MSH6 Haploinsufficiency at Relapse Contributes to the Development of Thiopurine Resistance in Pediatric B-Lymphoblastic Leukemia

Total Page:16

File Type:pdf, Size:1020Kb

MSH6 Haploinsufficiency at Relapse Contributes to the Development of Thiopurine Resistance in Pediatric B-Lymphoblastic Leukemia View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Copenhagen University Research Information System MSH6 haploinsufficiency at relapse contributes to the development of thiopurine resistance in pediatric B-lymphoblastic leukemia Evensen, Nikki A.; Madhusoodhan, P. Pallavi; Meyer, Julia; Saliba, Jason; Chowdhury, Ashfiyah; Araten, David J.; Nersting, Jacob; Bhatla, Teena; Vincent, Tiffaney L.; Teachey, David; Hunger, Stephen P.; Yang, Jun; Schmiegelow, Kjeld; Carroll, William L. Published in: Haematologica DOI: 10.3324/haematol.2017.176362 Publication date: 2018 Document version Publisher's PDF, also known as Version of record Document license: CC BY-NC Citation for published version (APA): Evensen, N. A., Madhusoodhan, P. P., Meyer, J., Saliba, J., Chowdhury, A., Araten, D. J., ... Carroll, W. L. (2018). MSH6 haploinsufficiency at relapse contributes to the development of thiopurine resistance in pediatric B-lymphoblastic leukemia. Haematologica, 103(5), 830-839. https://doi.org/10.3324/haematol.2017.176362 Download date: 09. Apr. 2020 ARTICLE Acute Lymphoblastic Leukemia MSH6 haploinsufficiency at relapse Ferrata Storti contributes to the development of thiopurine Foundation resistance in pediatric B-lymphoblastic leukemia Nikki A. Evensen,1 P. Pallavi Madhusoodhan,1 Julia Meyer,2 Jason Saliba,1 Ashfiyah Chowdhury,1 David J. Araten,3 Jacob Nersting,4 Teena Bhatla,1 Tiffaney L. Vincent,5 David Teachey,5 Stephen P. Hunger,5 Jun Yang,6 Haematologica 2018 Kjeld Schmiegelow4 and William L. Carroll1 Volume 103(5):830-839 1Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU-Langone Medical Center, New York, NY, USA; 2Huntsman Cancer Institute, University of Utah Medical Center, Salt Lake City, USA; 3Department of Medicine, Perlmutter Cancer Center, NYU-Langone Medical Center, New York NY, USA; 4Department of Pediatrics and Adolescent Medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark; 5Department of Pediatrics and the Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA, USA and 6St. Jude Children’s Research Hospital, Memphis, TN, USA ABSTRACT urvival of children with relapsed acute lymphoblastic leukemia is poor, and understanding mechanisms underlying resistance is Sessential to developing new therapy. Relapse-specific heterozygous deletions in MSH6, a crucial part of DNA mismatch repair, are frequent- ly detected. Our aim was to determine whether MSH6 deletion results in a hypermutator phenotype associated with generation of secondary mutations involved in drug resistance, or if it leads to a failure to initiate apoptosis directly in response to chemotherapeutic agents. We knocked down MSH6 in mismatch repair proficient cell lines (697 and UOCB1) Correspondence: and showed significant increases in IC50s to 6-thioguanine and 6-mer- [email protected] captopurine (697: 26- and 9-fold; UOCB1: 5- and 8-fold) in vitro, as well as increased resistance to 6-mercaptopurine treatmentin vivo. No shift in IC50 was observed in deficient cells (Reh and RS4;11). 