1
|
Yang Y, Yuan H, Yao B, Zhao S, Wang X, Xu L, Zhang L. Genetic Adaptations of the Tibetan Pig to High-Altitude Hypoxia on the Qinghai-Tibet Plateau. Int J Mol Sci 2024; 25:11303. [PMID: 39457085 PMCID: PMC11508817 DOI: 10.3390/ijms252011303] [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: 08/19/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The Tibetan Plateau's distinctive high-altitude environment, marked by extreme cold and reduced oxygen levels, presents considerable survival challenges for both humans and mammals. Natural selection has led to the accumulation of adaptive mutations in Tibetan pigs, enabling them to develop distinctive adaptive phenotypes. Here, we aim to uncover the genetic mechanisms underlying the adaptation of Tibetan pigs to high-altitude hypoxia. Therefore, we conducted a systematic analysis of 140 whole-genome sequencing (WGS) data points from different representing pig populations. Our analysis identified a total of 27,614,561 mutations, including 22,386,319 single-nucleotide variants (SNVs) and 5,228,242 insertions/deletions (INDELs, size < 50 bp). A total of 11% (2,678,569) of the SNVs were newly identified in our project, significantly expanding the dataset of genetic variants in Tibetan pigs. Compared to other pig breeds, Tibetan pigs are uniquely adapted to high-altitude environments, exhibiting the highest genetic diversity and the lowest inbreeding coefficient. Employing the composite of multiple signals (CMS) method, we scanned the genome-wide Darwinian positive selection signals and identified 32,499 Tibetan pig positively selected SNVs (TBPSSs) and 129 selected genes (TBPSGs), including 213 newly discovered genes. Notably, we identified eight genes (PHACTR1, SFI1, EPM2A, SLC30A7, NKAIN2, TNNI3K, and PLIN2) with strong nature selection signals. They are likely to improve cardiorespiratory function and fat metabolism to help Tibetan pigs become adapted to the high-altitude environment. These findings provide new insights into the genetic mechanisms of high-altitude adaptation and the adaptive phenotypes of Tibetan pigs.
Collapse
Affiliation(s)
- Yanan Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (H.Y.); (B.Y.); (S.Z.); (X.W.); (L.X.); (L.Z.)
| | | | | | | | | | | | | |
Collapse
|
2
|
Fang S, Zhang H, Long H, Zhang D, Chen H, Yang X, Pan H, Pan X, Liu D, E G. Phylogenetic Relations and High-Altitude Adaptation in Wild Boar ( Sus scrofa), Identified Using Genome-Wide Data. Animals (Basel) 2024; 14:2984. [PMID: 39457914 PMCID: PMC11503864 DOI: 10.3390/ani14202984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
The Qinghai-Tibet Plateau (QTP) wild boar is an excellent model for investigating high-altitude adaptation. In this study, we analyzed genome-wide data from 93 wild boars compiled from various studies worldwide, including the QTP, southern and northern regions of China, Europe, Northeast Asia, and Southeast Asia, to explore their phylogenetic patterns and high-altitude adaptation based on genome-wide selection signal analysis and run of homozygosity (ROH) estimation. The findings demonstrate the alignment between the phylogenetic associations among wild boars and their geographical location. An ADMIXTURE analysis indicated a relatively close genetic relationship between QTP and southern Chinese wild boars. Analyses of the fixation index and cross-population extended haplotype homozygosity between populations revealed 295 candidate genes (CDGs) associated with high-altitude adaptation, such as TSC2, TELO2, SLC5A1, and SLC5A4. These CDGs were significantly overrepresented in pathways such as the mammalian target of rapamycin signaling and Fanconi anemia pathways. In addition, 39 ROH islands and numerous selective CDGs (e.g., SLC5A1, SLC5A4, and VCP), which are implicated in glucose metabolism and mitochondrial function, were discovered in QTP wild boars. This study not only assessed the phylogenetic history of QTP wild boars but also advanced our comprehension of the genetic mechanisms underlying the adaptation of wild boars to high altitudes.
Collapse
Affiliation(s)
- Shiyong Fang
- College of Animal Science and Technology, Southwest University, Chongqing 400716, China; (S.F.); (H.Z.); (H.L.)
| | - Haoyuan Zhang
- College of Animal Science and Technology, Southwest University, Chongqing 400716, China; (S.F.); (H.Z.); (H.L.)
| | - Haoyuan Long
- College of Animal Science and Technology, Southwest University, Chongqing 400716, China; (S.F.); (H.Z.); (H.L.)
| | - Dongjie Zhang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China;
| | - Hongyue Chen
- Chongqing Animal Husbandry Technology Extension Station, Chongqing 401121, China;
| | - Xiuqin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China;
| | - Hongmei Pan
- Chongqing Academy of Animal Sciences, Chongqing 408599, China;
| | - Xiao Pan
- Chongqing Hechuan Animal Husbandry Station, Chongqing 401520, China;
| | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China;
| | - Guangxin E
- College of Animal Science and Technology, Southwest University, Chongqing 400716, China; (S.F.); (H.Z.); (H.L.)
| |
Collapse
|
3
|
Wang L, Liu WQ, Du J, Li M, Wu RF, Li M. Comparative DNA methylation reveals epigenetic adaptation to high altitude in snub-nosed monkeys. Zool Res 2024; 45:1013-1026. [PMID: 39147716 PMCID: PMC11491775 DOI: 10.24272/j.issn.2095-8137.2024.050] [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: 02/17/2024] [Accepted: 04/25/2024] [Indexed: 08/17/2024] Open
Abstract
DNA methylation plays a crucial role in environmental adaptations. Here, using whole-genome bisulfite sequencing, we generated comprehensive genome-wide DNA methylation profiles for the high-altitude Yunnan snub-nosed monkey ( Rhinopithecus bieti) and the closely related golden snub-nosed monkey ( R. roxellana). Our findings indicated a slight increase in overall DNA methylation levels in golden snub-nosed monkeys compared to Yunnan snub-nosed monkeys, suggesting a higher prevalence of hypermethylated genomic regions in the former. Comparative genomic methylation analysis demonstrated that genes associated with differentially methylated regions were involved in membrane fusion, vesicular formation and trafficking, hemoglobin function, cell cycle regulation, and neuronal differentiation. These results suggest that the high-altitude-related epigenetic modifications are extensive, involving a complete adaptation process from the inhibition of single Ca 2+ channel proteins to multiple proteins collaboratively enhancing vesicular function or inhibiting cell differentiation and proliferation. Functional assays demonstrated that overexpression or down-regulation of candidate genes, such as SNX10, TIMELESS, and CACYBP, influenced cell viability under stress conditions. Overall, this research suggests that comparing DNA methylation across closely related species can identify novel candidate genomic regions and genes associated with local adaptations, thereby deepening our understanding of the mechanisms underlying environmental adaptations.
Collapse
Affiliation(s)
- Ling Wang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei-Qiang Liu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juan Du
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Meng Li
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rui-Feng Wu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ming Li
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| |
Collapse
|
4
|
Wang Z, Liu Y, Wang H, Roy A, Liu H, Han F, Zhang X, Lu Q. Genome and transcriptome of Ips nitidus provide insights into high-altitude hypoxia adaptation and symbiosis. iScience 2023; 26:107793. [PMID: 37731610 PMCID: PMC10507238 DOI: 10.1016/j.isci.2023.107793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/15/2023] [Accepted: 08/29/2023] [Indexed: 09/22/2023] Open
Abstract
Ips nitidus is a well-known conifer pest that has contributed significantly to spruce forest disturbance in the Qinghai-Tibet Plateau and seriously threatens the ecological balance of these areas. We report a chromosome-level genome of I. nitidus determined by PacBio and Hi-C technology. Phylogenetic inference showed that it diverged from the common ancestor of I. typographus ∼2.27 mya. Gene family expansion in I. nitidus was characterized by DNA damage repair and energy metabolism, which may facilitate adaptation to high-altitude hypoxia. Interestingly, differential gene expression analysis revealed upregulated genes associated with high-altitude hypoxia adaptation and downregulated genes associated with detoxification after feeding and tunneling in fungal symbiont Ophiostoma bicolor-colonized substrates. Our findings provide evidence of the potential adaptability of I. nitidus to conifer host, high-altitude hypoxia and insight into how fungal symbiont assist in this process. This study enhances our understanding of insect adaptation, symbiosis, and pest management.
Collapse
Affiliation(s)
- Zheng Wang
- Key Laboratory of Forest Protection of National Forestry and Grassland Administration, Ecology and Nature Conservation Institute, Chinese Academy of Forestry, Beijing 100091, China
- Shandong Research Center for Forestry Harmful Biological Control Engineering and Technology, College of Plant Protection, Shandong Agricultural University, Tai’an 271018, China
| | - Ya Liu
- Key Laboratory of Forest Protection of National Forestry and Grassland Administration, Ecology and Nature Conservation Institute, Chinese Academy of Forestry, Beijing 100091, China
| | - Huimin Wang
- Key Laboratory of Forest Protection of National Forestry and Grassland Administration, Ecology and Nature Conservation Institute, Chinese Academy of Forestry, Beijing 100091, China
| | - Amit Roy
- Faculty of Forestry and Wood Sciences, EXTEMIT-K and EVA.4.0 Unit, Czech University of Life Sciences, Kamýcká 1176, Prague 6, 165 00 Suchdol, Czech Republic
| | - Huixiang Liu
- Shandong Research Center for Forestry Harmful Biological Control Engineering and Technology, College of Plant Protection, Shandong Agricultural University, Tai’an 271018, China
| | | | - Xingyao Zhang
- Key Laboratory of Forest Protection of National Forestry and Grassland Administration, Ecology and Nature Conservation Institute, Chinese Academy of Forestry, Beijing 100091, China
| | - Quan Lu
- Key Laboratory of Forest Protection of National Forestry and Grassland Administration, Ecology and Nature Conservation Institute, Chinese Academy of Forestry, Beijing 100091, China
| |
Collapse
|
5
|
Yu X, Wei P, Zhao S, Chen Z, Li X, Zhang W, Liu C, Yang Y, Li X, Liu X. Population transcriptomics uncover the relative roles of positive selection and differential expression in Batrachium bungei adaptation to the Qinghai-Tibetan plateau. PLANT CELL REPORTS 2023; 42:879-893. [PMID: 36973418 DOI: 10.1007/s00299-023-03005-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/14/2023] [Indexed: 05/06/2023]
Abstract
KEY MESSAGE Positive selection genes are related to metabolism, while differentially expressed genes are related to photosynthesis, suggesting that genetic adaptation and expression regulation may play independent roles in different gene classes. Genome-wide investigation of the molecular mechanisms for high-altitude adaptation is an intriguing topic in evolutionary biology. The Qinghai-Tibet Plateau (QTP) with its extremely variable environments is an ideal site for studying high-altitude adaptation. Here, we used transcriptome data of 100 individuals from 20 populations collected from various altitudes on the QTP to investigate the adaptive mechanisms of the aquatic plant Batrachium bungei at both the genetic and transcriptional level. To explore genes and biological pathways that may contribute to QTP adaptation, we employed a two-step approach, in which we identified positively selected genes and differentially expressed genes using the landscape genomic and differential expression approaches. The positive selection analysis showed that genes involved in metabolic regulation played a crucial role in B. bungei adaptation to the extreme environments of the QTP, especially intense ultraviolet radiation. Altitude-based differential expression analysis suggested that B. bungei could increase the rate of energy dissipation or reduce the efficiency of light energy absorption by down regulating the expression of photosynthesis-related genes to adapt to the strong ultraviolet radiation. Weighted gene co-expression network analysis identified ribosomal genes as hubs of altitude adaptation in B. bungei. Only a small part of genes (about 10%) overlapped between positively selected genes and differentially expressed genes in B. bungei, suggesting that genetic adaptation and gene expression regulation might play relatively independent roles in different categories of functional genes. Taken together, this study enriches our understanding of the high-altitude adaptation mechanism of B. bungei on the QTP.
