Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Genes Dev ; 32(7-8): 524-536, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29636371

ABSTRACT

Chromosomal deletion rearrangements mediated by repetitive elements often involve repeats separated by several kilobases and sequences that are divergent. While such rearrangements are likely induced by DNA double-strand breaks (DSBs), it has been unclear how the proximity of DSBs relative to repeat sequences affects the frequency of such events. We generated a reporter assay in mouse cells for a deletion rearrangement involving repeats separated by 0.4 Mb. We induced this repeat-mediated deletion (RMD) rearrangement with two DSBs: the 5' DSB that is just downstream from the first repeat and the 3' DSB that is varying distances upstream of the second repeat. Strikingly, we found that increasing the 3' DSB/repeat distance from 3.3 kb to 28.4 kb causes only a modest decrease in rearrangement frequency. We also found that RMDs are suppressed by KU70 and RAD51 and promoted by RAD52, CtIP, and BRCA1. In addition, we found that 1%-3% sequence divergence substantially suppresses these rearrangements in a manner dependent on the mismatch repair factor MSH2, which is dominant over the suppressive role of KU70. We suggest that a DSB far from a repeat can stimulate repeat-mediated rearrangements, but multiple pathways suppress these events.


Subject(s)
Chromosome Breakage , Chromosome Deletion , Repetitive Sequences, Nucleic Acid , Animals , DNA/chemistry , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/physiology , Ku Autoantigen/physiology , Mice , MutS Homolog 2 Protein/physiology , Rad51 Recombinase/physiology , Rad52 DNA Repair and Recombination Protein/physiology , Tumor Suppressor p53-Binding Protein 1/physiology
2.
Mol Cell ; 59(4): 603-14, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26212458

ABSTRACT

Ataxia telangiectasia-mutated and Rad3-related (ATR) protein kinase, a master regulator of DNA-damage response, is activated by RPA-coated single-stranded DNA (ssDNA) generated at stalled replication forks or DNA double-strand breaks (DSBs). Here, we identify the mismatch-binding protein MutSß, a heterodimer of MSH2 and MSH3, as a key player in this process. MSH2 and MSH3 form a complex with ATR and its regulatory partner ATRIP, and their depletion compromises the formation of ATRIP foci and phosphorylation of ATR substrates in cells responding to replication-associated DSBs. Purified MutSß binds to hairpin loop structures that persist in RPA-ssDNA complexes and promotes ATRIP recruitment. Mutations in the mismatch-binding domain of MSH3 abolish the binding of MutSß to DNA hairpin loops and its ability to promote ATR activation by ssDNA. These results suggest that hairpin loops might form in ssDNA generated at sites of DNA damage and trigger ATR activation in a process mediated by MutSß.


Subject(s)
DNA Breaks, Double-Stranded , DNA-Binding Proteins/physiology , MutS Homolog 2 Protein/physiology , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Repair , DNA, Single-Stranded/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Enzyme Activation , HEK293 Cells , Homologous Recombination , Humans , MutS Homolog 2 Protein/chemistry , MutS Homolog 3 Protein , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein Transport
3.
Mol Cell ; 43(4): 649-62, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21855803

ABSTRACT

Posttranslational modification of PCNA by ubiquitin plays an important role in coordinating the processes of DNA damage tolerance during DNA replication. The monoubiquitination of PCNA was shown to facilitate the switch between the replicative DNA polymerase with the low-fidelity polymerase eta (η) to bypass UV-induced DNA lesions during replication. Here, we show that in response to oxidative stress, PCNA becomes transiently monoubiquitinated in an S phase- and USP1-independent manner. Moreover, Polη interacts with mUb-PCNA at sites of oxidative DNA damage via its PCNA-binding and ubiquitin-binding motifs. Strikingly, while functional base excision repair is not required for this modification of PCNA or Polη recruitment to chromatin, the presence of hMsh2-hMsh6 is indispensable. Our findings highlight an alternative pathway in response to oxidative DNA damage that may coordinate the removal of oxidatively induced clustered DNA lesions and could explain the high levels of oxidized DNA lesions in MSH2-deficient cells.


Subject(s)
DNA Damage , DNA-Binding Proteins/physiology , DNA-Directed DNA Polymerase/physiology , MutS Homolog 2 Protein/physiology , Oxidative Stress , Proliferating Cell Nuclear Antigen/physiology , Arabidopsis Proteins , Cell Line , Chromatin/metabolism , DNA Polymerase beta/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Endopeptidases/metabolism , Humans , MutS Homolog 2 Protein/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Specific Proteases , Ubiquitination , X-ray Repair Cross Complementing Protein 1
4.
Nucleic Acids Res ; 45(17): 10068-10078, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28973443