697 MSH6 knock- down resulted in increased DNA thioguanine nucleotide levels com- Received: July 13, 2017. pared to non-targeted cells (3070vs. 1722 fmol/μg DNA) with no differ- Accepted: February 7, 2018. ence observed in mismatch repair deficient cells. Loss of MSH6 did not Pre-published: February 15, 2018. give rise to microsatellite instability in cell lines or clinical samples, nor did it significantly increase mutation rate, but rather resulted in a defect in cell cycle arrest upon thiopurine exposure. MSH6 knockdown cells doi:10.3324/haematol.2017.176362 showed minimal activation of checkpoint regulator CHK1, γH2AX (DNA damage marker) and p53 levels upon treatment with thiopurines, Check the online version for the most updated consistent with intrinsic chemoresistance due to failure to recognize information on this article, online supplements, thioguanine nucleotide mismatching and initiate mismatch repair. and information on authorship & disclosures: Aberrant MSH6 adds to the list of alterations/mutations associated with www.haematologica.org/content/103/5/830 acquired resistance to purine analogs emphasizing the importance of thiopurine therapy. ©2018 Ferrata Storti Foundation Material published in Haematologica is covered by copyright. All rights are reserved to the Ferrata Storti Foundation. Use of Introduction published material is allowed under the following terms and conditions: https://creativecommons.org/licenses/by-nc/4.0/legalcode. Relapsed B-precursor acute lymphoblastic leukemia (B-ALL) is a leading cause of Copies of published material are allowed for personal or inter- cancer mortality amongst children. Development of chemoresistance is a crucial nal use. Sharing published material for non-commercial pur- factor contributing to relapse, therefore understanding the biological mechanisms poses is subject to the following conditions: https://creativecommons.org/licenses/by-nc/4.0/legalcode, underlying this resistance is imperative for discovering innovative treatment strate- 1 sect. 3. Reproducing and sharing published material for com- gies. Recent work has begun to highlight the direct role of relapse mercial purposes is not allowed without permission in writing specific/enriched genetic alterations in the emergence of clones that have gained a from the publisher. selective advantage under the pressure of specific chemotherapeutics, such as NT5C2, TBL1XR1, PRPS1, and CREBBP.1-5 Many of these mutations cause resist- ance specifically to thiopurines, which are the backbone of maintenance therapy 830 haematologica | 2018; 103(5) MSH6 haploinsufficiency contributes to resistance and have proven vital for achieving cures.6 Our analysis of RPMI1640 medium. HEK293T (ATCC) cells were grown in copy number alterations (CNAs) in diagnosis/relapse pairs DMEM medium. All media were supplemented with 10% FBS, revealed a relapse specific hemizygous deletion on chro- 1% penicillin/streptomycin under 5% CO2 at 37°C. mosome 2p16.3 involving MSH6 in 4-10% of patients.7,8 MutS homolog 6 (MSH6) is a major component of the Drug preparation, viral preparation, immunoblotting, mismatch repair (MMR) system, which is a highly con- apoptosis assays, and cell cycle served biological process that recognizes and repairs Standard protocols were followed and have been previously errors in nascent DNA strands during replication to main- described.3,21 More detailed information is provided in the Online tain genomic integrity. Initial recognition of replicative Supplementary Appendix. errors is carried out by protein heterodimers consisting of either MSH6 and MSH2 (hMutSa), or MSH3 and MSH2 Patients’ samples (hMutSβ). Upon recognition of a mismatch, hMutSa Cryopreserved pediatric B-ALL specimens were obtained from recruits MutLa (MLH1-PMS2) which engages down- the Children's Oncology Group (COG) ALL cell bank. All patients stream proteins and enzymes involved in DNA repair.9,10 were treated on COG protocols for newly diagnosed ALL. All sub- Constitutional defects in MMR, including monoallelic jects provided consent for banking and future research use of these mutations in Lynch syndrome and biallelic loss in consti- specimens in accordance with the regulations of the institutional tutional mismatch repair deficiency (CMMRD), are review boards of all participating institutions. strongly linked to carcinogenesis, where loss of MMR functionality causes increased mutability