4me3 at sites marked by γH2AX and impaired recruitment of DNA repair factors, preventing resolution of damage and resulting in robust sensitization to radiation therapy. In DNA-repair-proficient cancer cells, knockdown of the H3K4me3 demethylase KDM5B, but not other Jumonji enzymes, mimics pharmacological inhibition, and KDM5B overexpression rescues this phenotype and increases radioresistance. The H3K4me3 demethylase inhibitor PBIT also sensitizes cancer cells to radiation, while an H3K27me3 demethylase inhibitor does not. In vivo co-administration of radiation with JIB-04 significantly prolongs the survival of mice with tumors even long after cessation of treatment. In human patients, lung squamous cell carcinomas highly ex-pressing KDM5B respond poorly to radiation. Thus, we propose the use of Jumonji KDM inhibitors as potent radiosensitizers. Radioresistance is an obstacle to lung cancer cures. Bayo et al. reveal that JARID1B removes H3K4me3 marks at sites of DNA damage. Genetic or pharmacological inhibition of JARID1B robustly radiosensitizes cancers in vitro and in vivo through defects in DNA repair, providing a therapeutic option for radioresistant tumors.
Collapse
Affiliation(s)
- Juan Bayo
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA; Instituto de Investigaciones en Medicina Traslacional, CONICET, Universidad Austral, Argentina
| | - Tram Anh Tran
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lei Wang
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Samuel Peña-Llopis
- Division of Translational Oncology, Essen University Hospital, German Cancer Consortium (DKTK), Partner Site Essen and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Amit K Das
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Elisabeth D Martinez
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA; Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
43
|
De Vitis C, Corleone G, Salvati V, Ascenzi F, Pallocca M, De Nicola F, Fanciulli M, di Martino S, Bruschini S, Napoli C, Ricci A, Bassi M, Venuta F, Rendina EA, Ciliberto G, Mancini R. B4GALT1 Is a New Candidate to Maintain the Stemness of Lung Cancer Stem Cells. J Clin Med 2019; 8:E1928. [PMID: 31717588 PMCID: PMC6912435 DOI: 10.3390/jcm8111928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND According to the cancer stem cells (CSCs) hypothesis, a population of cancer cells with stem cell properties is responsible for tumor propagation, drug resistance, and disease recurrence. Study of the mechanisms responsible for lung CSCs propagation is expected to provide better understanding of cancer biology and new opportunities for therapy. METHODS The Lung Adenocarcinoma (LUAD) NCI-H460 cell line was grown either as 2D or as 3D cultures. Transcriptomic and genome-wide chromatin accessibility studies of 2D vs. 3D cultures were carried out using RNA-sequencing and Assay for Transposase Accessible Chromatin with high-throughput sequencing (ATAC-seq), respectively. Reverse transcription polymerase chain reaction (RT-PCR) was also carried out on RNA extracted from primary cultures derived from malignant pleural effusions to validate RNA-seq results. RESULTS RNA-seq and ATAC-seq data disentangled transcriptional and genome accessibility variability of 3D vs. 2D cultures in NCI-H460 cells. The examination of genomic landscape of genes upregulated in 3D vs. 2D cultures led to the identification of 2D cultures led to the identification of Beta-1,4-galactosyltranferase 1 (B4GALT1) as the top candidate. B4GALT1 as the top candidate. B4GALT1 was validated as a stemness factor, since its silencing caused strong inhibition of 3D spheroid formation. CONCLUSION Combined transcriptomic and chromatin accessibility study of 3D vs. 2D LUAD cultures led to the identification of B4GALT1 as a new factor involved in the propagation and maintenance of LUAD CSCs.