ABSTRACT

CTG•CAG repeat expansions cause at least twelve inherited neurological diseases. Expansions require the presence, not the absence, of the mismatch repair protein MutSß (Msh2-Msh3 heterodimer). To evaluate properties of MutSß that drive expansions, previous studies have tested under-expression, ATPase function or polymorphic variants of Msh2 and Msh3, but in disparate experimental systems. Additionally, some variants destabilize MutSß, potentially masking the effects of biochemical alterations of the variations. Here, human Msh3 was mutated to selectively inactivate MutSß. Msh3-/- cells are severely defective for CTG•CAG repeat expansions but show full activity on contractions. Msh3-/- cells provide a single, isogenic system to add back Msh3 and test key biochemical features of MutSß on expansions. Msh3 overexpression led to high expansion activity and elevated levels of MutSß complex, indicating that MutSß abundance drives expansions. An ATPase-defective Msh3 expressed at normal levels was as defective in expansions as Msh3-/- cells, indicating that Msh3 ATPase function is critical for expansions. Expression of two Msh3 polymorphic variants at normal levels showed no detectable change in expansions, suggesting these polymorphisms primarily affect Msh3 protein stability, not activity. In summary, CTG•CAG expansions are limited by the abundance of MutSß and rely heavily on Msh3 ATPase function.


Subject(s)
Adenosine Triphosphate/metabolism , DNA Mismatch Repair , MutS Homolog 3 Protein/physiology , Trinucleotide Repeat Expansion/physiology , Amino Acid Substitution , Astrocytes , Brain Neoplasms , CRISPR-Cas Systems , Cell Line , Colorectal Neoplasms , Dimerization , Gene Knockout Techniques , Genes, Reporter , Genetic Vectors , Humans , Hydrolysis , MutS Homolog 2 Protein/physiology , MutS Homolog 3 Protein/deficiency , MutS Homolog 3 Protein/genetics , Mutation, Missense , Neoplastic Syndromes, Hereditary , Point Mutation
5.
Blood ; 127(1): 102-12, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26385350

ABSTRACT

Somatic hypermutation and class-switch recombination of the immunoglobulin (Ig) genes occur in germinal center (GC) B cells and are initiated through deamination of cytidine to uracil by activation-induced cytidine deaminase (AID). Resulting uracil-guanine mismatches are processed by uracil DNA glycosylase (UNG)-mediated base-excision repair and MSH2-mediated mismatch repair (MMR) to yield mutations and DNA strand lesions. Although off-target AID activity also contributes to oncogenic point mutations and chromosome translocations associated with GC and post-GC B-cell lymphomas, the role of downstream AID-associated DNA repair pathways in the pathogenesis of lymphoma is unknown. Here, we show that simultaneous deficiency of UNG and MSH2 or MSH2 alone causes genomic instability and a shorter latency to the development of BCL6-driven diffuse large B-cell lymphoma (DLBCL) in a murine model. The additional development of several BCL6-independent malignancies in these mice underscores the critical role of MMR in maintaining general genomic stability. In contrast, absence of UNG alone is highly protective and prevents the development of BCL6-driven DLBCL. We further demonstrate that clonal and nonclonal mutations arise within non-Ig AID target genes in the combined absence of UNG and MSH2 and that DNA strand lesions arise in an UNG-dependent manner but are offset by MSH2. These findings lend insight into a complex interplay whereby potentially deleterious UNG activity and general genomic instability are opposed by the protective influence of MSH2, producing a net protective effect that promotes immune diversification while simultaneously attenuating malignant transformation of GC B cells.


Subject(s)
Cell Transformation, Neoplastic/pathology , Cytidine Deaminase/metabolism , DNA Repair/genetics , DNA-Binding Proteins/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , MutS Homolog 2 Protein/physiology , Uracil-DNA Glycosidase/physiology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Female , Flow Cytometry , Gene Expression Profiling , Germinal Center , Immunoenzyme Techniques , Immunoglobulin Class Switching/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Proto-Oncogene Proteins c-bcl-6 , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Somatic Hypermutation, Immunoglobulin/genetics , Spectral Karyotyping , Tumor Cells, Cultured
6.
Mol Cell Biochem ; 412(1-2): 297-305, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26728996