and predisposi- Microsatellite instability analysis tion to malignancy.11-15 Previous work has linked defects Microsatellite instability (MSI) analysis was performed using in MMR to drug resistance, including thiopurines, in var- MSI Analysis System, v.1.2 (Promega, Madison, WI, USA) follow- ious cancers.16-19 However, it is uncertain if resistance in ing the manufacturer’s protocol. Detailed information is provided MMR defective clones occurs through the acquisition of in the Online Supplementary Appendix. secondary mutations as a consequence of mutagenic ther- apy, or the outgrowth of clones that have intrinsic drug Measurement of mutation rate resistance. Our lab previously demonstrated that lower Spontaneous mutation rate was measured using a flow cytom- expression of MSH6 in patient samples was associated etry assay previously described by Araten et al.22 that detects the with increased ex vivo resistance to 6-mercaptopurine and presence of numerous glycosylphosphatidylinositol-linked (GPI) prednisone,7 highlighting the clinical importance of under- membrane proteins (see Online Supplementary Appendix). Briefly, standing the role of this genetic alteration in B-ALL. GPI(+) isolated clones from the NT and MSH6-KD cell lines were The mechanism of action of thiopurines is based upon expanded either untreated or treated with 6-TG (0.040 μg/mL and the insertion of a false nucleotide, namely a thioguanine 0.100 μg/mL, respectively, based on IC50 values determined for (TGN), into DNA that when thiomethylated pairs with a clones). Cells were then stained for GPI-dependent markers thymine instead of a cytosine.18 Cytotoxicity is thought to including FLAER-Alexa 488 (Pinewood), CD48, CD52, and CD59 be dependent on the MMR machinery recognizing the (Serotec),23 and analyzed by flow cytometry. The mutant frequen- mismatch and attempting to match the TGN on the cy (f) was calculated as the number of GPI(-) events divided by the parental
Recommended publications
  • Conformational Trapping of Mismatch Recognition Complex MSH2/MSH3
    Conformational trapping of Mismatch Recognition PNAS PLUS Complex MSH2/MSH3 on repair-resistant DNA loops Walter H. Langa,1,2, Julie E. Coatsb,1, Jerzy Majkaa, Greg L. Huraa, Yuyen Linb, Ivan Rasnikb,3, and Cynthia T. McMurraya,c,d,3 aLawrence Berkeley National Laboratory, Life Sciences Division, 1 Cyclotron Road, Berkeley, CA 94720; cDepartment of Molecular Pharmacology and Experimental Therapeutics; dDepartment of Biochemistry and Molecular Biology, Mayo Foundation, 200 First Street, Rochester, MN 55905; and bDepartment of Physics, Emory University, 400 Dowman Drive, MSC N214, Atlanta, GA 30322 Edited* by Peter H. von Hippel, Institute of Molecular Biology, Eugene, OR, and approved August 3, 2011 (received for review April 6, 2011) Insertion and deletion of small heteroduplex loops are common which fails to effectively couple DNA binding with downstream mutations in DNA, but why some loops are prone to mutation and repair signaling. We envision that conformational regulation of others are efficiently repaired is unknown. Here we report that the small loop repair occurs at the level of the junction dynamics. mismatch recognition complex, MSH2/MSH3, discriminates between a repair-competent and a repair-resistant loop by sensing the con- Results formational dynamics of their junctions. MSH2/MSH3 binds, bends, Conformational Integrity of MSH2/MSH3 and the DNA “Junction” Tem- and dissociates from repair-competent loops to signal downstream plates. We characterized the DNA-binding affinity and nucleotide repair. Repair-resistant Cytosine-Adenine-Guanine (CAG) loops adopt binding properties of MSH2/MSH3 bound to looped templates, a unique DNA junction that traps nucleotide-bound MSH2/MSH3, either a ðCAÞ4 loop or CAG hairpin loops of either 7 or 13 and inhibits its dissociation from the DNA.