Collapse
Affiliation(s)
- Claudia De Vitis
- Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, “Sapienza” University of Rome, 00161 Rome, Italy; (C.D.V.); (R.M.)
| | - Giacomo Corleone
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Valentina Salvati
- Preclinical Models and New Therapeutic Agents Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Francesca Ascenzi
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Matteo Pallocca
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Francesca De Nicola
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Maurizio Fanciulli
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Simona di Martino
- Pathology Unit, IRCSS “Regina Elena” National Cancer Institute, 00144 Rome, Italy;
| | - Sara Bruschini
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Christian Napoli
- Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea Hospital, “Sapienza” University of Rome, 00189 Rome, Italy;
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine, Division of Pneumology, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy;
| | - Massimiliano Bassi
- Department of Thoracic Surgery, University of Rome Sapienza, 00161 Rome, Italy; (M.B.); (F.V.)
| | - Federico Venuta
- Department of Thoracic Surgery, University of Rome Sapienza, 00161 Rome, Italy; (M.B.); (F.V.)
| | - Erino Angelo Rendina
- Department of Thoracic Surgery, Sant’Andrea Hospital, “Sapienza” University of Rome, 00189 Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, “Sapienza” University of Rome, 00161 Rome, Italy; (C.D.V.); (R.M.)
| |
Collapse
|
44
|
Kim JH. Chromatin Remodeling and Epigenetic Regulation in Plant DNA Damage Repair. Int J Mol Sci 2019; 20:ijms20174093. [PMID: 31443358 PMCID: PMC6747262 DOI: 10.3390/ijms20174093] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/19/2022] Open
Abstract
DNA damage response (DDR) in eukaryotic cells is initiated in the chromatin context. DNA damage and repair depend on or have influence on the chromatin dynamics associated with genome stability. Epigenetic modifiers, such as chromatin remodelers, histone modifiers, DNA (de-)methylation enzymes, and noncoding RNAs regulate DDR signaling and DNA repair by affecting chromatin dynamics. In recent years, significant progress has been made in the understanding of plant DDR and DNA repair. SUPPRESSOR OF GAMMA RESPONSE1, RETINOBLASTOMA RELATED1 (RBR1)/E2FA, and NAC103 have been proven to be key players in the mediation of DDR signaling in plants, while plant-specific chromatin remodelers, such as DECREASED DNA METHYLATION1, contribute to chromatin dynamics for DNA repair. There is accumulating evidence that plant epigenetic modifiers are involved in DDR and DNA repair. In this review, I examine how DDR and DNA repair machineries are concertedly regulated in Arabidopsis thaliana by a variety of epigenetic modifiers directing chromatin remodeling and epigenetic modification. This review will aid in updating our knowledge on DDR and DNA repair in plants.
Collapse
Affiliation(s)
- Jin-Hong Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do 56212, Korea.
| |
Collapse
|
45
|
Horemans N, Spurgeon DJ, Lecomte-Pradines C, Saenen E, Bradshaw C, Oughton D, Rasnaca I, Kamstra JH, Adam-Guillermin C. Current evidence for a role of epigenetic mechanisms in response to ionizing radiation in an ecotoxicological context. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 251:469-483. [PMID: 31103007 DOI: 10.1016/j.envpol.2019.04.125] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/14/2019] [Accepted: 04/27/2019] [Indexed: 05/22/2023]
Abstract
The issue of potential long-term or hereditary effects for both humans and wildlife exposed to low doses (or dose rates) of ionising radiation is a major concern. Chronic exposure to ionising radiation, defined as an exposure over a large fraction of the organism's lifespan or even over several generations, can possibly have consequences in the progeny. Recent work has begun to show that epigenetics plays an important role in adaptation of organisms challenged to environmental stimulae. Changes to so-called epigenetic marks such as histone modifications, DNA methylation and non-coding RNAs result in altered transcriptomes and proteomes, without directly changing the DNA sequence. Moreover, some of these environmentally-induced epigenetic changes tend to persist over generations, and thus, epigenetic modifications are regarded as the conduits for environmental influence on the genome. Here, we review the current knowledge of possible involvement of epigenetics in the cascade of responses resulting from environmental exposure to ionising radiation. In addition, from a comparison of lab and field obtained data, we investigate evidence on radiation-induced changes in the epigenome and in particular the total or locus specific levels of DNA methylation. The challenges for future research and possible use of changes as an early warning (biomarker) of radiosensitivity and individual exposure is discussed. Such a biomarker could be used to detect and better understand the mechanisms of toxic action and inter/intra-species susceptibility to radiation within an environmental risk assessment and management context.