Collapse
Affiliation(s)
- Xiaolei Yu
- State Key Laboratory of Hybrid Rice, Laboratory of Plant Systematics and Evolutionary Biology, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Pei Wei
- State Key Laboratory of Hybrid Rice, Laboratory of Plant Systematics and Evolutionary Biology, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Shuqi Zhao
- State Key Laboratory of Hybrid Rice, Laboratory of Plant Systematics and Evolutionary Biology, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Zhuyifu Chen
- State Key Laboratory of Hybrid Rice, Laboratory of Plant Systematics and Evolutionary Biology, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Xinzhong Li
- Laboratory of Extreme Environmental Biological Resources and Adaptive Evolution, Research Center for Ecology, School of Sciences, Tibet University, Lhasa, 850000, Tibet, China
| | - Wencai Zhang
- Laboratory of Extreme Environmental Biological Resources and Adaptive Evolution, Research Center for Ecology, School of Sciences, Tibet University, Lhasa, 850000, Tibet, China
| | - Chenlai Liu
- State Key Laboratory of Hybrid Rice, Laboratory of Plant Systematics and Evolutionary Biology, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Yujiao Yang
- State Key Laboratory of Hybrid Rice, Laboratory of Plant Systematics and Evolutionary Biology, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Xiaoyan Li
- Biology Experimental Teaching Center, School of Life Science, Wuhan University, Wuhan, 430072, Hubei, China.
| | - Xing Liu
- State Key Laboratory of Hybrid Rice, Laboratory of Plant Systematics and Evolutionary Biology, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China.
- Laboratory of Extreme Environmental Biological Resources and Adaptive Evolution, Research Center for Ecology, School of Sciences, Tibet University, Lhasa, 850000, Tibet, China.
| |
Collapse
|
6
|
Mudambi S, Fitzgerald M, Pera P, Washington D, Chamberlain S, Fidrus E, Hegedűs C, Remenyik E, Shafirstein G, Bellnier D, Paragh G. KDM1A inhibition increases UVA toxicity and enhances photodynamic therapy efficacy. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2023; 39:226-234. [PMID: 35968606 PMCID: PMC10089661 DOI: 10.1111/phpp.12826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 07/25/2022] [Accepted: 08/08/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Lysine-specific histone demethylase 1 (KDM1A/LSD1) regulates multiple cellular functions, including cellular proliferation, differentiation, and DNA repair. KDM1A is overexpressed in squamous cell carcinoma of the skin and inhibition of KDM1A can suppress cutaneous carcinogenesis. Despite the role of KDM1A in skin and DNA repair, the effect of KDM1A inhibition on cellular ultraviolet (UV) response has not been studied. METHODS The ability of KDM1A inhibitor bizine to modify cell death after UVA and UVB exposure was tested in normal human keratinocytes and melanocytes, HaCaT, and FaDu cell lines. KDM1A was also downregulated using shRNA and inhibited by phenelzine in HaCaT and FaDu cells to confirm the role of KDM1A in UVA response. In addition, cellular reactive oxygen species (ROS) changes were assessed by a lipid-soluble fluorescent indicator of lipid oxidation, and ROS-related gene regulation using qPCR. During photodynamic therapy (PDT) studies HaCaT and FaDu cells were treated with aminolaevulinic acid (5-ALA) or HPPH (2-[1-hexyloxyethyl]-2-devinyl pyropheophorbide-a) sodium and irradiated with 0-8 J/cm2 red LED light. RESULTS KDM1A inhibition sensitized cells to UVA radiation-induced cell death but not to UVB. KDM1A inhibition increased ROS generation as detected by increased lipid peroxidation and the upregulation of ROS-responsive genes. The effectiveness of both ALA and HPPH PDT significantly improved in vitro in HaCaT and FaDu cells after KDM1A inhibition. CONCLUSION KDM1A is a regulator of cellular UV response and KDM1A inhibition can improve PDT efficacy.
Collapse
Affiliation(s)
- Shaila Mudambi
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
- Department of Dermatology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
| | - Megan Fitzgerald
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
- Department of Dermatology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
| | - Paula Pera
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
- Department of Dermatology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
| | - Deschana Washington
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
| | - Sarah Chamberlain
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
- Photodynamic Therapy Center, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
| | - Eszter Fidrus
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei korut 98, Debrecen, Hungary, H-4032
| | - Csaba Hegedűs
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei korut 98, Debrecen, Hungary, H-4032
| | - Eva Remenyik
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei korut 98, Debrecen, Hungary, H-4032
| | - Gal Shafirstein
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
- Photodynamic Therapy Center, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
| | - David Bellnier
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
- Photodynamic Therapy Center, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
| | - Gyorgy Paragh
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
- Department of Dermatology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, United States 14263
| |
Collapse
|
7
|
Whitley MJ, Suwanpradid J, Lai C, Jiang SW, Cook JL, Zelac DE, Rudolph R, Corcoran DL, Degan S, Spasojevic I, Levinson H, Erdmann D, Reid C, Zhang JY, Robson SC, Healy E, Havran WL, MacLeod AS. ENTPD1 (CD39) Expression Inhibits UVR-Induced DNA Damage Repair through Purinergic Signaling and Is Associated with Metastasis in Human Cutaneous Squamous Cell Carcinoma. J Invest Dermatol 2021; 141:2509-2520. [PMID: 33848530 DOI: 10.1016/j.jid.2021.02.753] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/21/2021] [Accepted: 02/07/2021] [Indexed: 12/24/2022]
Abstract
UVR and immunosuppression are major risk factors for cutaneous squamous cell carcinoma (cSCC). Regulatory T cells promote cSCC carcinogenesis, and in other solid tumors, infiltrating regulatory T cells and CD8+ T cells express ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1) (also known as CD39), an ectoenzyme that catalyzes the rate-limiting step in converting extracellular adenosine triphosphate (ATP) to extracellular adenosine (ADO). We previously showed that extracellular purine nucleotides influence DNA damage repair. In this study, we investigate whether DNA damage repair is modulated through purinergic signaling in cSCC. We found increased ENTPD1 expression on T cells within cSCCs when compared with the expression on T cells from blood or nonlesional skin, and accordingly, concentrations of derivative extracellular adenosine diphosphate (ADP), adenosine monophosphate (AMP), and ADO are increased in tumors compared with those in normal skin. Importantly, ENTPD1 expression is significantly higher in human cSCCs that metastasize than in those that are nonmetastatic. We also identify in a mouse model that ENTPD1 expression is induced by UVR in an IL-27-dependent manner. Finally, increased extracellular ADO is shown to downregulate the expression of NAP1L2, a nucleosome assembly protein we show to be important for DNA damage repair secondary to UVR. Together, these data suggest a role for ENTPD1 expression on skin-resident T cells to regulate DNA damage repair through purinergic signaling to promote skin carcinogenesis and metastasis.
Collapse
Affiliation(s)
- Melodi Javid Whitley
- Department of Duke Dermatology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jutamas Suwanpradid
- Department of Duke Dermatology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Chester Lai
- Dermatopharmacology, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; Department of Dermatology, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Simon W Jiang
- Department of Duke Dermatology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jonathan L Cook
- Department of Duke Dermatology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Daniel E Zelac
- Department of Dermatology and Mohs Surgery, Scripps Clinic, La Jolla, California, USA
| | - Ross Rudolph
- Division of Plastic Surgery, Scripps Clinic, San Diego, California, USA; Division of Plastic Surgery, University of California San Diego, San Diego, California, USA
| | - David L Corcoran
- Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, North Carolina, USA; Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Simone Degan
- Department of Duke Dermatology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA; PK/PD Core Lab, Duke Cancer Institute, Durham, North Carolina, USA
| | - Howard Levinson
- Division of Plastic, Maxillofacial, and Oral Surgery, Duke Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Detlev Erdmann
- Division of Plastic, Maxillofacial, and Oral Surgery, Duke Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Claire Reid
- Dermatopharmacology, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; Department of Dermatology, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Jennifer Y Zhang
- Department of Duke Dermatology, Duke University School of Medicine, Durham, North Carolina, USA; Pinnell Center for Investigative Dermatology, Department of Duke Dermatology, Duke University School of Medicine, Durham, North Carolina, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Simon C Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, Massachusetts, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Boston, Massachusetts, USA
| | - Eugene Healy
- Dermatopharmacology, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; Department of Dermatology, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Wendy L Havran
- Department of Immunology and Microbiology, The Scripps Research Institute, San Diego, California, USA
| | - Amanda S MacLeod
- Department of Duke Dermatology, Duke University School of Medicine, Durham, North Carolina, USA; Pinnell Center for Investigative Dermatology, Department of Duke Dermatology, Duke University School of Medicine, Durham, North Carolina, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA; Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA.
| |
Collapse
|
8
|
Epigenetic modification and a role for the E3 ligase RNF40 in cancer development and metastasis. Oncogene 2020; 40:465-474. [PMID: 33199825 PMCID: PMC7819849 DOI: 10.1038/s41388-020-01556-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/15/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022]
Abstract
RNF40 (OMIM: 607700) is a really interesting new gene (RING) finger E3 ubiquitin ligase containing multiple coiled-coil domains and a C-terminal RING finger motif, which engage in protein–DNA and protein–protein interactions. RNF40 encodes a polypeptide of 1001 amino acids with a predicted molecular mass of 113,678 Da. RNF40 and its paralog RNF20 form a stable heterodimer complex that can monoubiquitylate histone H2B at lysine 120 as well as other nonhistone proteins. Cancer is a major public health problem and the second leading cause of death. Through its protein ubiquitylation activity, RNF40 acts as a tumor suppressor or oncogene to play major epigenetic roles in cancer development, progression, and metastasis, highlighting the essential function of RNF40 and the importance of studying it. In this review, we summarize current knowledge about RNF40 gene structure and the role of RNF40 in histone H2B monoubiquitylation, DNA damage repair, apoptosis, cancer development, and metastasis. We also underscore challenges in applying this information to cancer prognosis and prevention and highlight the urgent need for additional investigations of RNF40 as a potential target for cancer therapeutics.
Collapse
|
9
|
Gsell C, Richly H, Coin F, Naegeli H. A chromatin scaffold for DNA damage recognition: how histone methyltransferases prime nucleosomes for repair of ultraviolet light-induced lesions. Nucleic Acids Res 2020; 48:1652-1668. [PMID: 31930303 PMCID: PMC7038933 DOI: 10.1093/nar/gkz1229] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
The excision of mutagenic DNA adducts by the nucleotide excision repair (NER) pathway is essential for genome stability, which is key to avoiding genetic diseases, premature aging, cancer and neurologic disorders. Due to the need to process an extraordinarily high damage density embedded in the nucleosome landscape of chromatin, NER activity provides a unique functional caliper to understand how histone modifiers modulate DNA damage responses. At least three distinct lysine methyltransferases (KMTs) targeting histones have been shown to facilitate the detection of ultraviolet (UV) light-induced DNA lesions in the difficult to access DNA wrapped around histones in nucleosomes. By methylating core histones, these KMTs generate docking sites for DNA damage recognition factors before the chromatin structure is ultimately relaxed and the offending lesions are effectively excised. In view of their function in priming nucleosomes for DNA repair, mutations of genes coding for these KMTs are expected to cause the accumulation of DNA damage promoting cancer and other chronic diseases. Research on the question of how KMTs modulate DNA repair might pave the way to the development of pharmacologic agents for novel therapeutic strategies.