    [Show full text]
  • Dihydrofolate Reductase Amplification and Sensitization to Methotrexate of Methotrexate-Resistant Colon Cancer Cells
    Published OnlineFirst February 3, 2009; DOI: 10.1158/1535-7163.MCT-08-0759 424 Dihydrofolate reductase amplification and sensitization to methotrexate of methotrexate-resistant colon cancer cells Cristina Morales,1 Maria J. Garcı´a,4 Maria Ribas,1 withdrawal and reexposure to MTX. Passive loss of the Rosa Miro´,2 Mar Mun˜oz,3 Carlos Caldas,4 DHFR amplicon by withdrawal of the drug results in MTX- and Miguel A. Peinado1,3 sensitive cells exhibiting a substantial reduction of their capacity or even an incapacity to generate resistance when 1Institut d’Investigacio´Biome`dica de Bellvitge, L’Hospitalet; submitted to a second cycle of MTX treatment. On a 2 Institut de Biotecnologia i Biomedicina, Departament de Biologia second round of drug administration, the resistant cells CelÁlular, Fisiologia i Immunologia, Universitat Autonoma de Barcelona, Bellaterra; 3Institut de Medicina Predictiva i generate a different amplicon structure, suggesting that the Personalitzada del Ca`ncer, Badalona, Barcelona, Spain and formation of the amplicon as in the first cycle of treatment 4Breast Cancer Functional Genomics Laboratory, Cancer is not feasible. These results indicate that DHFR gene Research UK Cambridge Research Institute and Department of amplification is a ‘‘wear and tear’’ process in HT29 cells Oncology, University of Cambridge, Li Ka Shing Centre, and that MTX-resistant cells may become responsive to a Cambridge, United Kingdom second round of treatment if left untreated during a sufficient period of time. [Mol Cancer Ther 2009; Abstract 8(2):424–32] Gene amplification is one of the most frequent manifes- tations of genomic instability in human tumors and plays an important role in tumor progression and acquisition of Introduction drug resistance.
    [Show full text]
  • Recruitment of Mismatch Repair Proteins to the Site of DNA Damage in Human Cells
    3146 Research Article Recruitment of mismatch repair proteins to the site of DNA damage in human cells Zehui Hong, Jie Jiang, Kazunari Hashiguchi, Mikiko Hoshi, Li Lan and Akira Yasui* Department of Molecular Genetics, Institute of Development, Aging and Cancer, Tohoku University, Seiryomachi 4-1, Aobaku, Sendai 980-8575, Japan *Author for correspondence (e-mail: [email protected]) Accepted 8 July 2008 Journal of Cell Science 121, 3146-3154 Published by The Company of Biologists 2008 doi:10.1242/jcs.026393 Summary Mismatch repair (MMR) proteins contribute to genome stability the PCNA-binding domain of MSH6. MSH2 is recruited to the by excising DNA mismatches introduced by DNA polymerase. DNA damage site through interactions with either MSH3 or Although MMR proteins are also known to influence cellular MSH6, and is required for recruitment of MLH1 to the damage responses to DNA damage, how MMR proteins respond to DNA site. We found, furthermore, that MutSβ is also recruited to damage within the cell remains unknown. Here, we show that UV-irradiated sites in nucleotide-excision-repair- and PCNA- MMR proteins are recruited immediately to the sites of various dependent manners. Thus, MMR and its proteins function not types of DNA damage in human cells. MMR proteins are only in replication but also in DNA repair. recruited to single-strand breaks in a poly(ADP-ribose)- dependent manner as well as to double-strand breaks. Using Supplementary material available online at mutant cells, RNA interference and expression of fluorescence- http://jcs.biologists.org/cgi/content/full/121/19/3146/DC1
    [Show full text]
  • Machine Learning-Based Approach Highlights the Use of a Genomic Variant Profile for Precision Medicine in Ovarian Failure
