DNA Mismatch Repair in the Context of Chromatin Yaping Huang* and Guo‑Min Li

Total Page:16

File Type:pdf, Size:1020Kb

DNA Mismatch Repair in the Context of Chromatin Yaping Huang* and Guo‑Min Li Huang and Li Cell Biosci (2020) 10:10 https://doi.org/10.1186/s13578-020-0379-7 Cell & Bioscience REVIEW Open Access DNA mismatch repair in the context of chromatin Yaping Huang* and Guo‑Min Li Abstract DNA mismatch repair (MMR) maintains replication fdelity by correcting mispaired nucleotides incorporated by DNA polymerases. Defects in MMR lead to cancers characterized by microsatellite instability. Recently, chromatin mecha‑ nisms that regulate MMR have been discovered, which sheds new light on MMR defciency and its role in tumorigen‑ esis. This review summarizes these chromatin‑level mechanisms that regulate MMR and their implications for tumor development. Background proteins and their modifcations. Indeed, recent studies DNA mismatch repair (MMR) maintains replication have indicated that trimethylation of histone H3 lysine 36 fdelity in a replication-coupled manner, primarily by cor- (H3K36me3) plays a role in MMR by recruiting MutSα to recting misincorporated nucleotides in the nascent DNA replicating chromatin [5]. In addition, chromatin assem- strand [1–4]. Te typical MMR reaction in human cells bly/remodeling factors also interact with MMR proteins comprises three major steps. First, the mismatch rec- to coordinate MMR and nucleosome formation [6, 7]. ognition protein MutSα (MSH2–MSH6 heterodimer) Loss-of-function mutations or promoter hyper-methyl- or MutSβ (MSH2–MSH3 heterodimer) recognizes the ation of MMR genes, such as MSH2 and MLH1, increase mismatch, which triggers concerted interactions with susceptibility to cancers, including hereditary non-poly- proliferating cell nuclear antigen (PCNA) and MutLα posis colorectal cancer (HNPCC), also called lynch syn- (MLH1–PMS2 heterodimer), leading to the recruitment drome [1, 2, 4, 8, 9]. Te demonstration of H3K36me3 as of exonuclease 1 (EXO1) to a single-strand DNA break. an essential component for MMR in vivo [5] may provide Ten, Exo1-catalyzed DNA excises the mispaired base new insights into MMR defciency and cancer suscep- from the nick up to and beyond the mismatch in a man- tibility in the chromatin context. H3K36me3 is a well- ner dependent on MutSα (or MutSβ), MutLα, and repli- known histone post-translational modifcation mark, and cation protein A (RPA). Finally, the DNA gap is flled by its cellular level is regulated by its trimethyltransferase DNA polymerase δ in the presence of PCNA, RPA, and SETD2 and lysine demethylase KDM4 [10]. Inappropri- replication factor C (RFC), followed by DNA ligase I-cat- ate expression of and/or defects in these histone writer alyzed nick ligation. and eraser genes probably infuence cellular H3K36me3 In eukaryotes, DNA is wrapped around histone octam- levels, leading to the loss of MMR function. ers, which, together with DNA, compose nucleosomes Recent studies have also shown that mutations in his- to form chromatin. Tus, all DNA metabolic reactions, tone 3 (H3), particularly H3K36M/I and H3G34V/R, including MMR, are precisely regulated by the struc- which frequently occur in various tumors [11–14], afect tures of chromatin, particularly its component histone H3K36me3 expression levels [15, 16]. Tis review will focus on the most recent developments in the feld con- *Correspondence: [email protected] cerning the regulation of MMR in the context of chroma- Department of Radiation Oncology, University of Texas Southwestern tin and its association with cancer susceptibility. Readers Medical Center, Dallas, TX 75390, USA © The Author(s) 2020. This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://crea‑ tivecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdo‑ main/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data. Huang and Li Cell Biosci (2020) 10:10 Page 2 of 8 are also referred to several recent reviews in this area PWWP domain. Tus, yeast MutSα must be recruited [17–20]. by a histone mark diferent from H3K36me3, which is consistent with the report that set2 defciency doesn’t Main text infuence mutation frequency in yeast [25]. A recent H3K36me3 distribution dictates local mutation frequency study revealed that the PWWP domain of MSH6 in Cap- Te biochemistry of MMR is essentially well established, sella rubella interacts better with H3K4me3 than with but how the MMR system is regulated in the chromatin H3K36me3 [26], suggesting that H3K4me3, rather than context is not fully understood. In 2013, Li et al. [5] iden- H3K36me3, is the preferred histone mark for loading tifed H3K36me3 as an important MMR regulator in vivo MutSα to chromatin in plants. Te diference in MutSα by recruiting mismatch recognition protein MutSα to recruitment among diferent organisms may suggest an replicating