Collapse
Affiliation(s)
- Corina Gsell
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Holger Richly
- Boehringer Ingelheim Pharma, Department of Molecular Biology, Birkendorfer Str. 65, 88397 Biberach an der Riß, Germany
| | - Frédéric Coin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labélisée Ligue contre le Cancer, Illkirch Cedex, Strasbourg, France
| | - Hanspeter Naegeli
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| |
Collapse
|
10
|
Pavey S, Pinder A, Fernando W, D'Arcy N, Matigian N, Skalamera D, Lê Cao KA, Loo-Oey D, Hill MM, Stark M, Kimlin M, Burgess A, Cloonan N, Sturm RA, Gabrielli B. Multiple interaction nodes define the postreplication repair response to UV-induced DNA damage that is defective in melanomas and correlated with UV signature mutation load. Mol Oncol 2019; 14:22-41. [PMID: 31733171 PMCID: PMC6944116 DOI: 10.1002/1878-0261.12601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/06/2019] [Accepted: 11/14/2019] [Indexed: 01/25/2023] Open
Abstract
Ultraviolet radiation‐induced DNA mutations are a primary environmental driver of melanoma. The reason for this very high level of unrepaired DNA lesions leading to these mutations is still poorly understood. The primary DNA repair mechanism for UV‐induced lesions, that is, the nucleotide excision repair pathway, appears intact in most melanomas. We have previously reported a postreplication repair mechanism that is commonly defective in melanoma cell lines. Here we have used a genome‐wide approach to identify the components of this postreplication repair mechanism. We have used differential transcript polysome loading to identify transcripts that are associated with UV response, and then functionally assessed these to identify novel components of this repair and cell cycle checkpoint network. We have identified multiple interaction nodes, including global genomic nucleotide excision repair and homologous recombination repair, and previously unexpected MASTL pathway, as components of the response. Finally, we have used bioinformatics to assess the contribution of dysregulated expression of these pathways to the UV signature mutation load of a large melanoma cohort. We show that dysregulation of the pathway, especially the DNA damage repair components, are significant contributors to UV mutation load, and that dysregulation of the MASTL pathway appears to be a significant contributor to high UV signature mutation load.
Collapse
Affiliation(s)
- Sandra Pavey
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Alex Pinder
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Winnie Fernando
- Mater Research, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Nicholas D'Arcy
- Mater Research, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Nicholas Matigian
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia.,QFAB Bioinformatics, The University of Queensland, Brisbane, QLD, Australia
| | - Dubravka Skalamera
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia.,Mater Research, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Kim-Anh Lê Cao
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Dorothy Loo-Oey
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Michelle M Hill
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia.,QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Mitchell Stark
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Michael Kimlin
- University of the Sunshine Coast, Sippy Downs, QLD, Australia
| | | | - Nicole Cloonan
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Richard A Sturm
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Brian Gabrielli
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia.,Mater Research, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
11
|
Chen JH, Huang Y, Brachi B, Yun QZ, Zhang W, Lu W, Li HN, Li WQ, Sun XD, Wang GY, He J, Zhou Z, Chen KY, Ji YH, Shi MM, Sun WG, Yang YP, Zhang RG, Abbott RJ, Sun H. Genome-wide analysis of Cushion willow provides insights into alpine plant divergence in a biodiversity hotspot. Nat Commun 2019; 10:5230. [PMID: 31745089 PMCID: PMC6864086 DOI: 10.1038/s41467-019-13128-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 10/22/2019] [Indexed: 01/25/2023] Open
Abstract
The Hengduan Mountains (HDM) biodiversity hotspot exhibits exceptional alpine plant diversity. Here, we investigate factors driving intraspecific divergence within a HDM alpine species Salix brachista (Cushion willow), a common component of subnival assemblages. We produce a high-quality genome assembly for this species and characterize its genetic diversity, population structure and pattern of evolution by resequencing individuals collected across its distribution. We detect population divergence that has been shaped by a landscape of isolated sky island-like habitats displaying strong environmental heterogeneity across elevational gradients, combined with population size fluctuations that have occurred since approximately the late Miocene. These factors are likely important drivers of intraspecific divergence within Cushion willow and possibly other alpine plants with a similar distribution. Since intraspecific divergence is often the first step toward speciation, the same factors can be important contributors to the high alpine species diversity in the HDM.
Collapse
Affiliation(s)
- Jia-Hui Chen
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China.
| | - Yuan Huang
- School of Life Sciences, Yunnan Normal University, 650092, Kunming, Yunnan, P. R. China
| | | | - Quan-Zheng Yun
- Beijing Ori-Gene Science and Technology Co., Ltd, 102206, Beijing, P.R. China
| | - Wei Zhang
- State Key Laboratory of Protein and Plant Gene Research, Peking-Tsinghua Center for Life Sciences, and School of Life Sciences, Peking University, 100871, Beijing, P.R. China
- School of Life Sciences, Peking University, 100871, Beijing, P.R. China
| | - Wei Lu
- School of Life Sciences, Peking University, 100871, Beijing, P.R. China
| | - Hong-Na Li
- Beijing Ori-Gene Science and Technology Co., Ltd, 102206, Beijing, P.R. China
| | - Wen-Qing Li
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
| | - Xu-Dong Sun
- Institute of Tibetan Plateau Research at Kunming, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
- The Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
| | - Guang-Yan Wang
- Institute of Tibetan Plateau Research at Kunming, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
- The Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
| | - Jun He
- The Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
| | - Zhuo Zhou
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
| | - Kai-Yun Chen
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
| | - Yun-Heng Ji
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
| | - Ming-Ming Shi
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
| | - Wen-Guang Sun
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China
| | - Yong-Ping Yang
- Institute of Tibetan Plateau Research at Kunming, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China.
- The Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China.
| | - Ren-Gang Zhang
- Beijing Ori-Gene Science and Technology Co., Ltd, 102206, Beijing, P.R. China
| | - Richard J Abbott
- School of Biology, University of St. Andrews, St. Andrews, Fife, KY16 9TH, UK.
| | - Hang Sun
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, Yunnan, P. R. China.
| |
Collapse
|
12
|
Silva LE, Souza RC, Kitano ES, Monteiro LF, Iwai LK, Forti FL. Proteomic and Interactome Approaches Reveal PAK4, PHB-2, and 14-3-3η as Targets of Overactivated Cdc42 in Cellular Responses to Genomic Instability. J Proteome Res 2019; 18:3597-3614. [DOI: 10.1021/acs.jproteome.9b00260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Luiz E. Silva
- Laboratory of Signaling in Biomolecular Systems (LSSB), Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo-SP CEP 05508-900, Brazil
| | - Renan C. Souza
- Laboratory of Signaling in Biomolecular Systems (LSSB), Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo-SP CEP 05508-900, Brazil
| | - Eduardo S. Kitano
- Special Laboratory of Applied Toxicology (LETA), Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo-SP 05503-000, Brazil
| | - Lucas F. Monteiro
- Laboratory of Signaling in Biomolecular Systems (LSSB), Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo-SP CEP 05508-900, Brazil
| | - Leo K. Iwai
- Special Laboratory of Applied Toxicology (LETA), Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo-SP 05503-000, Brazil
| | - Fabio L. Forti
- Laboratory of Signaling in Biomolecular Systems (LSSB), Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo-SP CEP 05508-900, Brazil
| |
Collapse
|
13
|
Baxter LL, Watkins-Chow DE, Pavan WJ, Loftus SK. A curated gene list for expanding the horizons of pigmentation biology. Pigment Cell Melanoma Res 2019; 32:348-358. [PMID: 30339321 PMCID: PMC10413850 DOI: 10.1111/pcmr.12743] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 09/01/2018] [Accepted: 09/29/2018] [Indexed: 12/27/2022]
Abstract
Over the past century, studies of human pigmentary disorders along with mouse and zebrafish models have shed light on the many cellular functions associated with visible pigment phenotypes. This has led to numerous genes annotated with the ontology term "pigmentation" in independent human, mouse, and zebrafish databases. Comparisons among these datasets revealed that each is individually incomplete in documenting all genes involved in integument-based pigmentation phenotypes. Additionally, each database contained inherent species-specific biases in data annotation, and the term "pigmentation" did not solely reflect integument pigmentation phenotypes. This review presents a comprehensive, cross-species list of 650 genes involved in pigmentation phenotypes that was compiled with extensive manual curation of genes annotated in OMIM, MGI, ZFIN, and GO. The resulting cross-species list of genes both intrinsic and extrinsic to integument pigment cells provides a valuable tool that can be used to expand our knowledge of complex, pigmentation-associated pathways.
Collapse
Affiliation(s)
- Laura L Baxter
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Dawn E Watkins-Chow
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Stacie K Loftus
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
14
|
Persistence and Tolerance of DNA Damage Induced by Chronic UVB Irradiation of the Human Genome. J Invest Dermatol 2017; 138:405-412. [PMID: 28951242 DOI: 10.1016/j.jid.2017.08.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/25/2017] [Accepted: 08/25/2017] [Indexed: 12/17/2022]
Abstract
Exposure to solar UVB radiation leads to the formation of the highly mutagenic cyclobutane pyrimidine dimers (CPDs), the DNA damage responsible for mutations found in skin cancer. The frequency of CPD formation and the repair rate of those lesions are two important parameters to determine the probability of UVR-induced mutations. Previous work has shown that chronic irradiation with sublethal doses of UVB radiation (chronic low-dose UVB radiation) leads to the accumulation of residual CPD that persists over time. We have thus investigated the persistence, localization, and consequences on genome stability of those chronic low-dose UVB radiation-induced residual CPDs. We show that chronic low-dose UVB radiation-induced residual CPDs persist on DNA and are diluted via semiconservative replication. They are overrepresented in the heterochromatin and at the TT dipyrimidine sites, and they catalyze the incidence of sister chromatin exchange. Our results shed some light on the impact of chronic UVB radiation exposure on DNA, with a focus on residual CPDs, their distribution, and consequences.
Collapse
|
15
|
Yu L, Wang GD, Ruan J, Chen YB, Yang CP, Cao X, Wu H, Liu YH, Du ZL, Wang XP, Yang J, Cheng SC, Zhong L, Wang L, Wang X, Hu JY, Fang L, Bai B, Wang KL, Yuan N, Wu SF, Li BG, Zhang JG, Yang YQ, Zhang CL, Long YC, Li HS, Yang JY, Irwin DM, Ryder OA, Li Y, Wu CI, Zhang YP. Genomic analysis of snub-nosed monkeys (Rhinopithecus) identifies genes and processes related to high-altitude adaptation. Nat Genet 2016; 48:947-52. [DOI: 10.1038/ng.3615] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 06/13/2016] [Indexed: 12/31/2022]
|
16
|
Johnson CA, Collis SJ. Ciliogenesis and the DNA damage response: a stressful relationship. Cilia 2016; 5:19. [PMID: 27335639 PMCID: PMC4916530 DOI: 10.1186/s13630-016-0040-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/22/2016] [Indexed: 01/27/2023] Open
Abstract
Both inherited and sporadic mutations can give rise to a plethora of human diseases. Through myriad diverse cellular processes, sporadic mutations can arise through a failure to accurately replicate the genetic code or by inaccurate separation of duplicated chromosomes into daughter cells. The human genome has therefore evolved to encode a large number of proteins that work together with regulators of the cell cycle to ensure that it remains error-free. This is collectively known as the DNA damage response (DDR), and genome stability mechanisms involve a complex network of signalling and processing factors that ensure redundancy and adaptability of these systems. The importance of genome stability mechanisms is best illustrated by the dramatic increased risk of cancer in individuals with underlying disruption to genome maintenance mechanisms. Cilia are microtubule-based sensory organelles present on most vertebrate cells, where they facilitate transduction of external signals into the cell. When not embedded within the specialised ciliary membrane, components of the primary cilium's basal body help form the microtubule organising centre that controls cellular trafficking and the mitotic segregation of chromosomes. Ciliopathies are a collection of diseases associated with functional disruption to cilia function through a variety of different mechanisms. Ciliopathy phenotypes can vary widely, and although some cellular overgrowth phenotypes are prevalent in a subset of ciliopathies, an increased risk of cancer is not noted as a clinical feature. However, recent studies have identified surprising genetic and functional links between cilia-associated proteins and genome maintenance factors. The purpose of this mini-review is to therefore highlight some of these discoveries and discuss their implications with regards to functional crosstalk between the DDR and ciliogenesis pathways, and how this may impact on the development of human disease.