    Journal of Personalized Medicine Article Machine Learning-Based Approach Highlights the Use of a Genomic Variant Profile for Precision Medicine in Ovarian Failure Ismael Henarejos-Castillo 1,2 , Alejandro Aleman 1, Begoña Martinez-Montoro 3, Francisco Javier Gracia-Aznárez 4, Patricia Sebastian-Leon 1,3, Monica Romeu 5, Jose Remohi 2,6, Ana Patiño-Garcia 4,7 , Pedro Royo 3, Gorka Alkorta-Aranburu 4 and Patricia Diaz-Gimeno 1,3,* 1 IVI Foundation-Instituto de Investigación Sanitaria La Fe, Av. Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026 Valencia, Spain; [email protected] (I.H.-C.); [email protected] (A.A.); [email protected] (P.S.-L.) 2 Department of Paediatrics, Obstetrics and Gynaecology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; [email protected] 3 IVI-RMA Pamplona, Reproductive Medicine, C/Sangüesa, Número 15-Planta Baja, 31003 Pamplona, Spain; [email protected] (B.M.-M.); [email protected] (P.R.) 4 CIMA Lab Diagnostics, University of Navarra, IdiSNA, Avda Pio XII, 55, 31008 Pamplona, Spain; [email protected] (F.J.G.-A.); [email protected] (A.P.-G.); [email protected] (G.A.-A.) 5 Hospital Universitario y Politécnico La Fe, Av. Fernando Abril Martorell 106, 46026 Valencia, Spain; [email protected] 6 IVI-RMA Valencia, Reproductive Medicine, Plaça de la Policia Local, 3, 46015 Valencia, Spain 7 Laboratorio de Pediatría-Unidad de Genética Clínica, Clínica Universidad de Navarra, Avda Pio XII, 55, Citation: Henarejos-Castillo, I.; 31008 Pamplona, Spain Aleman, A.; Martinez-Montoro, B.; * Correspondence: [email protected] Gracia-Aznárez, F.J.; Sebastian-Leon, P.; Romeu, M.; Remohi, J.; Patiño- Abstract: Ovarian failure (OF) is a common cause of infertility usually diagnosed as idiopathic, Garcia, A.; Royo, P.; Alkorta-Aranburu, with genetic causes accounting for 10–25% of cases.
    [Show full text]
  • MSH3 Protein Expression and Nodal Status in MLH1-Deficient Colorectal Cancers Running Title
    Author Manuscript Published OnlineFirst on April 10, 2012; DOI: 10.1158/1078-0432.CCR-12-0175 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Title: MSH3 Protein Expression and Nodal Status in MLH1-Deficient Colorectal Cancers Running Title: MSH3 status and Nodal Metastasis in MSI CRC Authors: Luigi Laghi 1,2, Paolo Bianchi 1, Gabriele Delconte 2, Giuseppe Celesti 1, Giuseppe Di Caro 1,3, Monica Pedroni 4, Anna Maria Chiaravalli 5, Barbara Jung 6, Carlo Capella 5, Maurizio Ponz de Leon 4, Alberto Malesci 2,7 Institutions: 1 Laboratory of Molecular Gastroenterology , IRCCS Istituto Clinico Humanitas Rozzano (Milan), Italy. 2 Department of Gastroenterology, IRCCS Istituto Clinico Humanitas - Rozzano (Milan), Italy. 3 PhD Program of the School in Pathology and Neuropathology - University of Milan, Italy. 4 Department of Internal Medicine, Medical Faculty, Modena and Reggio Emilia University, Modena, Italy. 5 Department of Human Morphology, Anatomic Pathology Unit, University of Insubria, Varese, Italy. 6 Division of Gastroenterology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA. 7 Department of Translational Medicine – University of Milan- Milan, Italy Addresses for correspondence: Dr.Luigi Laghi Prof. Alberto Malesci Dept. of Gastroenterology Dept. of Gastroenterology IRCCS Istituto Clinico Humanitas IRCCS Istituto Clinico Humanitas-University of Milan Via Manzoni, 56 Via Manzoni, 56 20089 Rozzano – Milan 20089 Rozzano - Milan ITALY ITALY e-mail: [email protected] e-mail: [email protected] phone: +39. 02. 8224. 4572 phone: +39. 02. 8224. 4542 fax: +39. 02. 8224. 4590 fax: +39. 02. 8224. 4590 Keywords: colorectal cancer, microsatellite instability, cancer genes, prognosis, biological markers 1 Downloaded from clincancerres.aacrjournals.org on September 25, 2021.