chromatin through its physical interaction adapted evolution for MMR regulation in the chromatin with the PWWP domain of human MutSα. A recent context, although the canonical MMR function is highly chromatin immunoprecipitation followed by sequencing conserved. Terefore, additional studies are needed to (ChIP-Seq) study [21] has illustrated the genome-wide determine why diferent systems use diferent mecha- distribution of H3K36me3 and MutSα in HeLa cells. Both nisms for MutSα recruitment. H3K36me3 and MutSα are unevenly distributed in chro- matin [10, 21]. H3K36me3 and MutSα are more enriched Chromatin remodeling in MMR in euchromatin, exons, and 3′ gene bodies than in het- MMR occurs in the chromatin context, where nucleo- erochromatin, introns, and 5′ gene bodies, respectively. some obstacles must be handled for efcient repair Consistent with MMR’s genome maintenance role, the before the newly synthesized mismatch-containing H3K36me3–MutSα distribution is inversely correlated DNA is packaged into nucleosomes. Tus, MMR needs with mutation frequencies in these genomic regions, as to coordinate with chromatin remodeling factors and/or the mutation frequencies in euchromatin, exons, and 3′ nucleosome assembly factors to repair misincorporated gene bodies are much lower than in heterochromatin, nucleotides (Fig. 1). CAF-1 is the most studied nucleo- introns, and 5′ gene bodies, respectively. In other words, some assembly factor involved in MMR during replica- the abundance of H3K36me3 and MutSα is inversely cor- tion. CAF-1 directly interacts with MMR components related to the local mutation frequency [21]. MutSα and PCNA, and MutSα inhibits CAF-1-mediated It is known that replication timing determines mutation chromatin assembly [27]. Recent biochemical studies frequency: early replicating genes exhibit lower mutation further indicate that MutSα inhibits CAF-1- and ASF1A- frequencies than late replicating genes. Tis phenomenon H3-H4-dependent packaging of DNA mismatches into can be well explained by the distribution and enrichment (H3–H4)2 tetramers [28]. Tese studies support the idea of H3K36me3/MutSα in chromatin. Analyzing the rela- that MMR occurs before the mismatch is packaged into tionship between replication timing, H3K36me3/MutSα nucleosomes. In addition, CAF-1- and ASF1A-H3-H4- enrichment, and mutation frequency revealed that early dependent nucleosome assembly quickly represses the replicating chromatin regions are highly enriched for unnecessary degradation of the discontinuous strand [7, H3K36me3/MutSα and display a lower mutation fre- 28]. Tus, nucleosome assembly is compatible with MMR quency than late replicating regions, which contain fewer during replication. In contrast to CAF-1, the chromatin H3K36me3/MutSα signals [21]. Tese observations sug- remodeler Smarcad1 is recruited by MSH2 to mismatch- gest that H3K36me3-mediated MMR ensures the replica- containing DNA to exclude nucleosomes from the repair tion accuracy of early replicating genes/chromatin, where site [29]. It is interesting to note that, although MSH2, actively transcribed genes are located. In addition to safe- a subunit of MutSα, can be recruited to chromatin via guarding actively transcribed genes during DNA replica- H3K36me3 [5], a recent study showed that ARID1A, tion, H3K36me3-mediated MMR also appears to protect a subunit of the chromatin remodeling complex SWI/ these genes during transcription by directly or indirectly SNF, recruits MSH2 to the chromatin during replica- removing DNA lesions associated with transcription tion through direct interaction [30]. Histone variants [21]. However, how H3K36me3 regulates MMR in difer- also play important roles in DNA transactions, in addi- ent DNA transactions, e.g., replication and transcription, tion to nucleosome assembly and chromatin remodeling. remains to be investigated. It was reported that deposition of H2A.Z by chromatin It is worth noting that not all eukaryotic cells use remodeling enzyme SWR-C stimulates Exo1 activity and H3K36me3
Recommended publications
  • Paul Modrich Howard Hughes Medical Institute and Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, USA