Collapse
Affiliation(s)
- Colin A. Johnson
- />Section of Ophthalmology and Neurosciences, Wellcome Trust Brenner Building, Leeds Institute of Molecular Medicine, St. James’s University Hospital, Leeds, LS9 7TF UK
| | - Spencer J. Collis
- />Genome Stability Group, Department of Oncology and Metabolism, Academic Unit of Molecular Oncology, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX UK
| |
Collapse
|
17
|
Ascer LG, Magalhaes YT, Espinha G, Osaki JH, Souza RC, Forti FL. CDC42 Gtpase Activation Affects Hela Cell DNA Repair and Proliferation Following UV Radiation-Induced Genotoxic Stress. J Cell Biochem 2016; 116:2086-97. [PMID: 25780896 DOI: 10.1002/jcb.25166] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 03/13/2015] [Indexed: 12/25/2022]
Abstract
Cell division control protein 42 (CDC42) homolog is a small Rho GTPase enzyme that participates in such processes as cell cycle progression, migration, polarity, adhesion, and transcription. Recent studies suggest that CDC42 is a potent tumor suppressor in different tissues and is related to aging processes. Although DNA damage is crucial in aging, a potential role for CDC42 in genotoxic stress remains to be explored. Migration, survival/proliferation and DNA damage/repair experiments were performed to demonstrate CDC42 involvement in the recovery of HeLa cells exposed to ultraviolet radiation-induced stress. Sub-lines of HeLa cells ectopically expressing the constitutively active CDC42-V12 mutant were generated to examine whether different CDC42-GTP backgrounds might reflect different sensitivities to UV radiation. Our results show that CDC42 constitutive activation does not interfere with HeLa cell migration after UV radiation. However, the minor DNA damage exhibited by the CDC42-V12 mutant exposed to UV radiation most likely results in cell cycle arrest at the G2/M checkpoint and reduced proliferation and survival. HeLa cells and Mock clones, which express endogenous wild-type CDC42 and show normal activity, are more resistant to UV radiation. None of these effects are altered by pharmacological CDC42 inhibition. Finally, the phosphorylation status of the DNA damage response proteins γ-H2AX and p-Chk1 was found to be delayed and attenuated, respectively, in CDC42-V12 clones. In conclusion, the sensitivity of HeLa cells to ultraviolet radiation increases with CDC42 over-activation due to inadequate DNA repair signaling, culminating in G2/M cell accumulation, which is translated into reduced cellular proliferation and survival.
Collapse
Affiliation(s)
- Liv G Ascer
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil
| | - Yuli T Magalhaes
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil
| | - Gisele Espinha
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil
| | - Juliana H Osaki
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil
| | - Renan C Souza
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil
| | - Fabio L Forti
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
18
|
Grbatinić I, Milošević NT. Incipient UV-Induced Structural Changes in Neutrophil Granulocytes: Morphometric and Texture Analysis of Two-Dimensional Digital Images. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2016; 22:387-393. [PMID: 26906218 DOI: 10.1017/s1431927616000532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The aim of this study is to determine the ability and consequent significance of fractal and lacunarity analysis together with computational morphometric and gray-level co-occurrence matrix (GLCM) analysis in detecting subtle initial UVB-induced chromatin and cytosolic changes in neutrophil granulocytes. In addition, the direction and potential significance of the observed changes is speculated. Feulgen-stained neutrophils are pictured and their digitalized images are analyzed in specialized software for digital image processing and ImageJ analysis. Significant statistical difference is observed (p0.05). For other parameters there was mostly high statistical significance (p>0.05). Significant unmatched correlations were found as sensitive markers of early morphological changes in cells exposed to UV light. In addition, the correlation between nuclear area and entropy was determined and was highly significant (p<0.001). UVB light, due to its high absorbance by DNA molecules, leads to double behavior of the cells. On one hand, cells start to rearrange but on the other UV light starts very early to immediately damage the cell. All these processes are very subtle in their intensity and GLCM analysis and computational imaging methods based on fractal geometry, i.e. fractal and morphometric analysis, in particular their combination, are very sensitive for detecting and describing these early chromatin changes.
Collapse
Affiliation(s)
- Ivan Grbatinić
- 1Laboratory of Digital Image Processing,School of Medicine,University of Belgrade,Visegradka 2, Belgrade,Serbia
| | - Nebojša T Milošević
- 2Department of Biophysics,School of Medicine,University of Belgrade,Visegradka 2, Belgrade,Serbia
| |
Collapse
|
19
|
Wang Y, Gao D, Chu B, Gao C, Cao D, Liu H, Jiang Y. Exposure of CCRF-CEM cells to acridone derivative 8a triggers tumor death via multiple mechanisms. Proteomics 2016; 16:1177-90. [DOI: 10.1002/pmic.201500317] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Yini Wang
- Department of Chemistry; Tsinghua University; Beijing P. R. China
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
| | - Dan Gao
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Bizhu Chu
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Chunmei Gao
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Deliang Cao
- Department of Medical Microbiology; Immunology and Cell Biology; Simmons Cancer Institute; Southern Illinois University School of Medicine, Springfield; IL USA
| | - Hongxia Liu
- The Key Laboratory of Tumor Metabolomics at Shenzhen; Shenzhen P. R. China
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
| | - Yuyang Jiang
- The State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology; Graduate School at Shenzhen; Tsinghua University; Shenzhen P. R. China
- School of Medicine; Tsinghua University; Beijing P. R. China
| |
Collapse
|
20
|
Liu R, Jin L, Long K, Chai J, Ma J, Tang Q, Tian S, Hu Y, Lin L, Wang X, Jiang A, Li X, Li M. Detection of genetic diversity and selection at the coding region of the melanocortin receptor 1 ( MC1R ) gene in Tibetan pigs and Landrace pigs. Gene 2016; 575:537-542. [DOI: 10.1016/j.gene.2015.09.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 08/13/2015] [Accepted: 09/15/2015] [Indexed: 11/30/2022]
|
21
|
Manova V, Gruszka D. DNA damage and repair in plants - from models to crops. FRONTIERS IN PLANT SCIENCE 2015; 6:885. [PMID: 26557130 PMCID: PMC4617055 DOI: 10.3389/fpls.2015.00885] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 10/05/2015] [Indexed: 05/17/2023]
Abstract
The genomic integrity of every organism is constantly challenged by endogenous and exogenous DNA-damaging factors. Mutagenic agents cause reduced stability of plant genome and have a deleterious effect on development, and in the case of crop species lead to yield reduction. It is crucial for all organisms, including plants, to develop efficient mechanisms for maintenance of the genome integrity. DNA repair processes have been characterized in bacterial, fungal, and mammalian model systems. The description of these processes in plants, in contrast, was initiated relatively recently and has been focused largely on the model plant Arabidopsis thaliana. Consequently, our knowledge about DNA repair in plant genomes - particularly in the genomes of crop plants - is by far more limited. However, the relatively small size of the Arabidopsis genome, its rapid life cycle and availability of various transformation methods make this species an attractive model for the study of eukaryotic DNA repair mechanisms and mutagenesis. Moreover, abnormalities in DNA repair which proved to be lethal for animal models are tolerated in plant genomes, although sensitivity to DNA damaging agents is retained. Due to the high conservation of DNA repair processes and factors mediating them among eukaryotes, genes and proteins that have been identified in model species may serve to identify homologous sequences in other species, including crop plants, in which these mechanisms are poorly understood. Crop breeding programs have provided remarkable advances in food quality and yield over the last century. Although the human population is predicted to "peak" by 2050, further advances in yield will be required to feed this population. Breeding requires genetic diversity. The biological impact of any mutagenic agent used for the creation of genetic diversity depends on the chemical nature of the induced lesions and on the efficiency and accuracy of their repair. More recent targeted mutagenesis procedures also depend on host repair processes, with different pathways yielding different products. Enhanced understanding of DNA repair processes in plants will inform and accelerate the engineering of crop genomes via both traditional and targeted approaches.
Collapse
Affiliation(s)
- Vasilissa Manova
- Department of Molecular Genetics, Institute of Plant Physiology and Genetics, Bulgarian Academy of SciencesSofia
| | - Damian Gruszka
- Department of Genetics, Faculty of Biology and Environment Protection, University of SilesiaKatowice, Poland
| |
Collapse
|
22
|
Abstract
DNA damage is a constant threat to cells, causing cytotoxicity as well as inducing genetic alterations. The steady-state abundance of DNA lesions in a cell is minimized by a variety of DNA repair mechanisms, including DNA strand break repair, mismatch repair, nucleotide excision repair, base excision repair, and ribonucleotide excision repair. The efficiencies and mechanisms by which these pathways remove damage from chromosomes have been primarily characterized by investigating the processing of lesions at defined genomic loci, among bulk genomic DNA, on episomal DNA constructs, or using in vitro substrates. However, the structure of a chromosome is heterogeneous, consisting of heavily protein-bound heterochromatic regions, open regulatory regions, actively transcribed genes, and even areas of transient single stranded DNA. Consequently, DNA repair pathways function in a much more diverse set of chromosomal contexts than can be readily assessed using previous methods. Recent efforts to develop whole genome maps of DNA damage, repair processes, and even mutations promise to greatly expand our understanding of DNA repair and mutagenesis. Here we review the current efforts to utilize whole genome maps of DNA damage and mutation to understand how different chromosomal contexts affect DNA excision repair pathways.
Collapse
Affiliation(s)
- John J Wyrick
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA; Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA.
| | - Steven A Roberts
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
23
|
Bee L, Marini S, Pontarin G, Ferraro P, Costa R, Albrecht U, Celotti L. Nucleotide excision repair efficiency in quiescent human fibroblasts is modulated by circadian clock. Nucleic Acids Res 2015; 43:2126-37. [PMID: 25662220 PMCID: PMC4344517 DOI: 10.1093/nar/gkv081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The efficiency of Nucleotide Excision Repair (NER)process is crucial for maintaining genomic integrity because in many organisms, including humans, it represents the only system able to repair a wide range of DNA damage. The aim of the work was to investigate whether the efficiency of the repair of photoproducts induced by UV-light is affected by the circadian phase at which irradiation occurred. NER activity has been analyzed in human quiescent fibroblasts (in the absence of the cell cycle effect), in which circadian rhythmicity has been synchronized with a pulse of dexamethasone. Our results demonstrate that both DNA damage induction and repair efficiency are strictly dependent on the phase of the circadian rhythm at which the cells are UV-exposed. Furthermore, the differences observed between fibroblasts irradiated at different circadian times (CTs) are abolished when the clock is obliterated. In addition, we observe that chromatin structure is regulated by circadian rhythmicity. Maximal chromatin relaxation occurred at the same CT when photoproduct formation and removal were highest. Our data suggest that the circadian clock regulates both the DNA sensitivity to UV damage and the efficiency of NER by controlling chromatin condensation mainly through histone acetylation.