    [Show full text]
  • A Functional Assay for Mutations in Tumor Suppressor Genes Caused by Mismatch Repair Deficiency
    © 2001 Oxford University Press Human Molecular Genetics, 2001, Vol. 10, No. 24 2737–2743 A functional assay for mutations in tumor suppressor genes caused by mismatch repair deficiency Hanlee P. Ji and Mary-Claire King* Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195-7720, USA Received May 25, 2001; Revised and Accepted September 19, 2001 The coding sequences of multiple human tumor MMR-deficient strains of S.cerevisiae has increased our suppressor genes include microsatellite sequences understanding of the role of MSI in human tumors (5–7). that are prone to mutations. Saccharomyces cerevisiae In cancers from individuals with HNPCC, the elevation in strains deficient in DNA mismatch repair (MMR) can mutation rates causes inactivation of specific tumor suppressor be used to determine de novo mutation rates of these gene, producing a selective growth advantage and thus human tumor suppressor genes as well as any other contributing to stepwise neoplastic progression (8). Multiple genes have been identified with microsatellite mutations gene sequence. Microsatellites in human TGFBR2, including TGFBR2, IGFR2, MSH3, MSH6, PTEN, BAX and PTEN and APC genes were placed in yeast vectors MBD4 (8–15). The majority of these microsatellites consist of and analyzed in isogenic yeast strains that were wild- mononucleotide repeats which accumulate 1 bp deletions or type or deletion mutants for MSH2 or MLH1. In MMR- insertions. deficient strains, the vector containing the (A)10 Observational studies have identified these microsatellite microsatellite sequence of TGFBR2 had a mutation mutation hotspots; candidate genes are sequenced for micro- rate (mutations/cell division) of 1.4 × 10–4, compared satellite mutations in a series of tumors and cell lines exhib- to a mutation rate of 1.7 × 10–6 in the wild-type strain.
    [Show full text]
  • Germline and Tumor Whole Genome Sequencing As a Diagnostic Tool to Resolve Suspected Lynch Syndrome
    medRxiv preprint doi: https://doi.org/10.1101/2020.03.12.20034991; this version posted March 17, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Germline and Tumor Whole Genome Sequencing as a Diagnostic Tool to Resolve Suspected Lynch Syndrome Bernard J. Pope1,2,3, Mark Clendenning1,2, Christophe Rosty1,2,4,5, Khalid Mahmood1,2,3, Peter Georgeson1,2, Jihoon E. Joo1,2, Romy Walker1,2, Ryan A. Hutchinson1,2, Harindra Jayasekara1,2,6, Sharelle Joseland1,2, Julia Como1,2, Susan Preston1,2, Amanda B. Spurdle7, Finlay A. Macrae8,9,10, Aung K. Win11, John L. Hopper11, Mark A. Jenkins11, Ingrid M. Winship9,10, Daniel D. Buchanan1,2,10* 1 Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia 2 University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia 3 Melbourne Bioinformatics, The University of Melbourne, Parkville, Victoria, Australia 4 Envoi Pathology, Brisbane, Queensland, Australia 5 University of Queensland, School of Medicine, Herston, Queensland, Australia 6 Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia 7 Molecular Cancer Epidemiology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia 8 Colorectal Medicine and Genetics, The Royal Melbourne
    [Show full text]
  • Review DNA Mismatch Repair Genes and Colorectal Cancer
    148 Gut 2000;47:148–153 Review Gut: first published as 10.1136/gut.47.1.148 on 1 July 2000. Downloaded from DNA mismatch repair genes and colorectal cancer Summary hereditary component and the work of Warthin and Lynch Positional cloning and linkage analysis have shown that was seen as being anecdotal. However, by the 1980s many inactivation of one of the mismatch repair genes (hMLH1, reports of a “cancer family syndrome” were appearing in hMSH2, hPMS1, hPMS2, GTBP/hMSH6) is responsible the medical literature.56 Cancer family syndrome then for the microsatellite instability or replication error became subdivided into Lynch syndrome I (families with (RER+) seen in more than 90% of hereditary non- mainly colorectal cancers at an early age) and Lynch syn- polyposis colorectal cancers (HNPCC) and 15% of drome II (families with colorectal and extracolonic sporadic RER+ colorectal cancers. In HNPCC, a germline cancers, particularly of the female genital tract).7 All of this mutation (usually in hMLH1 or hMSH2) is accompanied diVerent terminology was eventually clarified with the by one further event (usually allelic loss) to inactivate a introduction of the term hereditary non-polyposis colorec- mismatch repair gene. In contrast, somatic mutations in tal cancer (HNPCC) to emphasise the lack of multiple the mismatch repair genes are not frequently found in spo- colonic polyps and to separate it from the polyposis radic RER+ colorectal cancers. Hypermethylation of the syndromes. hMLH1 promoter region has recently been described, and this epigenetic change is the predominant cause of Discovery of human mismatch repair genes inactivation of mismatch repair genes in sporadic RER+ Following the study of large kindreds using linkage analy- colorectal and other cancers.