Collapse
Affiliation(s)
- Nele Horemans
- Belgian Nuclear Research Centre, Boeretang 200, B-2400, Mol, Belgium; Centre for Environmental Research, University of Hasselt, Agoralaan, 3590, Diepenbeek, Belgium.
| | - David J Spurgeon
- Centre for Ecology and Hydrology, MacLean Building, Benson Lane, Wallingford, Oxon, OX10 8BB, UK
| | - Catherine Lecomte-Pradines
- Institut de Radioprotection et de Sûreté Nucléaire, PSE-ENV/SRTE/LECO, Cadarache, Saint Paul Lez Durance, France
| | - Eline Saenen
- Belgian Nuclear Research Centre, Boeretang 200, B-2400, Mol, Belgium
| | - Clare Bradshaw
- Department of Ecology, Environment and Plant Sciences, Stockholm University, 106 91, Stockholm, Sweden
| | - Deborah Oughton
- Centre for Environmental Radioactivity (CERAD), Norwegian University of Life Sciences, 1430, Aas, Norway
| | - Ilze Rasnaca
- Centre for Ecology and Hydrology, MacLean Building, Benson Lane, Wallingford, Oxon, OX10 8BB, UK
| | - Jorke H Kamstra
- Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Christelle Adam-Guillermin
- Institut de Radioprotection et de Sûreté Nucléaire, PSE-SANTE, Cadarache, Saint Paul Lez Durance, France
| |
Collapse
|
46
|
Zhang Q, Thakur C, Shi J, Sun J, Fu Y, Stemmer P, Chen F. New discoveries of mdig in the epigenetic regulation of cancers. Semin Cancer Biol 2019; 57:27-35. [PMID: 31276784 PMCID: PMC6844078 DOI: 10.1016/j.semcancer.2019.06.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 06/17/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022]
Abstract
Mineral dust-induced gene (mdig) encodes a member of the evolutionarily conserved JmjC family proteins that play fundamental roles in regulating chromatin-based processes as well as transcription of the genes in eukaryotic cells. This gene is also named as myc-induced nuclear antigen 53 (MINA), nucleolar protein 52 (NO52) and ribosomal oxygenase 2 (RIOX2). Increased expression of mdig had been noted in a number of human cancers, esp. lung cancer. Emerging evidence suggests that the oncogenic activity of mdig is most likely achieved through its regulation on the demethylation of histone proteins, despite it lacks the structural identities of the demethylases. Here, we discuss the latest discoveries on the characteristics of the mdig protein and its roles in a wide variety of normal and carcinogenic processes. We will also provide perspectives on how mdig is involved in the maintenance and differentiation of the embryonic stem cells, somatic stem cells and cancer stem cells.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Chitra Thakur
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Junwei Shi
- Nantong Pulmonary Hospital, 500 Yonghe Road, Gangzha Qu, Nantong, 226011, Jiangsu Province, China
| | - Jiaying Sun
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Yao Fu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Paul Stemmer
- Institute of Environmental Health Sciences, School of Medicine, Wayne State University, 6135 Woodward Avenue, Detroit, MI, 48202, USA
| | - Fei Chen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA.