Collapse
Affiliation(s)
- Leonardo Bee
- Department of Biology, University of Padova, Padova 35131, Italy
| | - Selena Marini
- Department of Biology, University of Padova, Padova 35131, Italy
| | | | - Paola Ferraro
- Department of Biology, University of Padova, Padova 35131, Italy
| | - Rodolfo Costa
- Department of Biology, University of Padova, Padova 35131, Italy
| | - Urs Albrecht
- Department of Biology, Unit for Biochemistry, University of Fribourg, Fribourg CH-1700, Switzerland
| | - Lucia Celotti
- Department of Biology, University of Padova, Padova 35131, Italy
| |
Collapse
|
24
|
Karentz D. Beyond xeroderma pigmentosum: DNA damage and repair in an ecological context. A tribute to James E. Cleaver. Photochem Photobiol 2014; 91:460-74. [PMID: 25395165 DOI: 10.1111/php.12388] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/29/2014] [Indexed: 12/12/2022]
Abstract
The ability to repair DNA is a ubiquitous characteristic of life on Earth and all organisms possess similar mechanisms for dealing with DNA damage, an indication of a very early evolutionary origin for repair processes. James E. Cleaver's career (initiated in the early 1960s) has been devoted to the study of mammalian ultraviolet radiation (UVR) photobiology, specifically the molecular genetics of xeroderma pigmentosum and other human diseases caused by defects in DNA damage recognition and repair. This work by Jim and others has influenced the study of DNA damage and repair in a variety of taxa. Today, the field of DNA repair is enhancing our understanding of not only how to treat and prevent human disease, but is providing insights on the evolutionary history of life on Earth and how natural populations are coping with UVR-induced DNA damage from anthropogenic changes in the environment such as ozone depletion.
Collapse
Affiliation(s)
- Deneb Karentz
- Department of Biology, University of San Francisco, San Francisco, CA
| |
Collapse
|
25
|
You X, Bian C, Zan Q, Xu X, Liu X, Chen J, Wang J, Qiu Y, Li W, Zhang X, Sun Y, Chen S, Hong W, Li Y, Cheng S, Fan G, Shi C, Liang J, Tom Tang Y, Yang C, Ruan Z, Bai J, Peng C, Mu Q, Lu J, Fan M, Yang S, Huang Z, Jiang X, Fang X, Zhang G, Zhang Y, Polgar G, Yu H, Li J, Liu Z, Zhang G, Ravi V, Coon SL, Wang J, Yang H, Venkatesh B, Wang J, Shi Q. Mudskipper genomes provide insights into the terrestrial adaptation of amphibious fishes. Nat Commun 2014; 5:5594. [PMID: 25463417 PMCID: PMC4268706 DOI: 10.1038/ncomms6594] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 10/16/2014] [Indexed: 01/18/2023] Open
Abstract
Mudskippers are amphibious fishes that have developed morphological and physiological adaptations to match their unique lifestyles. Here we perform whole-genome sequencing of four representative mudskippers to elucidate the molecular mechanisms underlying these adaptations. We discover an expansion of innate immune system genes in the mudskippers that may provide defence against terrestrial pathogens. Several genes of the ammonia excretion pathway in the gills have experienced positive selection, suggesting their important roles in mudskippers’ tolerance to environmental ammonia. Some vision-related genes are differentially lost or mutated, illustrating genomic changes associated with aerial vision. Transcriptomic analyses of mudskippers exposed to air highlight regulatory pathways that are up- or down-regulated in response to hypoxia. The present study provides a valuable resource for understanding the molecular mechanisms underlying water-to-land transition of vertebrates. Mudskippers are amphibious fishes that have adapted to live on mudflats. Here, the authors sequence the genomes of four different mudskipper species and highlight genetic changes that may have had an evolutionary role in the water-to-land transition of vertebrates.
Collapse
Affiliation(s)
- Xinxin You
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | - Chao Bian
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | - Qijie Zan
- Shenzhen Wild Animal Rescue Center, Shenzhen 518040, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xin Liu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Jieming Chen
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | | | - Ying Qiu
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | - Wujiao Li
- Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China
| | - Xinhui Zhang
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | - Ying Sun
- BGI-Shenzhen, Shenzhen 518083, China
| | - Shixi Chen
- College of Ocean and Earth Science, Xiamen University, Xiamen 361005, China
| | - Wanshu Hong
- College of Ocean and Earth Science, Xiamen University, Xiamen 361005, China
| | | | | | | | | | - Jie Liang
- BGI-Shenzhen, Shenzhen 518083, China
| | | | | | - Zhiqiang Ruan
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | - Jie Bai
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | - Chao Peng
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | - Qian Mu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Jun Lu
- 1] BGI-Shenzhen, Shenzhen 518083, China [2] Shenzhen BGI Fisheries Sci &Tech Co. Ltd, Shenzhen 518083, China
| | - Mingjun Fan
- Center for Fish Genomics, BGI-Wuhan, Wuhan 430075, China
| | - Shuang Yang
- 1] BGI-Shenzhen, Shenzhen 518083, China [2] Center for Fish Genomics, BGI-Wuhan, Wuhan 430075, China
| | | | | | | | | | | | - Gianluca Polgar
- Environmental and Life Sciences Programme, Faculty of Science, Universiti Brunei Darussalam, Jln Tungku Link, BE1410 Brunei Darussalam
| | - Hui Yu
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | - Jia Li
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China
| | - Zhongjian Liu
- Shenzhen Key Laboratory for Orchid Conservation and Utilization of the Orchid Conservation and Research Center of Shenzhen, Shenzhen 518114, China
| | - Guoqiang Zhang
- Shenzhen Key Laboratory for Orchid Conservation and Utilization of the Orchid Conservation and Research Center of Shenzhen, Shenzhen 518114, China
| | - Vydianathan Ravi
- Institute of Molecular and Cell Biology, A*STAR, Biopolis, Singapore 138673, Singapore
| | - Steven L Coon
- Molecular Genomics Laboratory, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jian Wang
- 1] BGI-Shenzhen, Shenzhen 518083, China [2] James D. Watson Institute of Genome Science, Hangzhou 310008, China
| | - Huanming Yang
- 1] BGI-Shenzhen, Shenzhen 518083, China [2] James D. Watson Institute of Genome Science, Hangzhou 310008, China [3] Princess Al Jawhara Center of Excellence in the Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Byrappa Venkatesh
- Institute of Molecular and Cell Biology, A*STAR, Biopolis, Singapore 138673, Singapore
| | - Jun Wang
- 1] BGI-Shenzhen, Shenzhen 518083, China [2] Princess Al Jawhara Center of Excellence in the Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia [3] Department of Biology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Qiong Shi
- 1] Shenzhen Key Lab of Marine Genomics, State Key Laboratory of Agricultural Genomics, Shenzhen 518083, China [2] BGI-Shenzhen, Shenzhen 518083, China [3] Shenzhen BGI Fisheries Sci &Tech Co. Ltd, Shenzhen 518083, China [4] Center for Fish Genomics, BGI-Wuhan, Wuhan 430075, China
| |
Collapse
|
26
|
Li Z, Mon H, Mitsunobu H, Zhu L, Xu J, Lee JM, Kusakabe T. Dynamics of polycomb proteins-mediated histone modifications during UV irradiation-induced DNA damage. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2014; 55:9-18. [PMID: 25308962 DOI: 10.1016/j.ibmb.2014.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/28/2014] [Accepted: 10/02/2014] [Indexed: 06/04/2023]
Abstract
Polycomb group (PcG) complexes are known to be chromatin modifiers and transcriptional repressors. In this work, we reported that the histone-modifying PcG complexes are able to participate in the repair process of ultraviolet (UV)-induced DNA lesions in the silkworm, Bombyx mori. The silkworm cells with depletion of PcG genes showed hypersensitive to UV-C irradiation and increased inhibition of cell proliferation. Interestingly, an SQ site in the silkworm-human chimeric H2A protein synthesized here was phosphorylated rapidly upon UV-C exposure, which could be used as a marker for monitoring the response to DNA damage in silkworm cells. Under these UV-C irradiated conditions, we found that PRC1-mediated ubiquitylation of H2AX, but not of H2AZ, were decreased and this deubiquitylation was independent of its phosphorylation event. In contrast, UV-C irradiation induced the increase of trimethylation of lysine 27 on histone H3 (H3K27me3), a mark of transcriptionally silent chromatin catalyzed by another PcG subcomplex, PRC2. Collectively, we provided the first evidence on chromatin remodeling in response to UV-C lesion in silkworm and revealed another layer role for PcG complexes-mediated histone modifications in contributing to creating an open chromatin structure for the efficient repair of DNA damages.
Collapse
Affiliation(s)
- Zhiqing Li
- Laboratory of Silkworm Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Hakozaki 6-10-1, Fukuoka 812-8581, Japan
| | - Hiroaki Mon
- Laboratory of Silkworm Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Hakozaki 6-10-1, Fukuoka 812-8581, Japan
| | - Hitoshi Mitsunobu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Li Zhu
- Laboratory of Silkworm Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Hakozaki 6-10-1, Fukuoka 812-8581, Japan
| | - Jian Xu
- Laboratory of Silkworm Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Hakozaki 6-10-1, Fukuoka 812-8581, Japan
| | - Jae Man Lee
- Laboratory of Silkworm Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Hakozaki 6-10-1, Fukuoka 812-8581, Japan
| | - Takahiro Kusakabe
- Laboratory of Silkworm Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Hakozaki 6-10-1, Fukuoka 812-8581, Japan.
| |
Collapse
|
27
|
RAD6 promotes homologous recombination repair by activating the autophagy-mediated degradation of heterochromatin protein HP1. Mol Cell Biol 2014; 35:406-16. [PMID: 25384975 DOI: 10.1128/mcb.01044-14] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Efficient DNA double-strand break (DSB) repair is critical for the maintenance of genome stability. Unrepaired or misrepaired DSBs cause chromosomal rearrangements that can result in severe consequences, such as tumorigenesis. RAD6 is an E2 ubiquitin-conjugating enzyme that plays a pivotal role in repairing UV-induced DNA damage. Here, we present evidence that RAD6 is also required for DNA DSB repair via homologous recombination (HR) by specifically regulating the degradation of heterochromatin protein 1α (HP1α). Our study indicates that RAD6 physically interacts with HP1α and ubiquitinates HP1α at residue K154, thereby promoting HP1α degradation through the autophagy pathway and eventually leading to an open chromatin structure that facilitates efficient HR DSB repair. Furthermore, bioinformatics studies have indicated that the expression of RAD6 and HP1α exhibits an inverse relationship and correlates with the survival rate of patients.