ABSTRACT

The mammalian DNA mismatch repair (MMR) system consists of a number of proteins that play important roles in repair of base pair mismatch mutations and in maintenance of genomic integrity. A defect in this system can cause genetic instability, which can lead to carcinogenesis. For instance, a germline mutation in one of the mismatch repair proteins, especially MLH1 or MSH2, is responsible for hereditary non-polyposis colorectal cancer. These MMR proteins also play an important role in the induction of apoptosis. Accordingly, altered expression of or a defect in MLH1 or MSH2 may confer resistance to anti-cancer drugs used in chemotherapy. We hypothesized that the ability of these two MMR proteins to regulate apoptosis are interdependent. Moreover, a defect in either one may confer resistance to chemotherapy by an inability to trigger apoptosis. To this end, we studied three cell lines-SW480, LoVo, and HTC116. These cell lines were selected based on their differential expression of MLH1 and MSH2 proteins. SW480 expresses both MLH1 and MSH2; LoVo expresses only MLH1 but not MSH2; HCT116 expresses only MSH2 but not MLH1 protein. MTT assays, a measure of cytotoxicity, showed that there were different cytotoxic effects of an anti-cancer drug, etoposide, on these cell lines, effects that were correlated with the MMR status of the cells. Cells that are deficient in MLH1 protein (HCT116 cells) were resistant to the drug. Cells that express both MLH1 and MSH2 proteins (SW480 cells) showed caspase-3 cleavage, an indicator of apoptosis. Cells that lack MLH1 (HCT116 cells) did not show any caspase-3 cleavage. Expression of full-length MLH1 protein was decreased in MMR proficient (SW480) cells during apoptosis; it remained unchanged in cells that lack MSH2 (LoVo cells). The expression of MSH2 protein remained unchanged during apoptosis both in MMR proficient (SW480) and deficient (HCT116) cells. Studies on translocation of MLH1 protein from nucleus to cytosolic fraction, an indicator of apoptosis, showed that MLH1 translocation only occurred in MMR proficient (SW480) cells upon induction of apoptosis further suggested a MSH2 dependent role of MLH1 in apoptosis. These data suggest a role of MLH1 in mediation of apoptosis in a MSH2-dependent manner. Taken together, our data supported an interdependence of mismatch repair proteins, particularly MLH1 and MSH2, in the mediation of apoptosis in human colorectal carcinoma cell lines.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis/physiology , Colorectal Neoplasms/pathology , DNA Mismatch Repair , MutS Homolog 2 Protein/physiology , Nuclear Proteins/physiology , Antineoplastic Agents/therapeutic use , Caspase 3/metabolism , Colorectal Neoplasms/drug therapy , Etoposide/therapeutic use , Humans , MutL Protein Homolog 1 , Proteolysis
7.
Eur J Immunol ; 44(7): 1925-35, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24771041

ABSTRACT

Excision of uracil introduced into the immunoglobulin loci by AID is central to antibody diversification. While predominantly carried out by the UNG uracil-DNA glycosylase as reflected by deficiency in immunoglobulin class switching in Ung(-/-) mice, the deficiency is incomplete, as evidenced by the emergence of switched IgG in the serum of Ung(-/-) mice. Lack of switching in mice deficient in both UNG and MSH2 suggested that mismatch repair initiated a backup pathway. We now show that most of the residual class switching in Ung(-/-) mice depends upon the endogenous SMUG1 uracil-DNA glycosylase, with in vitro switching to IgG1 as well as serum IgG3, IgG2b, and IgA greatly diminished in Ung(-/-) Smug1(-/-) mice, and that Smug1 partially compensates for Ung deficiency over time. Nonetheless, using a highly MSH2-dependent mechanism, Ung(-/-) Smug1(-/-) mice can still produce detectable levels of switched isotypes, especially IgG1. While not affecting the pattern of base substitutions, SMUG1 deficiency in an Ung(-/-) background further reduces somatic hypermutation at A:T base pairs. Our data reveal an essential requirement for uracil excision in class switching and in facilitating noncanonical mismatch repair for the A:T phase of hypermutation presumably by creating nicks near the U:G lesion recognized by MSH2.


Subject(s)
Immunoglobulin Class Switching , Mutation , Uracil-DNA Glycosidase/physiology , Uracil/metabolism , Animals , Immunoglobulin A/blood , Immunoglobulin G/blood , Mice , MutS Homolog 2 Protein/physiology
8.
Nucleic Acids Res ; 41(22): 10312-22, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24038355

ABSTRACT

Translesion DNA synthesis (TLS) can use specialized DNA polymerases to insert and/or extend nucleotides across lesions, thereby limiting stalled replication fork collapse and the potential for cell death. Recent studies have shown that monoubiquitinated proliferating cell nuclear antigen (PCNA) plays an important role in recruitment of Y-family TLS polymerases to stalled replication forks after DNA damage treatment. To explore the possible roles of other factors that regulate the ultraviolet (UV)-induced assembly of specialized DNA polymerases at arrested replication forks, we performed immunoprecipitation experiments combined with mass spectrometry and established that DNA polymerase kappa (Polκ) can partner with MSH2, an important mismatch repair protein associated with hereditary non-polyposis colorectal cancer. We found that depletion of MSH2 impairs PCNA monoubiquitination and the formation of foci containing Polκ and other TLS polymerases after UV irradiation of cells. Interestingly, expression of MSH2 in Rad18-deficient cells increased UV-induced Polκ and REV1 focus formation without detectable changes in PCNA monoubiquitination, indicating that MSH2 can regulate post-UV focus formation by specialized DNA polymerases in both PCNA monoubiquitination-dependent and -independent fashions. Moreover, we observed that MSH2 can facilitate TLS across cyclobutane pyrimidine dimers photoproducts in living cells, presenting a novel role of MSH2 in post-UV cellular responses.