| |
Collapse
|
47
|
Collemare J, Seidl MF. Chromatin-dependent regulation of secondary metabolite biosynthesis in fungi: is the picture complete? FEMS Microbiol Rev 2019; 43:591-607. [PMID: 31301226 PMCID: PMC8038932 DOI: 10.1093/femsre/fuz018] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 06/18/2019] [Indexed: 01/07/2023] Open
Abstract
Fungal secondary metabolites are small molecules that exhibit diverse biological activities exploited in medicine, industry and agriculture. Their biosynthesis is governed by co-expressed genes that often co-localize in gene clusters. Most of these secondary metabolite gene clusters are inactive under laboratory conditions, which is due to a tight transcriptional regulation. Modifications of chromatin, the complex of DNA and histone proteins influencing DNA accessibility, play an important role in this regulation. However, tinkering with well-characterised chemical and genetic modifications that affect chromatin alters the expression of only few biosynthetic gene clusters, and thus the regulation of the vast majority of biosynthetic pathways remains enigmatic. In the past, attempts to activate silent gene clusters in fungi mainly focused on histone acetylation and methylation, while in other eukaryotes many other post-translational modifications are involved in transcription regulation. Thus, how chromatin regulates the expression of gene clusters remains a largely unexplored research field. In this review, we argue that focusing on only few well-characterised chromatin modifications is significantly hampering our understanding of the chromatin-based regulation of biosynthetic gene clusters. Research on underexplored chromatin modifications and on the interplay between different modifications is timely to fully explore the largely untapped reservoir of fungal secondary metabolites.
Collapse
Affiliation(s)
| | - Michael F Seidl
- Corresponding author: Theoretical Biology and Bioinformatics, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands. E-mail: ; Present address: Theoretical Biology and Bioinformatics, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| |
Collapse
|
48
|
Is mitochondrial DNA profiling predictive for athletic performance? Mitochondrion 2019; 47:125-138. [PMID: 31228565 DOI: 10.1016/j.mito.2019.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 06/03/2019] [Accepted: 06/17/2019] [Indexed: 11/20/2022]
Abstract
Mitochondrial DNA encodes some proteins of the oxidative phosphorylation enzymatic complex, playing an important role in aerobic ATP production; therefore, it can contribute to the ability to respond to endurance exercise training. The accumulation of mitochondrial mutations and the migratory processes of populations have given a great contribution to the development of haplogroups with a different distribution in the world. Several studies have shown the important role of gene polymorphisms in aerobic performance. In this review, some mitochondrial haplogroups and multiple rare alleles were taken into consideration and could be linked to the athlete's physical performance of different ethnic groups.
Collapse
|
49
|
Efimova EV, Appelbe OK, Ricco N, Lee SSY, Liu Y, Wolfgeher DJ, Collins TN, Flor AC, Ramamurthy A, Warrington S, Bindokas VP, Kron SJ. O-GlcNAcylation Enhances Double-Strand Break Repair, Promotes Cancer Cell Proliferation, and Prevents Therapy-Induced Senescence in Irradiated Tumors. Mol Cancer Res 2019; 17:1338-1350. [PMID: 30885991 DOI: 10.1158/1541-7786.mcr-18-1025] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/04/2019] [Accepted: 03/14/2019] [Indexed: 12/16/2022]
Abstract
The metabolic reprogramming associated with characteristic increases in glucose and glutamine metabolism in advanced cancer is often ascribed to answering a higher demand for metabolic intermediates required for rapid tumor cell growth. Instead, recent discoveries have pointed to an alternative role for glucose and glutamine metabolites as cofactors for chromatin modifiers and other protein posttranslational modification enzymes in cancer cells. Beyond epigenetic mechanisms regulating gene expression, many chromatin modifiers also modulate DNA repair, raising the question whether cancer metabolic reprogramming may mediate resistance to genotoxic therapy and genomic instability. Our prior work had implicated N-acetyl-glucosamine (GlcNAc) formation by the hexosamine biosynthetic pathway (HBP) and resulting protein O-GlcNAcylation as a common means by which increased glucose and glutamine metabolism can drive double-strand break (DSB) repair and resistance to therapy-induced senescence in cancer cells. We have examined the effects of modulating O-GlcNAcylation on the DNA damage response (DDR) in MCF7 human mammary carcinoma in vitro and in xenograft tumors. Proteomic profiling revealed deregulated DDR pathways in cells with altered O-GlcNAcylation. Promoting protein O-GlcNAc modification by targeting O-GlcNAcase or simply treating animals with GlcNAc protected tumor xenografts against radiation. In turn, suppressing protein O-GlcNAcylation by blocking O-GlcNAc transferase activity led to delayed DSB repair, reduced cell proliferation, and increased cell senescence in vivo. Taken together, these findings confirm critical connections between cancer metabolic reprogramming, DDR, and senescence and provide a rationale to evaluate agents targeting O-GlcNAcylation in patients as a means to restore tumor sensitivity to radiotherapy. IMPLICATIONS: The finding that the HBP, via its impact on protein O-GlcNAcylation, is a key determinant of the DDR in cancer provides a mechanistic link between metabolic reprogramming, genomic instability, and therapeutic response and suggests novel therapeutic approaches for tumor radiosensitization.