Collapse
|
28
|
Zhang L, Nemzow L, Chen H, Hu JJ, Gong F. Whole genome expression profiling shows that BRG1 transcriptionally regulates UV inducible genes and other novel targets in human cells. PLoS One 2014; 9:e105764. [PMID: 25157878 PMCID: PMC4144907 DOI: 10.1371/journal.pone.0105764] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/19/2014] [Indexed: 12/16/2022] Open
Abstract
UV irradiation is known to cause cyclobutane pyrimidine dimers (CPDs) and pyrimidine (6–4) pyrimidone photoproducts (6-4PPs), and plays a large role in the development of cancer. Tumor suppression, through DNA repair and proper cell cycle regulation, is an integral factor in maintaining healthy cells and preventing development of cancer. Transcriptional regulation of the genes involved in the various tumor suppression pathways is essential for them to be expressed when needed and to function properly. BRG1, an ATPase catalytic subunit of the SWI/SNF chromatin remodeling complex, has been identified as a tumor suppressor protein, as it has been shown to play a role in Nucleotide Excision Repair (NER) of CPDs, suppress apoptosis, and restore checkpoint deficiency, in response to UV exposure. Although BRG1 has been shown to regulate transcription of some genes that are instrumental in proper DNA damage repair and cell cycle maintenance in response to UV, its role in transcriptional regulation of the whole genome in response to UV has not yet been elucidated. With whole genome expression profiling in SW13 cells, we show that upon UV induction, BRG1 regulates transcriptional expression of many genes involved in cell stress response. Additionally, our results also highlight BRG1's general role as a master regulator of the genome, as it transcriptionally regulates approximately 4.8% of the human genome, including expression of genes involved in many pathways. RT-PCR and ChIP were used to validate our genome expression analysis. Importantly, our study identifies several novel transcriptional targets of BRG1, such as ATF3. Thus, BRG1 has a larger impact on human genome expression than previously thought, and our studies will provide inroads for future analysis of BRG1's role in gene regulation.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Leah Nemzow
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Hua Chen
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Jennifer J. Hu
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Feng Gong
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
29
|
Hematopoietic myeloid cell differentiation diminishes nucleotide excision repair. Int J Hematol 2014; 100:260-5. [PMID: 25027282 DOI: 10.1007/s12185-014-1625-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 06/29/2014] [Accepted: 06/30/2014] [Indexed: 10/25/2022]
Abstract
Myeloid cell differentiation is the process by which stem cells develop into mature monocytes or granulocytes. This process is achieved by the sequential activation of variety of genes. Disruption of this process can result in immunodeficiency, bone marrow failure syndrome, or leukemia. Acute promyelocytic leukemia (APL) is characterized by the t(15;17) translocation and can be treated by a combination of all-trans retinoic acid (ATRA) and anthracycline. This treatment can induce leukemic cell differentiation, leading to extremely high remission rates. XAB2, a molecule involved in nucleotide excision repair (NER), is downregulated during granulocyte differentiation and shows reduced expression in NB4 APL-derived cells in vitro. Differentiation of APL by ATRA treatment reduced XAB2 expression levels in vivo. These observations suggest that cellular differentiation is associated with reduced NER activity and provides new insights into combined differentiation induction. NB4 cells were more susceptible than the immature myeloid leukemic cell lines, Kasumi-3 and Kasumi-1, to the DNA interstrand crosslinking agent cisplatin.
Collapse
|
30
|
Chen LC, Wu JC, Tuan YF, Tseng YK, Hseu YC, Chen SC. Molecular mechanisms of 3,3'-dichlorobenzidine-mediated toxicity in HepG2 cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2014; 55:407-420. [PMID: 24604609 DOI: 10.1002/em.21858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 01/29/2014] [Accepted: 01/29/2014] [Indexed: 06/03/2023]
Abstract
3,3'-Dichlorobenzidine (DCB) (CAS 91-94-1), a synthetic, chlorinated, primary aromatic amine, is typically used as an intermediate in the manufacturing of pigments for printing inks, textiles, paints, and plastics. In this study, we found that DCB could significantly inhibit the cell viability of HepG2 cells in a concentration-dependent manner. Flow cytometry revealed that DCB induced G2/M-phase arrest and apoptosis in HepG2 cells. DCB treatment dramatically induced the dissipation of mitochondrial membrane potential (Δψm ) and enhanced the enzymatic activities of caspase-9 and caspase-3 whilst hardly affecting caspase-8 activity. Furthermore, Western blotting indicated that DCB-induced apoptosis was accompanied by the down-regulation of Bcl-2/Bax ratio. These results suggested that DCB led to cytotoxicity involving activation of mitochondrial-dependent apoptosis through Bax/Bcl-2 pathways in HepG2 cells. Furthermore, HepG2 cells treated with DCB showed significant DNA damage as supported by the concentration-dependent increase in olive tail moments as determined by the comet assay and by concentration- and time-dependent increase in histone H2AX phosphorylation (γ-H2AX). Two-dimensional-difference gel electrophoresis (2D-DIGE), combined with mass spectrometry (MS), was used to unveil the differences in protein expression between cells exposed to 25 µM or 100 µM of DCB for 24 hr and the control cells. Twenty-seven differentially expressed proteins involved in DNA repair, unfolded protein response, metabolism, cell signaling, and apoptosis were identified. Among these, 14-3-3 theta, CGI-46, and heat-shock 70 protein 4 were confirmed using Western blot assay. Taken together, these data suggest that DCB is capable of inducing DNA damage and some cellular stress responses in HepG2 cells, thus eventually leading to cell death by apoptosis.
Collapse
MESH Headings
- 3,3'-Dichlorobenzidine/adverse effects
- Apoptosis/drug effects
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Carcinogens/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Caspase 3/metabolism
- Caspase 8/metabolism
- Caspase 9/metabolism
- Cell Cycle/drug effects
- Cell Proliferation/drug effects
- Electrophoresis, Gel, Two-Dimensional
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Membrane Potential, Mitochondrial/drug effects
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Lei-Chin Chen
- Department of Nutrition, I-Shou University, Kaohsiung City, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
31
|
Shapiro JA. Epigenetic control of mobile DNA as an interface between experience and genome change. Front Genet 2014; 5:87. [PMID: 24795749 PMCID: PMC4007016 DOI: 10.3389/fgene.2014.00087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/01/2014] [Indexed: 12/29/2022] Open
Abstract
Mobile DNA in the genome is subject to RNA-targeted epigenetic control. This control regulates the activity of transposons, retrotransposons and genomic proviruses. Many different life history experiences alter the activities of mobile DNA and the expression of genetic loci regulated by nearby insertions. The same experiences induce alterations in epigenetic formatting and lead to trans-generational modifications of genome expression and stability. These observations lead to the hypothesis that epigenetic formatting directed by non-coding RNA provides a molecular interface between life history events and genome alteration.
Collapse
Affiliation(s)
- James A. Shapiro
- Department of Biochemistry and Molecular Biology, University of ChicagoChicago, IL, USA
| |
Collapse
|
32
|
Chakarov S, Roeva I, Russev G. An Experimental Model for Assessment of Global DNA Repair Capacity. BIOTECHNOL BIOTEC EQ 2014. [DOI: 10.5504/bbeq.2011.0080] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
33
|
Adam S, Polo SE, Almouzni G. How to restore chromatin structure and function in response to DNA damage--let the chaperones play: delivered on 9 July 2013 at the 38th FEBS Congress in St Petersburg, Russia. FEBS J 2014; 281:2315-23. [PMID: 24673849 DOI: 10.1111/febs.12793] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 03/25/2014] [Indexed: 01/07/2023]
Abstract
Histone deposition onto DNA assisted by specific chaperones forms the chromatin basic unit and serves to package the genome within the cell nucleus. The resulting chromatin organization, often referred to as the epigenome, contributes to a unique transcriptional program that defines cell identity. Importantly, during cellular life, substantial alterations in chromatin structure may arise due to cell stress, including DNA damage, which not only challenges the integrity of the genome but also threatens the epigenome. Considerable efforts have been made to decipher chromatin dynamics in response to genotoxic stress, and to assess how it affects both genome and epigenome stability. Here, we review recent advances in understanding the mechanisms of DNA damage-induced chromatin plasticity in mammalian cells. We focus specifically on the dynamics of histone H3 variants in response to UV irradiation, and highlight the role of their dedicated chaperones in restoring both chromatin structure and function. Finally, we discuss how, in addition to restoring chromatin integrity, the cellular networks that signal and repair DNA damage may also provide a window of opportunity for modulating the information conveyed by chromatin.
Collapse
Affiliation(s)
- Salomé Adam
- Institut Curie, Centre de Recherche, Paris, France; Centre National de la Recherche Scientifique, UMR3664, Paris, France; Equipe Labellisée Ligue Contre le Cancer, Paris, France; Institut de Formation Doctorale, University Pierre & Marie Curie, Paris, France; Sorbonne University, PSL*, Paris, France; Epigenetics and Cell Fate Centre, UMR7216, Centre National de la Recherche Scientifique/Paris Diderot University, Paris, France
| | | | | |
Collapse
|
34
|
Abstract
The ability of cullin 4A (CUL4A), a scaffold protein, to recruit a repertoire of substrate adaptors allows it to assemble into distinct E3 ligase complexes to mediate turnover of key regulatory proteins. In the past decade, a considerable wealth of information has been generated regarding its biology, regulation, assembly, molecular architecture and novel functions. Importantly, unravelling of its association with multiple tumours and modulation by viral proteins establishes it as one of the key proteins that may play an important role in cellular transformation. Considering the role of its substrate in regulating the cell cycle and maintenance of genomic stability, understanding the detailed aspects of these processes will have significant consequences for the treatment of cancer and related diseases. This review is an effort to provide a broad overview of this multifaceted ubiquitin ligase and addresses its critical role in regulation of important biological processes. More importantly, its tremendous potential to be exploited for therapeutic purposes has been discussed.
Collapse
Affiliation(s)
- Puneet Sharma
- Department of Biochemistry, University of Delhi, South Campus, New Delhi, India
| | | |
Collapse
|
35
|
Polak P, Lawrence MS, Haugen E, Stoletzki N, Stojanov P, Thurman RE, Garraway LA, Mirkin S, Getz G, Stamatoyannopoulos JA, Sunyaev SR. Reduced local mutation density in regulatory DNA of cancer genomes is linked to DNA repair. Nat Biotechnol 2013; 32:71-5. [PMID: 24336318 PMCID: PMC4116484 DOI: 10.1038/nbt.2778] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 11/22/2013] [Indexed: 12/28/2022]
Abstract
Carcinogenesis and neoplastic progression are mediated by the accumulation of somatic mutations. Here we report that the local density of somatic mutations in cancer genomes is highly reduced specifically in accessible regulatory DNA defined by DNase I hypersensitive sites. This reduction is independent of any known factors influencing somatic mutation density and is observed in diverse cancer types, suggesting a general mechanism. By analyzing individual cancer genomes1, we show that the reduced local mutation density within regulatory DNA is linked to intact global genome repair machinery, with nearly complete abrogation of the hypomutation phenomenon in individual cancers that possess mutations in multiple nucleotide excision repair components. Together, our results connect chromatin structure, gene regulation and cancer-associated somatic mutation.