Subject(s)
DNA Damage , DNA/biosynthesis , MutS Homolog 2 Protein/physiology , Ultraviolet Rays , Animals , Cell Line , DNA Replication , DNA-Binding Proteins/analysis , DNA-Directed DNA Polymerase/analysis , DNA-Directed DNA Polymerase/metabolism , Humans , Mice , MutS Homolog 2 Protein/metabolism , Nucleotidyltransferases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Pyrimidine Dimers/metabolism , Replication Protein A/analysis , Ubiquitin-Protein Ligases , Ubiquitination
9.
Nucleic Acids Res ; 40(20): 10324-33, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22941650

ABSTRACT

Trinucleotide repeat (TNR) expansions cause at least 17 heritable neurological diseases, including Huntington's disease. Expansions are thought to arise from abnormal processing of TNR DNA by specific trans-acting proteins. For example, the DNA repair complex MutSß (MSH2-MSH3 heterodimer) is required in mice for on-going expansions of long, disease-causing alleles. A distinctive feature of TNR expansions is a threshold effect, a narrow range of repeat units (∼30-40 in humans) at which mutation frequency rises dramatically and disease can initiate. The goal of this study was to identify factors that promote expansion of threshold-length CTG•CAG repeats in a human astrocytic cell line. siRNA knockdown of the MutSß subunits MSH2 or MSH3 impeded expansions of threshold-length repeats, while knockdown of the MutSα subunit MSH6 had no effect. Chromatin immunoprecipitation experiments indicated that MutSß, but not MutSα, was enriched at the TNR. These findings imply a direct role for MutSß in promoting expansion of threshold-length CTG•CAG tracts. We identified the class II deacetylase HDAC5 as a novel promoting factor for expansions, joining the class I deacetylase HDAC3 that was previously identified. Double knockdowns were consistent with the possibility that MutSß, HDAC3 and HDAC5 act through a common pathway to promote expansions of threshold-length TNRs.


Subject(s)
DNA-Binding Proteins/physiology , Histone Deacetylases/physiology , MutS Homolog 2 Protein/physiology , Trinucleotide Repeat Expansion , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Histone Deacetylases/genetics , Humans , MutS Homolog 2 Protein/antagonists & inhibitors , MutS Homolog 2 Protein/genetics , MutS Homolog 3 Protein
10.
J Immunol ; 186(4): 2336-43, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21242524

ABSTRACT

In the absence of core nonhomologous end-joining (NHEJ) factors, Ab gene class-switch recombination (CSR) uses an alternative end-joining (A-EJ) pathway to recombine switch (S) region DNA breaks. Previous reports showing decreased S-junction microhomologies in MSH2-deficient mice and an exonuclease 1 (EXO1) role in yeast microhomology-mediated end joining suggest that mismatch repair (MMR) proteins might influence A-EJ-mediated CSR. We have directly investigated whether MMR proteins collectively or differentially influence the A-EJ mechanism of CSR by analyzing CSR in mice deficient in both XRCC4 and individual MMR proteins. We find CSR is reduced and that Igh locus chromosome breaks are reduced in the MMR/XRCC4 double-deficient B cells compared with B cells deficient in XRCC4 alone, suggesting MMR proteins function upstream of double-strand break formation to influence CSR efficiency in these cells. Our results show that MLH1, EXO1, and MSH2 are all important for efficient A-EJ-mediated CSR, and we propose that MMR proteins convert DNA nicks and point mutations into dsDNA breaks for both C-NHEJ and A-EJ pathways of CSR. We also find Mlh1-XRCC4(-) B cells have an increased frequency of direct S junctions, suggesting that MLH1 proteins may have additional functions that influence A-EJ-mediated CSR.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , B-Lymphocyte Subsets/metabolism , DNA Repair/genetics , DNA-Binding Proteins/deficiency , Exodeoxyribonucleases/physiology , Immunoglobulin Class Switching/genetics , MutS Homolog 2 Protein/physiology , Nuclear Proteins/physiology , Animals , Cells, Cultured , DNA Breaks, Double-Stranded , DNA Damage , DNA-Binding Proteins/genetics , Deoxyribonucleases, Type II Site-Specific , Mice , Mice, Knockout , Mice, Transgenic , MutL Protein Homolog 1 , Point Mutation
11.
Carcinogenesis ; 33(9): 1647-54, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22739024