Collapse
Affiliation(s)
- Elena V Efimova
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Oliver K Appelbe
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Natalia Ricco
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Steve S-Y Lee
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Yue Liu
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Donald J Wolfgeher
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Tamica N Collins
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Amy C Flor
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Aishwarya Ramamurthy
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Sara Warrington
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Vytautas P Bindokas
- Integrated Light Microscopy Facility, The University of Chicago, Chicago, IL
| | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL. .,Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL
| |
Collapse
|
50
|
Cogné B, Ehresmann S, Beauregard-Lacroix E, Rousseau J, Besnard T, Garcia T, Petrovski S, Avni S, McWalter K, Blackburn PR, Sanders SJ, Uguen K, Harris J, Cohen JS, Blyth M, Lehman A, Berg J, Li MH, Kini U, Joss S, von der Lippe C, Gordon CT, Humberson JB, Robak L, Scott DA, Sutton VR, Skraban CM, Johnston JJ, Poduri A, Nordenskjöld M, Shashi V, Gerkes EH, Bongers EM, Gilissen C, Zarate YA, Kvarnung M, Lally KP, Kulch PA, Daniels B, Hernandez-Garcia A, Stong N, McGaughran J, Retterer K, Tveten K, Sullivan J, Geisheker MR, Stray-Pedersen A, Tarpinian JM, Klee EW, Sapp JC, Zyskind J, Holla ØL, Bedoukian E, Filippini F, Guimier A, Picard A, Busk ØL, Punetha J, Pfundt R, Lindstrand A, Nordgren A, Kalb F, Desai M, Ebanks AH, Jhangiani SN, Dewan T, Coban Akdemir ZH, Telegrafi A, Zackai EH, Begtrup A, Song X, Toutain A, Wentzensen IM, Odent S, Bonneau D, Latypova X, Deb W, Redon S, Bilan F, Legendre M, Troyer C, Whitlock K, Caluseriu O, Murphree MI, Pichurin PN, Agre K, Gavrilova R, Rinne T, Park M, Shain C, Heinzen EL, Xiao R, Amiel J, Lyonnet S, Isidor B, Biesecker LG, Lowenstein D, Posey JE, Denommé-Pichon AS, Férec C, et alCogné B, Ehresmann S, Beauregard-Lacroix E, Rousseau J, Besnard T, Garcia T, Petrovski S, Avni S, McWalter K, Blackburn PR, Sanders SJ, Uguen K, Harris J, Cohen JS, Blyth M, Lehman A, Berg J, Li MH, Kini U, Joss S, von der Lippe C, Gordon CT, Humberson JB, Robak L, Scott DA, Sutton VR, Skraban CM, Johnston JJ, Poduri A, Nordenskjöld M, Shashi V, Gerkes EH, Bongers EM, Gilissen C, Zarate YA, Kvarnung M, Lally KP, Kulch PA, Daniels B, Hernandez-Garcia A, Stong N, McGaughran J, Retterer K, Tveten K, Sullivan J, Geisheker MR, Stray-Pedersen A, Tarpinian JM, Klee EW, Sapp JC, Zyskind J, Holla ØL, Bedoukian E, Filippini F, Guimier A, Picard A, Busk ØL, Punetha J, Pfundt R, Lindstrand A, Nordgren A, Kalb F, Desai M, Ebanks AH, Jhangiani SN, Dewan T, Coban Akdemir ZH, Telegrafi A, Zackai EH, Begtrup A, Song X, Toutain A, Wentzensen IM, Odent S, Bonneau D, Latypova X, Deb W, Redon S, Bilan F, Legendre M, Troyer C, Whitlock K, Caluseriu