Collapse
Affiliation(s)
- Paz Polak
- 1] Division of Genetics, Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA. [2] Harvard Medical School, Boston, Massachusetts, USA. [3] The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA. [4]
| | - Michael S Lawrence
- 1] The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA. [2]
| | - Eric Haugen
- Departments of Genome Sciences and Medicine (Oncology), University of Washington, Seattle, Washington, USA
| | - Nina Stoletzki
- 1] Division of Genetics, Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA. [2] Harvard Medical School, Boston, Massachusetts, USA. [3] The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Petar Stojanov
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Robert E Thurman
- Departments of Genome Sciences and Medicine (Oncology), University of Washington, Seattle, Washington, USA
| | - Levi A Garraway
- 1] Harvard Medical School, Boston, Massachusetts, USA. [2] The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA. [3] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA. [4] Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Sergei Mirkin
- Department of Biology, Tufts University, Medford, Massachusetts, USA
| | - Gad Getz
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - John A Stamatoyannopoulos
- Departments of Genome Sciences and Medicine (Oncology), University of Washington, Seattle, Washington, USA
| | - Shamil R Sunyaev
- 1] Division of Genetics, Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA. [2] Harvard Medical School, Boston, Massachusetts, USA. [3] The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
36
|
Jagirdar K, Yin K, Harrison M, Lim W, Muscat GEO, Sturm RA, Smith AG. The NR4A2 nuclear receptor is recruited to novel nuclear foci in response to UV irradiation and participates in nucleotide excision repair. PLoS One 2013; 8:e78075. [PMID: 24223135 PMCID: PMC3819332 DOI: 10.1371/journal.pone.0078075] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 09/17/2013] [Indexed: 12/11/2022] Open
Abstract
Ultraviolet radiation (UVR) is one of the most common mutagens encountered by humans and induces the formation of cyclobutane pyrimidine dimers (CPDs) and pyrimidine-(6-4)-pyrimidone photoproduct (6-4PP) lesions in the genomic DNA. To prevent the accumulation of deleterious mutations these lesions must be efficiently repaired, primarily by nucleotide excision repair. We have previously demonstrated that the NR4A family of nuclear receptors are crucial mediators of the DNA repair function of the MC1R signalling pathway in melanocytes. Here we explore the role of the NR4A2 protein in the DNA repair process further. Using EYFP tagged-NR4A2 we have demonstrated a UVR induced recruitment to distinct nuclear foci where they co-localise with known DNA repair proteins. We reveal that the N-terminal domain of the receptor is required for this translocation and identify a role for p38 and PARP signalling in this process. Moreover disruption of the functional integrity of the Ligand Binding Domain of the receptor by deleting the terminal helix 12 effectively blocks co-localisation of the receptor with DNA repair factors. Restored co-localisation of the mutant receptor with DNA repair proteins in the presence of a Histone Deacetylase Inhibitor suggests that impaired chromatin accessibility underpins the mis-localisation observed. Finally NR4A2 over-expression facilitated a more efficient clearance of UVR induced CPD and 6-4PP lesions. Taken together these data uncover a novel role for the NR4A nuclear receptors as direct facilitators of nucleotide excision repair.
Collapse
Affiliation(s)
- Kasturee Jagirdar
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Kelvin Yin
- School of Biomedical Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Matthew Harrison
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Wen Lim
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - George E. O. Muscat
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Richard A. Sturm
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Aaron G. Smith
- School of Biomedical Science, The University of Queensland, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
37
|
Genomic analyses identify distinct patterns of selection in domesticated pigs and Tibetan wild boars. Nat Genet 2013; 45:1431-8. [DOI: 10.1038/ng.2811] [Citation(s) in RCA: 341] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 10/04/2013] [Indexed: 12/28/2022]
|
38
|
Keuser B, Khobta A, Gallé K, Anderhub S, Schulz I, Pauly K, Epe B. Influences of histone deacetylase inhibitors and resveratrol on DNA repair and chromatin compaction. Mutagenesis 2013; 28:569-76. [PMID: 23814181 DOI: 10.1093/mutage/get034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Accessibility of DNA is a prerequisite for both DNA damage and repair. Therefore, the chromatin structure is expected to have major impact on both processes, with opposite consequences for the stability of the genome. To analyse the influence of chromatin compaction on the generation and repair of various types of DNA modifications, we modulated the global chromatin structure of AS52 Chinese hamster ovary cells and HeLa cells by treatment with either histone deacetylase inhibitors or resveratrol and measured the repair kinetics of (i) pyrimidine dimers induced by ultraviolet B, (ii) oxidised purines generated by photosensitisation and (iii) single-strand breaks induced by H2O2, using an alkaline elution technique. The decrease of chromatin compaction (detected as reduced DNA accessibility to DNase I) after treatment with trichostatin A or butyrate slightly increased the damage generation but had no significant effect on the global repair rates. In contrast, incubation of AS52 cells with resveratrol at concentrations that caused significant chromatin compaction and that had only moderate influence on cell proliferation gave rise to a strong decrease of the repair rates of all three types of DNA modifications. Similar, but less pronounced effects were observed in HeLa cells. The effects of resveratrol on the repair rates were not antagonised by the sirtuin inhibitor EX-527 or by an increase of the intracellular thiol levels.
Collapse
Affiliation(s)
- Bettina Keuser
- Institute of Pharmacy and Biochemistry, University of Mainz, Staudingerweg 5, 55099 Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Nonnekens J, Cabantous S, Slingerland J, Mari PO, Giglia-Mari G. In vivo interactions of TTDA mutant proteins within TFIIH. J Cell Sci 2013; 126:3278-83. [PMID: 23729738 DOI: 10.1242/jcs.126839] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Trichothiodystrophy group A (TTD-A) patients carry a mutation in the transcription factor II H (TFIIH) subunit TTDA. Using a novel in vivo tripartite split-GFP system, we show that TTDA interacts with the TFIIH subunit p52 and the p52-TTDA-GFP product is incorporated into TFIIH. p52-TTDA-GFP is able to bind DNA and is recruited to UV-damaged DNA. Furthermore, we show that two patient-mutated TTDA proteins can interact with p52, are able to bind to the DNA and can localize to damaged DNA. Our findings give new insights into the behavior of TTDA within the context of a living cell and thereby shed light on the complex phenotype of TTD-A patients.
Collapse
|
40
|
Choi SY, Jang H, Roe JS, Kim ST, Cho EJ, Youn HD. Phosphorylation and ubiquitination-dependent degradation of CABIN1 releases p53 for transactivation upon genotoxic stress. Nucleic Acids Res 2013; 41:2180-90. [PMID: 23303793 PMCID: PMC3575827 DOI: 10.1093/nar/gks1319] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
CABIN1 acts as a negative regulator of p53 by keeping p53 in an inactive state on chromatin. Genotoxic stress causes rapid dissociation of CABIN1 and activation of p53. However, its molecular mechanism is still unknown. Here, we reveal the phosphorylation- and ubiquitination-dependent degradation of CABIN1 upon DNA damage, releasing p53 for transcriptional activation. The DNA-damage-signaling kinases, ATM and CHK2, phosphorylate CABIN1 and increase the degradation of CABIN1 protein. Knockdown or overexpression of these kinases influences the stability of CABIN1 protein showing that their activity is critical for degradation of CABIN1. Additionally, CABIN1 was found to undergo ubiquitin-dependent proteasomal degradation mediated by the CRL4DDB2 ubiquitin ligase complex. Both phosphorylation and ubiquitination of CABIN1 appear to be relevant for controlling the level of CABIN1 protein upon genotoxic stress.
Collapse
Affiliation(s)
- Soo-Youn Choi
- Department of Biomedical Sciences, Department of Biochemistry and Molecular Biology, National Creative Research Center for Epigenome Reprogramming Network, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
41
|
Pines A, Vrouwe MG, Marteijn JA, Typas D, Luijsterburg MS, Cansoy M, Hensbergen P, Deelder A, de Groot A, Matsumoto S, Sugasawa K, Thoma N, Vermeulen W, Vrieling H, Mullenders L. PARP1 promotes nucleotide excision repair through DDB2 stabilization and recruitment of ALC1. ACTA ACUST UNITED AC 2012; 199:235-49. [PMID: 23045548 PMCID: PMC3471223 DOI: 10.1083/jcb.201112132] [Citation(s) in RCA: 186] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PARP1-mediated poly(ADP-ribosyl)ation of DDB2 prolongs its occupation on UV-damaged chromatin and promotes the recruitment of the chromatin remodeler ALC1. The WD40-repeat protein DDB2 is essential for efficient recognition and subsequent removal of ultraviolet (UV)-induced DNA lesions by nucleotide excision repair (NER). However, how DDB2 promotes NER in chromatin is poorly understood. Here, we identify poly(ADP-ribose) polymerase 1 (PARP1) as a novel DDB2-associated factor. We demonstrate that DDB2 facilitated poly(ADP-ribosyl)ation of UV-damaged chromatin through the activity of PARP1, resulting in the recruitment of the chromatin-remodeling enzyme ALC1. Depletion of ALC1 rendered cells sensitive to UV and impaired repair of UV-induced DNA lesions. Additionally, DDB2 itself was targeted by poly(ADP-ribosyl)ation, resulting in increased protein stability and a prolonged chromatin retention time. Our in vitro and in vivo data support a model in which poly(ADP-ribosyl)ation of DDB2 suppresses DDB2 ubiquitylation and outline a molecular mechanism for PARP1-mediated regulation of NER through DDB2 stabilization and recruitment of the chromatin remodeler ALC1.
Collapse
Affiliation(s)
- Alex Pines
- Department of Toxicogenetics, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gaillard H, Aguilera A. Transcription coupled repair at the interface between transcription elongation and mRNP biogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1829:141-50. [PMID: 23046879 DOI: 10.1016/j.bbagrm.2012.09.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 09/19/2012] [Accepted: 09/22/2012] [Indexed: 01/13/2023]
Abstract
During transcription, the nascent pre-mRNA associates with mRNA-binding proteins and undergoes a series of processing steps, resulting in export competent mRNA ribonucleoprotein complexes (mRNPs) that are transported into the cytoplasm. Throughout transcription elongation, RNA polymerases frequently deal with a number of obstacles that need to be removed for transcription resumption. One important type of hindrance consists of helix-distorting DNA lesions. Transcription-coupled repair (TC-NER), a specific sub-pathway of nucleotide excision repair, ensures a fast repair of such transcription-blocking lesions. While the nucleotide excision repair reaction is fairly well understood, its regulation and the way it deals with DNA transcription remains largely unknown. In this review, we update our current understanding of the factors involved in TC-NER and discuss their functional interplay with the processes of transcription elongation and mRNP biogenesis. This article is part of a Special Issue entitled: RNA polymerase II Transcript Elongation.
Collapse
|
43
|
The emerging roles of ATP-dependent chromatin remodeling enzymes in nucleotide excision repair. Int J Mol Sci 2012; 13:11954-11973. [PMID: 23109894 PMCID: PMC3472786 DOI: 10.3390/ijms130911954] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 08/30/2012] [Accepted: 08/31/2012] [Indexed: 12/14/2022] Open
Abstract
DNA repair in eukaryotic cells takes place in the context of chromatin, where DNA, including damaged DNA, is tightly packed into nucleosomes and higher order chromatin structures. Chromatin intrinsically restricts accessibility of DNA repair proteins to the damaged DNA and impacts upon the overall rate of DNA repair. Chromatin is highly responsive to DNA damage and undergoes specific remodeling to facilitate DNA repair. How damaged DNA is accessed, repaired and restored to the original chromatin state, and how chromatin remodeling coordinates these processes in vivo, remains largely unknown. ATP-dependent chromatin remodelers (ACRs) are the master regulators of chromatin structure and dynamics. Conserved from yeast to humans, ACRs utilize the energy of ATP to reorganize packing of chromatin and control DNA accessibility by sliding, ejecting or restructuring nucleosomes. Several studies have demonstrated that ATP-dependent remodeling activity of ACRs plays important roles in coordination of spatio-temporal steps of different DNA repair pathways in chromatin. This review focuses on the role of ACRs in regulation of various aspects of nucleotide excision repair (NER) in the context of chromatin. We discuss current understanding of ATP-dependent chromatin remodeling by various subfamilies of remodelers and regulation of the NER pathway in vivo.