ABSTRACT

The hMSH2(M688R) mismatch repair (MMR) gene mutation has been found in five large families from Tenerife, Spain, suggesting it is a Lynch syndrome or hereditary non-polyposis colorectal cancer (LS/HNPCC) founder mutation. In addition to classical LS/HNPCC tumors, these families present with a high incidence of central nervous system (CNS) tumors normally associated with Turcot or constitutional mismatch repair deficiency (CMMR-D) syndromes. Turcot and CMMR-D mutations may be biallelic, knocking out both copies of the MMR gene. The hMSH2(M688R) mutation is located in the ATP hydrolysis (ATPase) domain. We show that the hMSH2(M688R)-hMSH6 heterodimer binds to mismatched nucleotides but lacks normal ATP functions and inhibits MMR in vitro when mixed with the wild-type (WT) heterodimer. Another alteration that has been associated with LS/HNPCC, hMSH2(M688I)-hMSH6, displays no identifiable differences with the WT heterodimer. Interestingly, some extracolonic tumors from hMSH2(M688R) carriers may express hMSH2-hMSH6, yet display microsatellite instability (MSI). The functional analysis along with variability in tumor expression and the high incidence of CNS tumors suggests that hMSH2(M688R) may act as a dominant negative in some tissues, while the hMSH2(M688I) is most likely a benign polymorphism.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , MutS Homolog 2 Protein/genetics , Mutation , Amino Acid Sequence , DNA Mismatch Repair , DNA-Binding Proteins/genetics , Humans , Immunohistochemistry , Molecular Sequence Data , MutS Homolog 2 Protein/analysis , MutS Homolog 2 Protein/physiology
12.
World J Surg Oncol ; 10: 264, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23231927

ABSTRACT

BACKGROUND: Vasoactive intestinal polypeptide secreting tumors(VIPomas) are rare endocrine tumors of the pancreas with an estimated incidence of 0.1 per million per year. The molecular mechanisms that mediate development of VIPomas are poorly investigated and require definition. METHODS: A genome- and gene expression analysis of specimens of a primary pancreatic VIPoma with hepatic metastases was performed. The primary tumor, the metastases, the corresponding healthy tissue of the liver, and the pancreas were compared with each other using oligonucleotide microarrays and loss of heterozygosity (LOH). RESULTS: The results revealed multiple LOH events and several differentially expressed genes. Our finding of LOH and downregulation was conspicuous in the microarray analysis for the mismatch repair gene MSH2 in the primary pancreatic VIPoma tumor, the hepatic metastasis but not in the corresponding healthy tissue. Further a strong overexpression of the chemokine CXCR4 was detected in the hepatic metastases compared to its pancreatic primary. With a review of the literature we describe the molecular insights of metastatic development in VIPoma. CONCLUSION: In VIPoma, defects in the mismatch repair system especially in MSH2 may contribute to carcinogenesis, and increased CXCR4 may be associated with liver metastasis.


Subject(s)
MutS Homolog 2 Protein/physiology , Pancreatic Neoplasms/genetics , Receptors, CXCR4/physiology , Vipoma/genetics , Aged , DNA Mismatch Repair/genetics , Humans , Loss of Heterozygosity , Male , Microsatellite Repeats , MutS Homolog 2 Protein/genetics , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , Receptors, CXCR4/genetics , Vipoma/etiology , Vipoma/pathology
13.
Carcinogenesis ; 32(7): 1085-92, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21551128

ABSTRACT

Loss of E-cadherin expression is a critical step in the development and progression of gynecological tumors. Study of the precise role of E-cadherin has been hampered by the lack of satisfactory mouse model for E-cadherin deficiency. Likewise, DNA mismatch repair (MMR) is implicated in gynecological tumorigenesis, but knockout of MMR in mice predominantly causes hematologic neoplasms. Here, we show that combined disruption of E-cadherin and DNA MMR pathways increases incidence of endometrioid tumors in mice. Twenty percent of mice knockout for Msh2 enzyme and hemizygous for E-cadherin [Msh2(-/-)/Cdh1(+/-)] developed endometrioid-like tumors in the ovary, uterus and genital area. Characteristic of these tumors was a complete loss of E-cadherin expression. Sequence analysis of E-cadherin promoter region demonstrated that the loss of E-cadherin expression is caused by inactivating mutations, implying that E-cadherin is a mutational target in Msh2-deficient mice. In addition, Msh2(-/-)/Cdh1(+/-) mice showed a reduction in overall survival as compared with their Msh2(-/-) counterparts due to the development of more aggressive lymphomas, suggesting a specific role of E-cadherin in lymphomagenesis. In conclusion, Msh2(-/-)/Cdh1(+/-) mice provide a good model of gynecological tumorigenesis and may be useful for testing molecular target-specific therapies.