O, Murphree MI, Pichurin PN, Agre K, Gavrilova R, Rinne T, Park M, Shain C, Heinzen EL, Xiao R, Amiel J, Lyonnet S, Isidor B, Biesecker LG, Lowenstein D, Posey JE, Denommé-Pichon AS, Férec C, Yang XJ, Rosenfeld JA, Gilbert-Dussardier B, Audebert-Bellanger S, Redon R, Stessman HA, Nellaker C, Yang Y, Lupski JR, Goldstein DB, Eichler EE, Bolduc F, Bézieau S, Küry S, Campeau PM, Küry S, Campeau PM. Missense Variants in the Histone Acetyltransferase Complex Component Gene TRRAP Cause Autism and Syndromic Intellectual Disability. Am J Hum Genet 2019; 104:530-541. [PMID: 30827496 DOI: 10.1016/j.ajhg.2019.01.010] [Show More Authors] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
Acetylation of the lysine residues in histones and other DNA-binding proteins plays a major role in regulation of eukaryotic gene expression. This process is controlled by histone acetyltransferases (HATs/KATs) found in multiprotein complexes that are recruited to chromatin by the scaffolding subunit transformation/transcription domain-associated protein (TRRAP). TRRAP is evolutionarily conserved and is among the top five genes intolerant to missense variation. Through an international collaboration, 17 distinct de novo or apparently de novo variants were identified in TRRAP in 24 individuals. A strong genotype-phenotype correlation was observed with two distinct clinical spectra. The first is a complex, multi-systemic syndrome associated with various malformations of the brain, heart, kidneys, and genitourinary system and characterized by a wide range of intellectual functioning; a number of affected individuals have intellectual disability (ID) and markedly impaired basic life functions. Individuals with this phenotype had missense variants clustering around the c.3127G>A p.(Ala1043Thr) variant identified in five individuals. The second spectrum manifested with autism spectrum disorder (ASD) and/or ID and epilepsy. Facial dysmorphism was seen in both groups and included upslanted palpebral fissures, epicanthus, telecanthus, a wide nasal bridge and ridge, a broad and smooth philtrum, and a thin upper lip. RNA sequencing analysis of skin fibroblasts derived from affected individuals skin fibroblasts showed significant changes in the expression of several genes implicated in neuronal function and ion transport. Thus, we describe here the clinical spectrum associated with TRRAP pathogenic missense variants, and we suggest a genotype-phenotype correlation useful for clinical evaluation of the pathogenicity of the variants.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Sébastien Küry
- Centre Hospitalier Universitaire de Nantes, Service de Génétique Médicale, 9 quai Moncousu, 44093 Nantes, France; INSERM, CNRS, UNIV Nantes, l'institut du thorax, 44007 Nantes, France.
| | - Philippe M Campeau
- Centre Hospitalier Universitaire Sainte-Justine Research Centre, University of Montreal, Montreal, QC H3T 1C5, Canada; Department of Pediatrics, University of Montreal, Montreal, QC H3T1J4, Canada.
| |
Collapse
|