    Mechanisms in E. coli and Human Mismatch Repair Nobel Lecture, December 8, 2015 by Paul Modrich Howard Hughes Medical Institute and Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, USA. he idea that mismatched base pairs occur in cells and that such lesions trig- T ger their own repair was suggested 50 years ago by Robin Holliday in the context of genetic recombination [1]. Breakage and rejoining of DNA helices was known to occur during this process [2], with precision of rejoining attributed to formation of a heteroduplex joint, a region of helix where the two strands are derived from the diferent recombining partners. Holliday pointed out that if this heteroduplex region should span a genetic diference between the two DNAs, then it will contain one or more mismatched base pairs. He invoked processing of such mismatches to explain the recombination-associated phenomenon of gene conversion [1], noting that “If there are enzymes which can repair points of damage in DNA, it would seem possible that the same enzymes could recognize the abnormality of base pairing, and by exchange reactions rectify this.” Direct evidence that mismatches provoke a repair reaction was provided by bacterial transformation experiments [3–5], and our interest in this efect was prompted by the Escherichia coli (E. coli) work done in Matt Meselson’s lab at Harvard. Using artifcially constructed heteroduplex DNAs containing multiple mismatched base pairs, Wagner and Meselson [6] demonstrated that mismatches elicit a repair reaction upon introduction into the E. coli cell. Tey also showed that closely spaced mismatches, mismatches separated by a 1000 base pairs or so, are usually repaired on the same DNA strand.
    [Show full text]
  • Recognition of Cancer Mutations in Histone H3K36 by Epigenetic Writers and Readers Brianna J
    EPIGENETICS https://doi.org/10.1080/15592294.2018.1503491 REVIEW Recognition of cancer mutations in histone H3K36 by epigenetic writers and readers Brianna J. Kleina, Krzysztof Krajewski b, Susana Restrepoa, Peter W. Lewis c, Brian D. Strahlb, and Tatiana G. Kutateladzea aDepartment of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA; bDepartment of Biochemistry & Biophysics, The University of North Carolina School of Medicine, Chapel Hill, NC, USA; cWisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA ABSTRACT ARTICLE HISTORY Histone posttranslational modifications control the organization and function of chromatin. In Received 30 May 2018 particular, methylation of lysine 36 in histone H3 (H3K36me) has been shown to mediate gene Revised 1 July 2018 transcription, DNA repair, cell cycle regulation, and pre-mRNA splicing. Notably, mutations at or Accepted 12 July 2018 near this residue have been causally linked to the development of several human cancers. These KEYWORDS observations have helped to illuminate the role of histones themselves in disease and to clarify Histone; H3K36M; cancer; the mechanisms by which they acquire oncogenic properties. This perspective focuses on recent PTM; methylation advances in discovery and characterization of histone H3 mutations that impact H3K36 methyla- tion. We also highlight findings that the common cancer-related substitution of H3K36 to methionine (H3K36M) disturbs functions of not only H3K36me-writing enzymes but also H3K36me-specific readers. The latter case suggests that the oncogenic effects could also be linked to the inability of readers to engage H3K36M. Introduction from yeast to humans and has been shown to have a variety of functions that range from the control Histone proteins are main components of the of gene transcription and DNA repair, to cell cycle nucleosome, the fundamental building block of regulation and nutrient stress response [8].
    [Show full text]
  • DNA Repair with Its Consequences (E.G
    Cell Science at a Glance 515 DNA repair with its consequences (e.g. tolerance and pathways each require a number of apoptosis) as well as direct correction of proteins. By contrast, O-alkylated bases, Oliver Fleck* and Olaf Nielsen* the damage by DNA repair mechanisms, such as O6-methylguanine can be Department of Genetics, Institute of Molecular which may require activation of repaired by the action of a single protein, Biology, University of Copenhagen, Øster checkpoint pathways. There are various O6-methylguanine-DNA Farimagsgade 2A, DK-1353 Copenhagen K, Denmark forms of DNA damage, such as base methyltransferase (MGMT). MGMT *Authors for correspondence (e-mail: modifications, strand breaks, crosslinks removes the alkyl group in a suicide fl[email protected]; [email protected]) and mismatches. There are also reaction by transfer to one of its cysteine numerous DNA repair pathways. Each residues. Photolyases are able to split Journal of Cell Science 117, 515-517 repair pathway is directed to specific Published by The Company of Biologists 2004 covalent bonds of pyrimidine dimers doi:10.1242/jcs.00952 types of damage, and a given type of produced by UV radiation. They bind to damage can be targeted by several a UV lesion in a light-independent Organisms are permanently exposed to pathways. Major DNA repair pathways process, but require light (350-450 nm) endogenous and exogenous agents that are mismatch repair (MMR), nucleotide as an energy source for repair. Another damage DNA. If not repaired, such excision repair (NER), base excision NER-independent pathway that can damage can result in mutations, diseases repair (BER), homologous recombi- remove UV-induced damage, UVER, is and cell death.