Subject(s)
Base Pair Mismatch , Cadherins/genetics , Endometrial Neoplasms/epidemiology , Promoter Regions, Genetic , Animals , Base Sequence , Blotting, Western , DNA Primers , Endometrial Neoplasms/genetics , Female , Incidence , Mice , Mice, Inbred C57BL , Mice, Knockout , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/physiology
14.
Cancer Res ; 81(10): 2760-2773, 2021 05 15.
Article in English | MEDLINE | ID: mdl-34003775

ABSTRACT

Lynch syndrome is the most common cause of hereditary colorectal cancer and is secondary to germline alterations in one of four DNA mismatch repair (MMR) genes. Here we aimed to provide novel insights into the initiation of MMR-deficient (MMRd) colorectal carcinogenesis by characterizing the expression profile of MMRd intestinal stem cells (ISC). A tissue-specific MMRd mouse model (Villin-Cre;Msh2 LoxP/LoxP ) was crossed with a reporter mouse (Lgr5-EGFP-IRES-creERT2) to trace and isolate ISCs (Lgr5+) using flow cytometry. Three different ISC genotypes (Msh2-KO, Msh2-HET, and Msh2-WT) were isolated and processed for mRNA-seq and mass spectrometry, followed by bioinformatic analyses to identify expression signatures of complete MMRd and haplo-insufficiency. These findings were validated using qRT-PCR, IHC, and whole transcriptomic sequencing in mouse tissues, organoids, and a cohort of human samples, including normal colorectal mucosa, premalignant lesions, and early-stage colorectal cancers from patients with Lynch syndrome and patients with familial adenomatous polyposis (FAP) as controls. Msh2-KO ISCs clustered together with differentiated intestinal epithelial cells from all genotypes. Gene-set enrichment analysis indicated inhibition of replication, cell-cycle progression, and the Wnt pathway and activation of epithelial signaling and immune reaction. An expression signature derived from MMRd ISCs successfully distinguished MMRd neoplastic lesions of patients with Lynch syndrome from FAP controls. SPP1 was specifically upregulated in MMRd ISCs and colocalized with LGR5 in Lynch syndrome colorectal premalignant lesions and tumors. These results show that expression signatures of MMRd ISC recapitulate the initial steps of Lynch syndrome carcinogenesis and have the potential to unveil novel biomarkers of early cancer initiation. SIGNIFICANCE: The transcriptomic and proteomic profile of MMR-deficient intestinal stem cells displays a unique set of genes with potential roles as biomarkers of cancer initiation and early progression.


Subject(s)
Carcinogenesis/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Mismatch Repair , Gene Expression Regulation, Neoplastic , Intestines/physiopathology , Stem Cells/pathology , Transcriptome , Animals , Apoptosis , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , MutS Homolog 2 Protein/physiology , Prognosis , Proteome/analysis , Proteome/metabolism , Receptors, G-Protein-Coupled/physiology , Stem Cells/metabolism , Survival Rate , Tumor Cells, Cultured
15.
Br J Cancer ; 102(6): 1068-73, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20160730

ABSTRACT

BACKGROUND: The target substrates of DNA mismatch recognising factors MutSalpha (MSH2+MSH6) and MutSbeta (MSH2+MSH3) have already been widely researched. However, the extent of their functional redundancy and clinical substance remains unclear. Mismatch repair (MMR)-deficient tumours are strongly associated with microsatellite instability (MSI) and the degree and type of MSI seem to be dependent on the MMR gene affected, and is linked to its substrate specificities. Deficiency in MSH2 and MSH6 is associated with both mononucleotide and dinucleotide repeat instability. Although no pathogenic MSH3 mutations have been reported, its deficiency is also suggested to cause low dinucleotide repeat instability. METHODS: To assess the substrate specificities and functionality of MutSalpha and MutSbeta we performed an in vitro MMR assay using three substrate constructs, GT mismatch, 1 and 2 nucleotide insertion/deletion loops (IDLs) in three different cell lines. RESULTS: Our results show that though MutSalpha alone seems to be responsible for GT and IDL1 repair, MutSalpha and MutSbeta indeed have functional redundancy in IDL2 repair and in contrast with earlier studies, MutSbeta seems to exceed MutSalpha. CONCLUSION: The finding is clinically relevant because the strong role of MutSbeta in IDL2 repair indicates MSH3 deficiency in tumours with low dinucleotide and no mononucleotide repeat instability.


Subject(s)
DNA Mismatch Repair/genetics , DNA-Binding Proteins/physiology , Dinucleotide Repeats/genetics , MutS Homolog 2 Protein/physiology , Nucleic Acid Conformation , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , HCT116 Cells , HeLa Cells , Humans , INDEL Mutation/genetics , MutS Homolog 2 Protein/genetics , MutS Homolog 3 Protein , Mutation, Missense/physiology , Spodoptera , Substrate Specificity
16.
Proc Natl Acad Sci U S A ; 104(26): 10956-61, 2007 Jun 26.
Article in English | MEDLINE | ID: mdl-17573527