Collapse
|
44
|
Yao H, Rahman I. Role of histone deacetylase 2 in epigenetics and cellular senescence: implications in lung inflammaging and COPD. Am J Physiol Lung Cell Mol Physiol 2012; 303:L557-66. [PMID: 22842217 DOI: 10.1152/ajplung.00175.2012] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Histone deacetylase 2 (HDAC2) is a class I histone deacetylase that regulates various cellular processes, such as cell cycle, senescence, proliferation, differentiation, development, apoptosis, and glucocorticoid function in inhibiting inflammatory response. HDAC2 has been shown to protect against DNA damage response and cellular senescence/premature aging via an epigenetic mechanism in response to oxidative stress. These phenomena are observed in patients with chronic obstructive pulmonary disease (COPD). HDAC2 is posttranslationally modified by oxidative/carbonyl stress imposed by cigarette smoke and oxidants, leading to its reduction via an ubiquitination-proteasome dependent degradation in lungs of patients with COPD. In this perspective, we have discussed the role of HDAC2 posttranslational modifications and its role in regulation of inflammation, histone/DNA epigenetic modifications, DNA damage response, and cellular senescence, particularly in inflammaging, and during the development of COPD. We have also discussed the potential directions for future translational research avenues in modulating lung inflammaging and cellular senescence based on epigenetic chromatin modifications in diseases associated with increased oxidative stress.
Collapse
Affiliation(s)
- Hongwei Yao
- Dept. of Environmental Medicine, Lung Biology and Disease Program, Univ. of Rochester Medical Center, Rochester, NY 14642, USA
| | | |
Collapse
|
45
|
Perspectives on translational and therapeutic aspects of SIRT1 in inflammaging and senescence. Biochem Pharmacol 2012; 84:1332-9. [PMID: 22796566 DOI: 10.1016/j.bcp.2012.06.031] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 06/28/2012] [Accepted: 06/29/2012] [Indexed: 02/08/2023]
Abstract
Sirtuin1 (SIRT1), a type III protein deacetylase, is considered as a novel anti-aging protein involved in regulation of cellular senescence/aging and inflammation. SIRT1 level and activity are decreased during lung inflammaging caused by oxidative stress. The mechanism of SIRT1-mediated protection against inflammaging is associated with the regulation of inflammation, premature senescence, telomere attrition, senescence associated secretory phenotype, and DNA damage response. A variety of dietary polyphenols and pharmacological activators are shown to regulate SIRT1 so as to intervene the progression of type 2 diabetes, cancer, cardiovascular diseases, and chronic obstructive pulmonary disease associated with inflammaging. However, recent studies have shown the non-specific regulation of SIRT1 by the aforementioned pharmacological activators and polyphenols. In this perspective, we have briefly discussed the role of SIRT1 in regulation of cellular senescence and its associated secretory phenotype, DNA damage response, particularly in lung inflammaging and during the development of chronic obstructive pulmonary diseases. We have also discussed the potential directions for future translational therapeutic avenues for SIRT1 in modulating lung inflammaging associated with senescence in chronic lung diseases associated with increased oxidative stress.
Collapse
|
46
|
Luijsterburg MS, Lindh M, Acs K, Vrouwe MG, Pines A, van Attikum H, Mullenders LH, Dantuma NP. DDB2 promotes chromatin decondensation at UV-induced DNA damage. ACTA ACUST UNITED AC 2012; 197:267-81. [PMID: 22492724 PMCID: PMC3328393 DOI: 10.1083/jcb.201106074] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In addition to its role in DNA lesion recognition, the damaged DNA-binding protein DDB2 elicits unfolding of large-scale chromatin structure independently of the CRL4 ubiquitin ligase complex. Nucleotide excision repair (NER) is the principal pathway that removes helix-distorting deoxyribonucleic acid (DNA) damage from the mammalian genome. Recognition of DNA lesions by xeroderma pigmentosum group C (XPC) protein in chromatin is stimulated by the damaged DNA-binding protein 2 (DDB2), which is part of a CUL4A–RING ubiquitin ligase (CRL4) complex. In this paper, we report a new function of DDB2 in modulating chromatin structure at DNA lesions. We show that DDB2 elicits unfolding of large-scale chromatin structure independently of the CRL4 ubiquitin ligase complex. Our data reveal a marked adenosine triphosphate (ATP)–dependent reduction in the density of core histones in chromatin containing UV-induced DNA lesions, which strictly required functional DDB2 and involved the activity of poly(adenosine diphosphate [ADP]–ribose) polymerase 1. Finally, we show that lesion recognition by XPC, but not DDB2, was strongly reduced in ATP-depleted cells and was regulated by the steady-state levels of poly(ADP-ribose) chains.
Collapse
Affiliation(s)
- Martijn S Luijsterburg
- Department of Cell and Molecular Biology, Karolinska Institutet, S-17177 Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lan L, Nakajima S, Kapetanaki MG, Hsieh CL, Fagerburg M, Thickman K, Rodriguez-Collazo P, Leuba SH, Levine AS, Rapić-Otrin V. Monoubiquitinated histone H2A destabilizes photolesion-containing nucleosomes with concomitant release of UV-damaged DNA-binding protein E3 ligase. J Biol Chem 2012; 287:12036-49. [PMID: 22334663 PMCID: PMC3320950 DOI: 10.1074/jbc.m111.307058] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
How the nucleotide excision repair (NER) machinery gains access to damaged chromatinized DNA templates and how the chromatin structure is modified to promote efficient repair of the non-transcribed genome remain poorly understood. The UV-damaged DNA-binding protein complex (UV-DDB, consisting of DDB1 and DDB2, the latter of which is mutated in xeroderma pigmentosum group E patients, is a substrate-recruiting module of the cullin 4B-based E3 ligase complex, DDB1-CUL4BDDB2. We previously reported that the deficiency of UV-DDB E3 ligases in ubiquitinating histone H2A at UV-damaged DNA sites in the xeroderma pigmentosum group E cells contributes to the faulty NER in these skin cancer-prone patients. Here, we reveal the mechanism by which monoubiquitination of specific H2A lysine residues alters nucleosomal dynamics and subsequently initiates NER. We show that DDB1-CUL4BDDB2 E3 ligase specifically binds to mononucleosomes assembled with human recombinant histone octamers and nucleosome-positioning DNA containing cyclobutane pyrimidine dimers or 6-4 photoproducts photolesions. We demonstrate functionally that ubiquitination of H2A Lys-119/Lys-120 is necessary for destabilization of nucleosomes and concomitant release of DDB1-CUL4BDDB2 from photolesion-containing DNA. Nucleosomes in which these lysines are replaced with arginines are resistant to such structural changes, and arginine mutants prevent the eviction of H2A and dissociation of polyubiquitinated DDB2 from UV-damaged nucleosomes. The partial eviction of H3 from the nucleosomes is dependent on ubiquitinated H2A Lys-119/Lys-120. Our results provide mechanistic insight into how post-translational modification of H2A at the site of a photolesion initiates the repair process and directly affects the stability of the human genome.
Collapse
Affiliation(s)
- Li Lan
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Triptolide induces cell-cycle arrest and apoptosis of human multiple myeloma cells in vitro via altering expression of histone demethylase LSD1 and JMJD2B. Acta Pharmacol Sin 2012; 33:109-19. [PMID: 22120968 DOI: 10.1038/aps.2011.145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM To elucidate the relationship between triptolide-induced changes in histone methylation and its antitumor effect on human multiple myeloma (MM) cells in vitro. METHODS Human multiple myeloma cell line RPMI8226 was used. Apoptosis was evaluated using Annexin-V-FITC/PI-labeled flow cytometry, Hoechst 33258 staining, and transmission electron microscopy. Flow cytometry was used to detect the cell cycle distribution of the apoptotic cells. The presence of the LSD1, JMJD2B, H3K4me2, H3K9me2, and H3K36me2 proteins was verified by Western blot analysis. Semi-quantitative real-time PCR was performed to examine the expression of LSD1 and JMJD2B. RESULTS Triptolide (10-160 nmol/L) suppressed the proliferation of MM cells in a dose- and time-dependent manner with an IC(50) value of 99.2 ± 9.0 nmol/L at 24 h. Triptolide (50 nmol/L) induced G(0)/G(1) cell cycle arrest in MM cells. The agent (50-150 nmol/L) induced apoptosis of MM cells in a dose-dependent manner. The same concentrations of triptolide suppressed the expression of dimethylated H3K4, dimethylated H3K9 and dimethylated H3K36 by altering the expression of histone demethylase LSD1 and JMJD2B without affecting the expression of histone demethylase LSD1. CONCLUSION Triptolide potently inhibits the growth of MM cells via regulating the expression of histone demethylase LSD1 and JMJD2B, which lead to abnormal histone methylation.
Collapse
|
49
|
Farrell AW, Halliday GM, Lyons JG. Chromatin structure following UV-induced DNA damage-repair or death? Int J Mol Sci 2011; 12:8063-85. [PMID: 22174650 PMCID: PMC3233456 DOI: 10.3390/ijms12118063] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 10/05/2011] [Accepted: 10/31/2011] [Indexed: 12/15/2022] Open
Abstract
In eukaryotes, DNA is compacted into a complex structure known as chromatin. The unravelling of DNA is a crucial step in DNA repair, replication, transcription and recombination as this allows access to DNA for these processes. Failure to package DNA into the nucleosome, the individual unit of chromatin, can lead to genomic instability, driving a cell into apoptosis, senescence, or cellular proliferation. Ultraviolet (UV) radiation damage causes destabilisation of chromatin integrity. UV irradiation induces DNA damage such as photolesions and subjects the chromatin to substantial rearrangements, causing the arrest of transcription forks and cell cycle arrest. Highly conserved processes known as nucleotide and base excision repair (NER and BER) then begin to repair these lesions. However, if DNA repair fails, the cell may be forced into apoptosis. The modification of various histones as well as nucleosome remodelling via ATP-dependent chromatin remodelling complexes are required not only to repair these UV-induced DNA lesions, but also for apoptosis signalling. Histone modifications and nucleosome remodelling in response to UV also lead to the recruitment of various repair and pro-apoptotic proteins. Thus, the way in which a cell responds to UV irradiation via these modifications is important in determining its fate. Failure of these DNA damage response steps can lead to cellular proliferation and oncogenic development, causing skin cancer, hence these chromatin changes are critical for a proper response to UV-induced injury.
Collapse
Affiliation(s)
- Andrew W Farrell
- Discipline of Dermatology, Bosch Institute, Sydney Cancer Centre, The University of Sydney, NSW 2006, Australia; E-Mails: (A.W.F.); (J.G.L.)
| | | | | |
Collapse
|
50
|
Khan HA, Alhomida AS. A review of the logistic role of L-carnitine in the management of radiation toxicity and radiotherapy side effects. J Appl Toxicol 2011; 31:707-13. [PMID: 21818761 DOI: 10.1002/jat.1716] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 06/10/2011] [Accepted: 06/10/2011] [Indexed: 02/05/2023]
Abstract
Radiation therapy is a key modality in the treatment of different cancer types. Fatigue is the most common side effect of radiotherapy, while others include nausea, hair loss, skin irritation, anemia, infertility, cardiovascular disease, cognitive impairment and even the development of second cancers. Studies in experimental animals have shown protective effects of carnitine against exposure of various organs to ionizing radiation, whereas carnitine deficiency is known to enhance radiation-induced toxicity. This report summarizes the recent literature on the adverse effects of radiotherapy and the impact of radiation on carnitine homeostasis. Although some studies have demonstrated the prophylactic benefits of carnitine against the toxic effects of chemotherapy, the role of carnitine in the prognosis and management of cancer patients receiving radiotherapy is not clear and needs to be explored.
Collapse
Affiliation(s)
- Haseeb Ahmad Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia.
| | | |
Collapse
|