    [Show full text]
  • Complex Interacts with WRN and Is Crucial to Regulate Its Response to Replication Fork Stalling
    Oncogene (2012) 31, 2809–2823 & 2012 Macmillan Publishers Limited All rights reserved 0950-9232/12 www.nature.com/onc ORIGINAL ARTICLE The RAD9–RAD1–HUS1 (9.1.1) complex interacts with WRN and is crucial to regulate its response to replication fork stalling P Pichierri, S Nicolai, L Cignolo1, M Bignami and A Franchitto Department of Environment and Primary Prevention, Istituto Superiore di Sanita`, Rome, Italy The WRN protein belongs to the RecQ family of DNA preference toward substrates that mimic structures helicases and is implicated in replication fork restart, but associated with stalled replication forks (Brosh et al., how its function is regulated remains unknown. We show 2002; Machwe et al., 2006) and WS cells exhibit that WRN interacts with the 9.1.1 complex, one of enhanced instability at common fragile sites, chromo- the central factors of the replication checkpoint. This somal regions especially prone to replication fork interaction is mediated by the binding of the RAD1 stalling (Pirzio et al., 2008). How WRN favors recovery subunit to the N-terminal region of WRN and is of stalled forks and prevents DNA breakage upon instrumental for WRN relocalization in nuclear foci and replication perturbation is not fully understood. It has its phosphorylation in response to replication arrest. We been suggested that WRN might facilitate replication also find that ATR-dependent WRN phosphorylation restart by either promoting recombination or processing depends on TopBP1, which is recruited by the 9.1.1 intermediates at stalled forks in a way that counteracts complex in response to replication arrest. Finally, we unscheduled recombination (Franchitto and Pichierri, provide evidence for a cooperation between WRN and 2004; Pichierri, 2007; Sidorova, 2008).
    [Show full text]
  • Genome Instability in Secondary Solid Tumors Developing After Radiotherapy of Bilateral Retinoblastoma
    Oncogene (2001) 20, 8092 ± 8099 ã 2001 Nature Publishing Group All rights reserved 0950 ± 9232/01 $15.00 www.nature.com/onc Genome instability in secondary solid tumors developing after radiotherapy of bilateral retinoblastoma Sandrine-He leÁ ne LefeÁ vre1, Nicolas Vogt1, Anne-Marie Dutrillaux1, Laurent Chauveinc2, Dominique Stoppa-Lyonnet3, FrancËois Doz4, Laurence Desjardins5, Bernard Dutrillaux1,6, Sylvie Chevillard6 and Bernard Malfoy*,1 1Institut Curie ± CNRS UMR 147, 26 rue d'Ulm 75248 Paris Cedex 05, France; 2Institut Curie, Service de RadiotheÂrapie, 26 rue d'Ulm 75248 Paris Cedex 05, France; 3Institut Curie, Service de GeÂneÂtique Oncologique, 26 rue d'Ulm 75248 Paris Cedex 05, France; 4Institut Curie, Service de PeÂdiatrie, 26 rue d'Ulm 75248 Paris Cedex 05, France; 5Institut Curie, Service d'Ophtalmologie, 26 rue d'Ulm 75248 Paris Cedex 05, France; 6CEA, DSV DRR, 60 avenue du GeÂneÂral Leclerc 92265 Fontenay-aux-Roses, France Genome alterations of seven secondary tumors (®ve these cancers and the diculty in collecting a series of osteosarcomas, one malignant peripheral sheath nerve cases in clearly de®ned context. tumor, one leiomyosarcoma) occurring in the ®eld of It is well established that therapeutic irradiation can irradiation of patients treated for bilateral retinoblasto- induce secondary malignancies within or at the margin ma have been studied. These patients were predisposed to of the radiation ®eld after a long latent period. These develop radiation-induced tumors because of the presence tumors have dierent histology from the primary of a germ line mutation in the retinoblastoma gene lesions, with sarcomas being common (Robinson et (RB1). Tumor cells were characterized by a high al., 1988).
    [Show full text]
  • Genomic Instability Promoted by Overexpression of Mismatch Repair Factors in Yeast: a Model for Understanding Cancer Progression
    HIGHLIGHTED ARTICLE | INVESTIGATION Genomic Instability Promoted by Overexpression of Mismatch Repair Factors in Yeast: A Model for Understanding Cancer Progression Ujani Chakraborty,* Timothy A. Dinh,† and Eric Alani*,1 *Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853-2703 and †Curriculum in Genetics and Molecular Biology, Biological and Biomedical Sciences Program, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599 ORCID ID: 0000-0002-5011-9339 (E.A.) ABSTRACT Mismatch repair (MMR) proteins act in spellchecker roles to excise misincorporation errors that occur during DNA replication. Curiously, large-scale analyses of a variety of cancers showed that increased expression of MMR proteins often correlated with tumor aggressiveness, metastasis, and early recurrence. To better understand these observations, we used The Cancer Genome Atlas and Gene Expression across Normal and Tumor tissue databases to analyze MMR protein expression in cancers. We found that the MMR genes MSH2 and MSH6 are overexpressed more frequently than MSH3, and that MSH2 and MSH6 are often cooverex- pressed as a result of copy number amplifications of these genes. These observations encouraged us to test the effects of upregulating MMR protein levels in baker’s yeast, where we can sensitively monitor genome instability phenotypes associated with cancer initiation and progression. Msh6 overexpression (two- to fourfold) almost completely disrupted mechanisms that prevent recombination be- tween divergent DNA sequences by interacting with the DNA polymerase processivity clamp PCNA and by sequestering the Sgs1 helicase. Importantly, cooverexpression of Msh2 and Msh6 (eightfold) conferred, in a PCNA interaction-dependent manner, several genome instability phenotypes including increased mutation rate, increased sensitivity to the DNA replication inhibitor HU and the DNA-damaging agents MMS and 4-nitroquinoline N-oxide, and elevated loss-of-heterozygosity.
    [Show full text]
  • Screening for Genes That Accelerate the Epigenetic Aging Clock in Humans Reveals a Role for the H3K36 Methyltransferase NSD1 Daniel E
    Martin-Herranz et al. Genome Biology (2019) 20:146 https://doi.org/10.1186/s13059-019-1753-9 RESEARCH Open Access Screening for genes that accelerate the epigenetic aging clock in humans reveals a role for the H3K36 methyltransferase NSD1 Daniel E. Martin-Herranz1,2* , Erfan Aref-Eshghi3,4, Marc Jan Bonder1,5, Thomas M. Stubbs2, Sanaa Choufani6, Rosanna Weksberg6, Oliver Stegle1,5,7, Bekim Sadikovic3,4, Wolf Reik8,9,10*† and Janet M. Thornton1*† Abstract Background: Epigenetic clocks are mathematical models that predict the biological age of an individual using DNA methylation data and have emerged in the last few years as the most accurate biomarkers of the aging process. However, little is known about the molecular mechanisms that control the rate of such clocks. Here, we have examined the human epigenetic clock in patients with a variety of developmental disorders, harboring mutations in proteins of the epigenetic machinery. Results: Using the Horvath epigenetic clock, we perform an unbiased screen for epigenetic age acceleration in the blood of these patients. We demonstrate that loss-of-function mutations in the H3K36 histone methyltransferase NSD1, which cause Sotos syndrome, substantially accelerate epigenetic aging. Furthermore, we show that the normal aging process and Sotos syndrome share methylation changes and the genomic context in which they occur. Finally, we found that the Horvath clock CpG sites are characterized by a higher Shannon methylation entropy when compared with the rest of the genome, which is dramatically decreased in Sotos syndrome patients. Conclusions: These results suggest that the H3K36 methylation machinery is a key component of the epigenetic maintenance system in humans, which controls the rate of epigenetic aging, and this role seems to be conserved in model organisms.
    [Show full text]
  • Histone H3 Lysine 36 Methylation Affects Temperature-Induced Alternative Splicing and Flowering in Plants A
    Pajoro et al. Genome Biology (2017) 18:102 DOI 10.1186/s13059-017-1235-x RESEARCH Open Access Histone H3 lysine 36 methylation affects temperature-induced alternative splicing and flowering in plants A. Pajoro1,2, E. Severing3,4, G. C. Angenent1,2 and R. G. H. Immink1,2* Abstract Background: Global warming severely affects flowering time and reproductive success of plants. Alternative splicing of pre-messenger RNA (mRNA) is an important mechanism underlying ambient temperature-controlled responses in plants, yet its regulation is poorly understood. An increase in temperature promotes changes in plant morphology as well as the transition from the vegetative to the reproductive phase in Arabidopsis thaliana via changes in splicing of key regulatory genes. Here we investigate whether a particular histone modification affects ambient temperature-induced alternative splicing and flowering time. Results: We use a genome-wide approach and perform RNA-sequencing (RNA-seq) analyses and histone H3 lysine 36 tri-methylation (H3K36me3) chromatin immunoprecipitation sequencing (ChIP-seq) in plants exposed to different ambient temperatures. Analysis and comparison of these datasets reveal that temperature-induced differentially spliced genes are enriched in H3K36me3. Moreover, we find that reduction of H3K36me3 deposition causes alteration in temperature-induced alternative splicing. We also show that plants with mutations in H3K36me3 writers, eraser, or readers have altered high ambient temperature-induced flowering. Conclusions: Our results show a key role for the histone mark H3K36me3 in splicing regulation and plant plasticity to fluctuating ambient temperature. Our findings open new perspectives for the breeding of crops that can better cope with environmental changes due to climate change.
    [Show full text]
  • Clinical Significance of ERCC2, XPC, ERCC5 and XRCC3 Gene Polymorphisms in Diffuse Large B Cell Lymphoma
    DOI: 10.14744/ejmi.2020.56831 EJMI 2020;4(3):332–340 Research Article Clinical Significance of ERCC2, XPC, ERCC5 and XRCC3 Gene Polymorphisms in Diffuse Large B Cell Lymphoma Aykut Bahceci,1 Semra Paydas,2 Melek Ergin,3 Gulsah Seydaoglu,4 Gulsum Ucar5 1Department of Medical Oncology, Dr. Ersin Arslan Training and Research Hospital, Gaziantep, Turkey 2Department of Medical Oncology, Cukurova University Faculty of Medicine, Adana, Turkey 3Department of Patology, Cukurova University Faculty of Medicine, Adana, Turkey 4Department of Biostatistics, Cukurova University Faculty of Medicine, Adana, Turkey 5Department of Pediatric Hematology, Cukurova University Faculty of Medicine, Adana, Turkey Abstract Objectives: DNA repair genes protects the genome from DNA damage both of endogenous and exogenous stress fac- tors. Due to DNA repair gene polymorphisms, there are differences in the repair capacity between several cancer types. The aim of this study is to evaluate the association between some of the DNA repair gene polymorphisms and clinical outcome in Diffuse Large B-Cell Lymphoma (DLBCL). Methods: The association between clinical factors including stage at diagnosis, extra-nodal involvement, tumor bur- den, bone marrow involvement, relapse status, disease-free/overall survival times and DNA repair gene polymorphisms including ERCC2 (Lys751Gln), XPC (Gln939Lys), ERCC5 (Asp1104His) and XRCC3 (Thr241Met) in 58 patients with DLBCL. T-Shift Real-Time PCR was used to detect these mutations. Results: The median survival times were 60 months and 109 months in patients with CC genotype and CA/AA geno- type of XPC gene polymorphism, respectively (p=0.017). More interestingly, median survival times were 9 months and 109 months in patients with CC (XPC)/CC (XRCC3) and CA/AA (XPC)/CT/TT (XRCC3) for both XPC and XRCC3 gene polymorphisms, respectively (p=0.004).
    [Show full text]
  • Dynamics of Transcription-Dependent H3k36me3 Marking by the SETD2:IWS1:SPT6 Ternary Complex
    bioRxiv preprint doi: https://doi.org/10.1101/636084; this version posted May 14, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Dynamics of transcription-dependent H3K36me3 marking by the SETD2:IWS1:SPT6 ternary complex Katerina Cermakova1, Eric A. Smith1, Vaclav Veverka2, H. Courtney Hodges1,3,4,* 1 Department of Molecular & Cellular Biology, Center for Precision Environmental Health, and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA 2 Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic 3 Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA 4 Department of Bioengineering, Rice University, Houston, TX, 77005, USA * Lead contact; Correspondence to: [email protected] Abstract The genome-wide distribution of H3K36me3 is maintained SETD2 contributes to gene expression by marking gene through various mechanisms. In human cells, H3K36 is bodies with H3K36me3, which is thought to assist in the mono- and di-methylated by eight distinct histone concentration of transcription machinery at the small portion methyltransferases; however, the predominant writer of the of the coding genome. Despite extensive genome-wide data trimethyl mark on H3K36 is SETD21,11,12. Interestingly, revealing the precise localization of H3K36me3 over gene SETD2 is a major tumor suppressor in clear cell renal cell bodies, the physical basis for the accumulation, carcinoma13, breast cancer14, bladder cancer15, and acute maintenance, and sharp borders of H3K36me3 over these lymphoblastic leukemias16–18. In these settings, mutations sites remains rudimentary.
    [Show full text]
  • Origin of Genome Instability and Determinants of Mutational Landscape in Cancer Cells
    G C A T T A C G G C A T genes Review Origin of Genome Instability and Determinants of Mutational Landscape in Cancer Cells Sonam Mehrotra * and Indraneel Mittra Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; [email protected] * Correspondence: [email protected]; Tel.: +91-22-27405143 Received: 15 August 2020; Accepted: 18 September 2020; Published: 21 September 2020 Abstract: Genome instability is a crucial and early event associated with an increased predisposition to tumor formation. In the absence of any exogenous agent, a single human cell is subjected to about 70,000 DNA lesions each day. It has now been shown that physiological cellular processes including DNA transactions during DNA replication and transcription contribute to DNA damage and induce DNA damage responses in the cell. These processes are also influenced by the three dimensional-chromatin architecture and epigenetic regulation which are altered during the malignant transformation of cells. In this review, we have discussed recent insights about how replication stress, oncogene activation, chromatin dynamics, and the illegitimate recombination of cell-free chromatin particles deregulate cellular processes in cancer cells and contribute to their evolution. The characterization of such endogenous sources of genome instability in cancer cells can be exploited for the development of new biomarkers and more effective therapies for cancer treatment. Keywords: genome instability; replication stress; replication-transcription conflict; cell free chromatin; cancer 1. Introduction Genome instability is a characteristic feature observed in most human cancers. The accumulation of genetic alterations ranging from single nucleotide mutations to chromosome rearrangements can predispose cells towards malignancy.
    [Show full text]
  • Predisposition to Hematologic Malignancies in Patients With
    LETTERS TO THE EDITOR carcinomas but no internal cancer by the age of 29 years Predisposition to hematologic malignancies in and 9 years, respectively. patients with xeroderma pigmentosum Case XP540BE . This patient had a highly unusual pres - entation of MPAL. She was diagnosed with XP at the age Germline predisposition is a contributing etiology of of 18 months with numerous lentigines on sun-exposed hematologic malignancies, especially in children and skin, when her family emigrated from Morocco to the young adults. Germline predisposition in myeloid neo - USA. The homozygous North African XPC founder muta - plasms was added to the World Health Organization tion was present. 10 She had her first skin cancer at the age 1 2016 classification, and current management recommen - of 8 years, and subsequently developed more than 40 cuta - dations emphasize the importance of screening appropri - neous basal and squamous cell carcinomas, one melanoma 2 ate patients. Rare syndromes of DNA repair defects can in situ , and one ocular surface squamous neoplasm. She 3 lead to myeloid and/or lymphoid neoplasms. Here, we was diagnosed with a multinodular goiter at the age of 9 describe our experience with hematologic neoplasms in years eight months, with several complex nodules leading the defective DNA repair syndrome, xeroderma pigmen - to removal of her thyroid gland. Histopathology showed tosum (XP), including myelodysplastic syndrome (MDS), multinodular adenomatous/papillary hyperplasia. At the secondary acute myeloid leukemia (AML), high-grade age of 19 years, she presented with night sweats, fatigue, lymphoma, and an extremely unusual presentation of and lymphadenopathy. Laboratory studies revealed pancy - mixed phenotype acute leukemia (MPAL) with B, T and topenia with hemoglobin 6.8 g/dL, platelet count myeloid blasts.
    [Show full text]