ABSTRACT

Msh2-Msh3 and Msh2-Msh6 are two partially redundant mispair-recognition complexes that initiate mismatch repair in eukaryotes. Crystal structures of the prokaryotic homolog MutS suggest the mechanism by which Msh6 interacts with mispairs because key mispair-contacting residues are conserved in these two proteins. Because Msh3 lacks these conserved residues, we constructed a series of mutants to investigate the requirements for mispair interaction by Msh3. We found that a chimeric protein in which the mispair-binding domain (MBD) of Msh6 was replaced by the equivalent domain of Msh3 was functional for mismatch repair. This chimera possessed the mispair-binding specificity of Msh3 and revealed that communication between the MBD and the ATPase domain is conserved between Msh2-Msh3 and Msh2-Msh6. Further, the chimeric protein retained Msh6-like properties with respect to genetic interactions with the MutL homologs and an Msh2 MBD deletion mutant, indicating that Msh3-like behaviors beyond mispair specificity are not features controlled by the MBD.


Subject(s)
DNA-Binding Proteins/genetics , Fungal Proteins/genetics , MutS Homolog 2 Protein/genetics , Saccharomyces cerevisiae Proteins/genetics , Base Pair Mismatch , Binding Sites/genetics , Conserved Sequence , DNA Repair , DNA-Binding Proteins/physiology , Fungal Proteins/physiology , MutS Homolog 2 Protein/physiology , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Recombinant Fusion Proteins , Saccharomyces cerevisiae Proteins/physiology
17.
Biochemistry ; 48(40): 9492-502, 2009 Oct 13.
Article in English | MEDLINE | ID: mdl-19320425

ABSTRACT

The observation that Cadmium (Cd(2+)) inhibits Msh2-Msh6, which is responsible for identifying base pair mismatches and other discrepancies in DNA, has led to the proposal that selective targeting of this protein and consequent suppression of DNA repair or apoptosis promote the carcinogenic effects of the heavy metal toxin. It has been suggested that Cd(2+) binding to specific sites on Msh2-Msh6 blocks its DNA binding and ATPase activities. To investigate the mechanism of inhibition, we measured Cd(2+) binding to Msh2-Msh6, directly and by monitoring changes in protein structure and enzymatic activity. Global fitting of the data to a multiligand binding model revealed that binding of about 100 Cd(2+) ions per Msh2-Msh6 results in its inactivation. This finding indicates that the inhibitory effect of Cd(2+) occurs via a nonspecific mechanism. Cd(2+) and Msh2-Msh6 interactions involve cysteine sulfhydryl groups, and the high Cd(2+):Msh2-Msh6 ratio implicates other ligands such as histidine, aspartate, glutamate, and the peptide backbone as well. Our study also shows that cadmium inactivates several unrelated enzymes similarly, consistent with a nonspecific mechanism of inhibition. Targeting of a variety of proteins, including Msh2-Msh6, in this generic manner would explain the marked broad-spectrum impact of Cd(2+) on biological processes. We propose that the presence of multiple nonspecific Cd(2+) binding sites on proteins and their propensity to change conformation on interaction with Cd(2+) are critical determinants of the susceptibility of corresponding biological systems to cadmium toxicity.


Subject(s)
Base Pair Mismatch , Cadmium/chemistry , DNA Repair , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/physiology , MutS Homolog 2 Protein/antagonists & inhibitors , MutS Homolog 2 Protein/physiology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/physiology , Adenosine Triphosphate/metabolism , Binding Sites , Cadmium/physiology , Cadmium Chloride/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Hydrolysis , MutS Homolog 2 Protein/chemistry , Protein Binding , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/chemistry
18.
Mutat Res ; 674(1-2): 123-30, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-18955159

ABSTRACT

Ionizing radiation (IR) induces two classes of complex DNA damage, double-strand breaks (DSBs) and non-DSB bi-stranded oxidative clustered DNA lesions (OCDLs). OCDLs may consist of single strand breaks (SSBs), oxidized purines/pyrimidines and abasic sites within 5-10bp. These significant biological lesions are hypothesized to challenge the repair machinery and carry a high mutagenic potential. MSH2, a classical DNA mismatch repair protein, has been also implicated in other repair pathways associated with DSB and base lesion processing. MSH2 mutations have been identified in acute lymphoblastic leukemia (ALL) patients as well as in other types of cancers. Our research model involves two precursors B (pre-B) ALL human cell lines, NALM-6 cells, homozygous null for MSH2, and wild type 697 cells. Using a modified version of neutral and alkaline single cell gel electrophoresis (SCGE) with Escherichia coli repair enzymes as damage probes, the processing capacity of single strand breaks (SSBs), DSBs and OCDLs was assessed in NALM-6 and 697 cells exposed to a radiotherapy relevant gamma-ray dose of 5Gy. Using reverse transcriptase PCR and Western blotting we verified the complete lack of expression of MSH2 in the NALM-6 cells at the transcriptional and translational level. No differences were measured between NALM-6 and 697 cells in the induction levels of SSBs, DSBs and OCDLs after exposure to gamma-rays. However, 697 cells repaired each lesion more efficiently with significant differences observed after 1-3h post-irradiation. Lastly, our results indicate a significantly higher population of apoptotic 697 cells compared to NALM-6 cells 6-24h post-irradiation. Our studies suggest that MSH2 is probably involved in the processing of the biologically significant clustered DNA damages as well as the execution of apoptosis induced by ionizing radiation.