    [Show full text]
  • MSH2 Gene Muts Homolog 2
    MSH2 gene mutS homolog 2 Normal Function The MSH2 gene provides instructions for making a protein that plays an essential role in repairing DNA. This protein helps fix errors that are made when DNA is copied (DNA replication) in preparation for cell division. The MSH2 protein joins with one of two other proteins, MSH6 or MSH3 (each produced from a different gene), to form a two-protein complex called a dimer. This complex identifies locations on the DNA where errors have been made during DNA replication. Another group of proteins, the MLH1-PMS2 dimer, then binds to the MSH2 dimer and repairs the errors by removing the mismatched DNA and replicating a new segment. The MSH2 gene is one of a set of genes known as the mismatch repair (MMR) genes. Health Conditions Related to Genetic Changes Constitutional mismatch repair deficiency syndrome About 10 mutations in the MSH2 gene have been associated with a condition called constitutional mismatch repair deficiency (CMMRD) syndrome. Individuals with this condition are at increased risk of developing cancers of the colon (large intestine) and rectum (collectively referred to as colorectal cancer), brain, and blood (leukemia or lymphoma). These cancers usually first occur in childhood, with the vast majority of cancers in CMMRD syndrome diagnosed in people under the age of 18. Many people with CMMRD syndrome also develop changes in skin coloring (pigmentation), similar to those that occur in a condition called neurofibromatosis type 1. Individuals with CMMRD syndrome inherit two MSH2 gene mutations, one from each parent, while people with Lynch syndrome (described below) have a mutation in one copy of the MSH2 gene.
    [Show full text]
  • Differential Regulation of Expression of the Mammalian DNA Repair Genes by Growth Stimulation
    Oncogene (2004) 23, 8581–8590 & 2004 Nature Publishing Group All rights reserved 0950-9232/04 $30.00 www.nature.com/onc Differential regulation of expression of the mammalian DNA repair genes by growth stimulation Ritsuko Iwanaga1, Hideyuki Komori1 and Kiyoshi Ohtani*,1 1Human Gene Sciences Center, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan During DNA replication, DNA becomes more vulnerable did not change significantly during the cell cycle (Meyers to certain DNA damages. DNA repair genes involved in et al., 1997). However, previous finding that hMSH2 repair of the damages may be induced by growth protein was expressed in proliferating cells of the gut stimulation. However, regulation of DNA repair genes (Wilson et al., 1995) and recent findings that some of by growth stimulation has not been analysed in detail. In repair genes such as MSH2 and Rad51 were induced by this report, we analysed the regulation of expression of overexpression of the transcription factor E2F (Ishida mammalian MSH2, MSH3 and MLH1 genes involved in et al., 2001; Polager et al., 2002), which plays crucial mismatch repair, and Rad51 and Rad50 genes involved in roles in cell growth control, suggest that some of the homologous recombination repair, in relation to cell repair genes may be regulated by growth stimulation. In growth. Unexpectedly, we found a clear difference in spite of the expectation, the regulation of DNA repair regulation of these repair gene expression by growth genes by growth stimulation such as serum stimulation stimulation even in the same repair system. The expres- of quiescent fibroblasts has not been analysed in detail.
    [Show full text]
  • Functional Analysis of Human Mismatch Repair Gene Mutations Identifies Weak Alleles and Polymorphisms Capable of Polygenic Interactions
    Functional analysis of human mismatch repair gene mutations identifies weak alleles and polymorphisms capable of polygenic interactions Sandra L. Martinez and Richard D. Kolodner1 Ludwig Institute for Cancer Research, Departments of Medicine and Cellular and Molecular Medicine, and Moores-UCSD Cancer Center, UC San Diego School of Medicine, La Jolla, CA 92093-0669 Contributed by Richard D. Kolodner, January 22, 2010 (sent for review December 24, 2009) Many of the mutations reported as potentially causing Lynch morphisms in MSH2, MLH1, MSH6, and PMS2 (PMS1 in S. syndrome are missense mutations in human mismatch repair cerevisiae) and 14 reported missense mutations in these genes by (MMR) genes. Here, we used a Saccharomyces cerevisiae-based making equivalent mutations at the analogous position in the system to study polymorphisms and suspected missense mutations corresponding chromosomal gene in S. cerevisiae and evaluating in human MMR genes by modeling them at the appropriate S. their effect on MMR using mutator assays (Tables 1 and 2; S. cerevisiae chromosomal locus and determining their effect on cerevisiae allele designations are used in the text of this report). mutation rates. We identified a number of weak alleles of MMR Some of these variants have been characterized in other studies genes and MMR gene polymorphisms that are capable of interact- using S. cerevisiae-based assays (16–19, 23, 25) but mostly by ing with other weak alleles of MMR genes to produce strong poly- testing them as plasmid-borne mutations (16–19, 23)(Table 2). genic MMR defects. We also identified a number of alleles of MSH2 The mutator assays used include CAN1 gene inactivation that that act as if they inactivate the Msh2-Msh3 mispair recognition detects mutations inactivating the CAN1 gene and hom3-10 and complex thus causing weak MMR defects that interact with an lys2-10A frameshift reversions that detect mutations that revert msh6Δ mutation to result in complete MMR defects.
    [Show full text]
  • DNA Mismatch Repair Status Affects Cellular Response to Ara-C and Other Anti-Leukemic Nucleoside Analogs
    Letters to the Editor 1046 grants CA30969, CA29139, CA98543, CA98413, and CA114766 for unfavorable early treatment response. Blood 2009; 114: awarded to the Children’s Oncology Group. 1053–1062. 4 Gutierrez A, Sanda T, Grebliunaite R, Carracedo A, Salmena L, LYu1, ML Slovak2,5, K Mannoor1, C Chen1, SP Hunger3, Ahn Y et al. High frequency of PTEN, PI3K, and AKT abnormalities in T-cell acute lymphoblastic leukemia. Blood 2009; 114: AJ Carroll4, RA Schultz2, LG Shaffer2, BC Ballif2 and Y Ning1 1 647–650. Department of Pathology, University of Maryland School 5 Duker AL, Ballif BC, Bawle EV, Person RE, Mahadevan S, of Medicine, Baltimore, MD, USA; 2 Alliman S et al. Paternally inherited microdeletion at 15q11.2 Division of Cytogenetics, Signature Genomic Laboratories, confirms a significant role for the SNORD116 C/D box snoRNA Spokane, WA, USA; 3 cluster in Prader-Willi syndrome. Eur J Hum Genet 2010; 18: Center for Cancer and Blood Disorders, University of 1196–1201. Colorado Denver School of Medicine, Aurora, CO, USA and 6 Raschke S, Balz V, Efferth T, Schulz WA, Florl AR. Homozygous 4 Department of Genetics, University of Alabama at deletions of CDKN2A caused by alternative mechanisms in various Birmingham, Birmingham, AL, USA human cancer cell lines. Genes Chromosomes Cancer 2005; 42: E-mail: [email protected] 58–67. 5Current address: Division of Cytogenetics, Quest Diagnostics 7 De Keersmaecker K, Marynen P, Cools J. Genetic insights in the Nichols Institute, Chantilly, VA, USA. pathogenesis of T-cell acute lymphoblastic leukemia. Haemato- logica 2005; 90: 1116–1127. 8 Colaizzo-Anas T, Aplan PD.
    [Show full text]