Subject(s)
Cell Line, Tumor , DNA Damage/genetics , DNA Repair/genetics , MutS Homolog 2 Protein/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Gamma Rays , Humans , Hydrogen Peroxide/pharmacology , Models, Biological , Multigene Family/radiation effects , MutS Homolog 2 Protein/physiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Radiotherapy Dosage
19.
Hepatogastroenterology ; 56(91-92): 899-904, 2009.
Article in English | MEDLINE | ID: mdl-19621725

ABSTRACT

BACKGROUND/AIMS: Microsatellite instability (MSI) is a manifestation of a defective DNA mismatch repair system. It is caused by germline mutations of mismatch repair genes or CpG islands hypermethylation. The majority of cancers of hereditary nonpolyposis colorectal cancer (HNPCC) syndrome have MSI+ phenotype. The colorectal cancers show distinctive clinicopathological characteristics and prognoses according to the MSI status. However, there is a wide variety of results between MSI and clinicopathological parameters in gastric carcinomas. METHODOLOGY: Five hundred and twenty-one surgically resected gastric carcinomas were studied and the correlation with clinicopathological parameters, MSI status by using five microsatellite markers, expression of hMLH1 and hMSH2 protein by immunohistochemical stain, and methylation of hMLH1 and hMSH2 by methylation-specific polymerase chain reaction was analyzed. RESULTS: There were 50 (9.6%) high-frequency MSI (MSI-H) cases. The MSI-H gastric carcinomas were associated with older age, expanding type by Ming's classification, lymphatic invasion, tumor multiplicity, losses of hMLH1 and hMSH2 protein expressions. The methylation frequency of hMLH1 was 75.5% in MSI-H gastric carcinomas. CONCLUSIONS: Our results suggest that epigenetic inactivation of hMLH1 might play a role in the carcinogenesis of MSI-H gastric carcinomas. The immunohistochemical stain for hMLH1 protein expression could be used in routine diagnostic methods for predicting MSI status.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Carcinoma/genetics , Microsatellite Instability , MutS Homolog 2 Protein/physiology , Nuclear Proteins/physiology , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Carcinoma/metabolism , Carcinoma/pathology , Cohort Studies , Female , Humans , Male , Middle Aged , MutL Protein Homolog 1 , Neoplasm Invasiveness , Retrospective Studies , Sensitivity and Specificity , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
20.
Gut ; 57(6): 780-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18272544

ABSTRACT

OBJECTIVE: Chronic inflammation in ulcerative colitis is associated with increased risk for colorectal cancer. Its molecular pathway of cancer development is poorly understood. We investigated the role of neutrophil-derived cellular stress in an in vitro model of neutrophils as effectors and colon epithelial cells as targets, and tested for changes in cell cycle distribution and the appearance of replication errors. DESIGN: Colon epithelial cells with different mismatch repair phenotypes were co-cultured with activated neutrophils. Target cells were analysed for cell cycle distribution and replication errors by flow cytometry. Changes in nuclear and DNA-bound levels of mismatch repair- and checkpoint-related proteins were analysed by western blotting. RESULTS: Activated neutrophils cause an accumulation of target cells in G2/M, consistent with the installation of a DNA-damage checkpoint. Cells that do not express hMSH2, p53 or p21(waf1/cip1) failed to undergo the G2/M arrest. Phosphorylation of p53 at site Ser15 and Chk1 at Ser317, as well as accumulation of p21(waf1/cip1), was observed within 8-24 h. Superoxide dismutase and catalase were unable to overcome this G2/M arrest, possibly indicating that neutrophil products other than superoxide or H(2)O(2) are involved in this cellular response. Finally, exposure to activated neutrophils increased the number of replication errors. CONCLUSIONS: By using an in vitro co-culture model that mimics intestinal inflammation in ulcerative colitis, we provide molecular evidence for an hMSH2-dependent G2/M checkpoint arrest and for the presence of replication errors.


Subject(s)
Colitis, Ulcerative/pathology , Colon/pathology , MutS Homolog 2 Protein/physiology , Neutrophil Activation , Catalase/pharmacology , Cell Cycle/drug effects , Cell Cycle/physiology , Checkpoint Kinase 1 , Coculture Techniques , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Colon/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Mismatch Repair , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Phosphorylation/drug effects , Protein Kinases/metabolism , Superoxide Dismutase/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL