Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Med Teach ; 36(9): 804-11, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24804919

ABSTRACT

Several influential national/international bodies including The Bologna Accord, The Carnegie Foundation and The Future of Medical Education in Canada (FMEC) have called for increased coordination across the medical education continuum. FMEC recognizes accreditation as a "powerful lever" and encourages the alignment of undergraduate and postgraduate standards. The Carnegie Foundation includes a similar call for the creation of a more coherent accreditation system. As a first step, using the Canadian context, we present a methodological approach that assesses the example of how well LCME/CACMS undergraduate accreditation standards align with the Royal College of Physician and Surgeons of Canada (RCPSC) postgraduate training standards. We analyzed how closely the 132 LCME/CACMS Medical School accreditation standards aligned with the 155 post-graduate standards from the RCPSC accreditation General Standards (A and B). This comparative evaluation demonstrates that the standards do not align closely. Gaps, redundancies and key differences are highlighted. These results are the first step in understanding how accreditation needs to be adapted and re-aligned across the education continuum to provide consistent and coordinated training and these methods could easily be applied to other contexts and jurisdictions.


Subject(s)
Accreditation/organization & administration , Education, Medical, Continuing/standards , Education, Medical, Undergraduate/standards , Accreditation/standards , Canada , Clinical Competence/standards , Humans
2.
Am J Pathol ; 179(1): 411-21, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703420

ABSTRACT

The fusion tyrosine kinase NPM-ALK is central to the pathogenesis of ALK-positive anaplastic large cell lymphoma (ALK(+)ALCL). We recently identified that MSH2, a key DNA mismatch repair (MMR) protein integral to the suppression of tumorigenesis, is an NPM-ALK-interacting protein. In this study, we found in vitro evidence that enforced expression of NPM-ALK in HEK293 cells suppressed MMR function. Correlating with these findings, six of nine ALK(+)ALCL tumors displayed evidence of microsatellite instability, as opposed to none of the eight normal DNA control samples (P = 0.007, Student's t-test). Using co-immunoprecipitation, we found that increasing levels of NPM-ALK expression in HEK293 cells resulted in decreased levels of MSH6 bound to MSH2, whereas MSH2·NPM-ALK binding was increased. The NPM-ALK·MSH2 interaction was dependent on the activation/autophosphorylation of NPM-ALK, and the Y191 residue of NPM-ALK was a crucial site for this interaction and NPM-ALK-mediated MMR suppression. MSH2 was found to be tyrosine phosphorylated in the presence of NPM-ALK. Finally, NPM-ALK impeded the expected DNA damage-induced translocation of MSH2 out of the cytoplasm. To conclude, our data support a model in which the suppression of MMR by NPM-ALK is attributed to its ability to interfere with normal MSH2 biochemistry and function.


Subject(s)
DNA Mismatch Repair , DNA-Binding Proteins/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large-Cell, Anaplastic/pathology , MutS Homolog 2 Protein/metabolism , Protein-Tyrosine Kinases/metabolism , Cytoplasm/metabolism , DNA Damage , DNA-Binding Proteins/genetics , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/metabolism , Microsatellite Instability , MutS Homolog 2 Protein/genetics , Phosphorylation , Protein Multimerization , Protein Transport , Protein-Tyrosine Kinases/genetics , Tumor Cells, Cultured , Tyrosine/metabolism
3.
Pigment Cell Melanoma Res ; 23(3): 430-40, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20337986

ABSTRACT

The mitogen-activated protein kinase (MAPK) pathway is constitutively activated in the majority of melanomas, promoting cell survival, proliferation and migration. In addition, anti-apoptotic Bcl-2 family proteins Mcl-1, Bcl-xL and Bcl-2 are frequently overexpressed, contributing to melanoma's well-documented chemoresistance. Recently, it was reported that the combination of MAPK pathway inhibition by specific MEK inhibitors and Bcl-2 family inhibition by BH3-mimetic ABT-737 synergistically induces apoptotic cell death in melanoma cell lines. Here we provide the first evidence that inhibition of another key MAPK, p38, synergistically induces apoptosis in melanoma cells in combination with ABT-737. We also provide novel mechanistic data demonstrating that inhibition of p38 increases expression of pro-apoptotic Bcl-2 protein PUMA. Furthermore, we demonstrate that PUMA can be cleaved by a caspase-dependent mechanism during apoptosis and identify what appears to be the PUMA cleavage product. Thus, our findings suggest that the combination of ABT-737 and inhibition of p38 is a promising, new treatment strategy that acts through a novel PUMA-dependent mechanism.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Biphenyl Compounds/pharmacology , Melanoma/pathology , Nitrophenols/pharmacology , Proto-Oncogene Proteins/metabolism , Skin Neoplasms/pathology , Sulfonamides/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Caspases/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Enzyme Activation/drug effects , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Melanoma/enzymology , Myeloid Cell Leukemia Sequence 1 Protein , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Pyridines/pharmacology , Skin Neoplasms/enzymology , Up-Regulation/drug effects
4.
DNA Repair (Amst) ; 9(2): 161-8, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-20022306

ABSTRACT

Many studies have shown that DNA mismatch repair (MMR) has a role beyond that of repair in response to several types of DNA damage, including ultraviolet radiation (UV). We have demonstrated previously that the MMR-dependent component of UVB-induced apoptosis is integral to the suppression of UVB-induced tumorigenesis. Here we demonstrate that Msh6-dependent UVB-induced apoptotic pathway is both activated via the mitochondria and p53-independent. In addition, we have shown for the first time that caspase 2, an initiator caspase, localizes to the centrosomes in mitotic primary mouse embryonic fibroblasts, irrespective of MMR status and UVB treatment.


Subject(s)
Apoptosis , Caspase 2/metabolism , Centrosome/enzymology , DNA Mismatch Repair , Animals , Apoptosis/radiation effects , Caspase 9/metabolism , Cell Extracts , Cells, Cultured , Centrosome/radiation effects , Cytochromes c/metabolism , DNA Mismatch Repair/radiation effects , DNA-Binding Proteins/metabolism , Enzyme Activation/radiation effects , Membrane Potential, Mitochondrial/radiation effects , Mice , Microscopy, Confocal , Mitochondria/enzymology , Mitochondria/radiation effects , MutS Homolog 2 Protein/metabolism , Protein Transport/radiation effects , Receptors, Death Domain/metabolism , Signal Transduction/radiation effects , Subcellular Fractions/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , bcl-2-Associated X Protein/metabolism
5.
PLoS One ; 4(8): e6651, 2009 Aug 17.
Article in English | MEDLINE | ID: mdl-19684859

ABSTRACT

BACKGROUND: Malignant melanoma is resistant to almost all conventional forms of chemotherapy. Recent evidence suggests that anti-apoptotic proteins of the Bcl-2 family are overexpressed in melanoma and may contribute to melanoma's striking resistance to apoptosis. ABT-737, a small-molecule inhibitor of Bcl-2, Bcl-xl and Bcl-w, has demonstrated efficacy in several forms of leukemia, lymphoma as well as solid tumors. However, overexpression of Mcl-1, a frequent observance in melanoma, is known to confer ABT-737 resistance. METHODOLOGY/PRINCIPAL FINDINGS: Here we report that knockdown of Mcl-1 greatly reduces cell viability in combination with ABT-737 in six different melanoma cell lines. We demonstrate that the cytotoxic effect of this combination treatment is due to apoptotic cell death involving not only caspase-9 activation but also activation of caspase-8, caspase-10 and Bid, which are normally associated with the extrinsic pathway of apoptosis. Caspase-8 (and caspase-10) activation is abrogated by inhibition of caspase-9 but not by inhibitors of the death receptor pathways. Furthermore, while caspase-8/-10 activity is required for the full induction of cell death with treatment, the death receptor pathways are not. Finally, we demonstrate that basal levels of caspase-8 and Bid correlate with treatment sensitivity. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that the combination of ABT-737 and Mcl-1 knockdown represents a promising, new treatment strategy for malignant melanoma. We also report a death receptor-independent role for extrinsic pathway proteins in treatment response and suggest that caspase-8 and Bid may represent potential markers of treatment sensitivity.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Caspase 8/metabolism , Melanoma/pathology , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , Sulfonamides/pharmacology , Caspase 8/genetics , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Melanoma/enzymology , Melanoma/genetics , Myeloid Cell Leukemia Sequence 1 Protein , Piperazines/pharmacology , Polymorphism, Genetic
6.
Oncol Rep ; 18(6): 1403-11, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982623

ABSTRACT

The multi-functionality of the DNA mismatch repair (MMR) proteins has been demonstrated by their role in regulation of the cell cycle and apoptosis, as well as DNA repair. Using a unique MSH2-/- non-tumor human lymphoblastoid cell line we show that MMR facilitates G2/M arrest after UVB-induced DNA damage. Deficiency in MSH2 leads to a decrease in the induction of G2/M cell cycle checkpoint following UVB radiation in MSH2-null non-tumor cells. We also show evidence that the above-mentioned cells deficient in MSH2 have decreased levels of key cell cycle proteins such as CHK1 phosphorylated at Ser345, CDC25C phosphorylated at Ser216 and CDC2 phosphorylated at Tyr15, Thr14, compared to MSH2-proficient cells after UVB radiation. In addition, we demonstrate an altered p53 protein in the MSH2-null cell line. Our data show that the MMR protein MSH2 is involved in the regulation of normal cell cycle response after UVB-induced DNA damage.


Subject(s)
Cell Cycle/genetics , MutS Homolog 2 Protein/deficiency , Ultraviolet Rays , Cell Cycle/radiation effects , Cell Division , Cell Line , DNA Damage/radiation effects , DNA Mismatch Repair , Flow Cytometry , G2 Phase , Humans , Lymphocytes/physiology , Phosphorylation , Tumor Suppressor Protein p53/metabolism
7.
Carcinogenesis ; 28(10): 2131-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17615258

ABSTRACT

DNA mismatch repair (MMR) is a highly conserved system that repairs DNA adducts acquired during replication, as well as some forms of exogenous/endogenous DNA damage. Additionally, MMR proteins bind to DNA adducts that are not removed by MMR and influence damage-response mechanisms other than repair. Hereditary non-polyposis colorectal cancer, as well as mouse models for MMR deficiency, illustrate that MMR proteins are required for maintenance of genetic stability and tumor suppression. In both humans and mice, the phenotype associated with Msh6-associated tumorigenesis is distinct from that of Msh2. In this study, we hypothesized that Msh6-/-;p53+/- mice would display earlier tumor onset than their Msh6-/- or p53+/- counterparts, indicating that concomitant loss of these two tumor suppressors contributes to tumorigenesis via mechanisms that are only partially interrelated. We generated a Msh6-/-;p53+/- mouse model which succumbed to malignant disease at an accelerated rate and with a tumor spectrum distinct from both Msh6-/- and p53+/- models. Alteration of tumor phenotype in the Msh6-/-;p53+/- mice included a marked increase in microsatellite instability that was associated with loss of heterozygosity of the remaining p53 allele. Also, genetic instability was inversely correlated with survival. This manuscript marks the first in vivo investigation into the association between Msh6 and p53, and their combined role in the suppression of spontaneous tumorigenesis, cell survival and genomic stability. Our results support the hypothesis that p53 and Msh6 are functionally interrelated and that, with concomitant mutation, these tumor suppressors act together to accelerate tumorigenesis.


Subject(s)
DNA Mismatch Repair , DNA-Binding Proteins/physiology , Tumor Suppressor Protein p53/physiology , Animals , Chromosomal Instability , Conserved Sequence , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Death , Genes, p53 , Genotype , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Loss of Heterozygosity , Mice , Mice, Knockout , Microsatellite Repeats , Splenic Neoplasms/genetics , Splenic Neoplasms/pathology , Thymus Neoplasms/genetics , Thymus Neoplasms/pathology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
8.
Cancer Res ; 65(7): 2626-35, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15805259

ABSTRACT

Inheritance of a germline mutation in one of the DNA mismatch repair genes predisposes human individuals to hereditary nonpolyposis colorectal cancer, characterized by development of tumors predominantly in the colon, endometrium, and gastrointestinal tract. Mice heterozygous for a mismatch repair-null mutation generally do not have an increased risk of neoplasia. However, mice constitutively lacking mismatch repair are prone to tumor development from an early age, particularly thymic lymphomas. Mismatch repair-deficient mice crossed to Apc(+/-) mice develop an increased spontaneous intestinal tumor incidence, demonstrating that the tumor spectrum can be genetically influenced. Here, we bred Msh2- and Msh6-deficient mice to athymic nude mice, hypothesizing that a broader tumor spectrum may be observed if mice are able to survive longer without succumbing to thymic lymphomas. However, Msh2(-/-);Foxn1(nu/nu) and Msh6(-/-);Foxn1(nu/nu) mice developed primarily early-onset lymphoblastic lymphomas. Using B-cell-specific markers, we found these tumors to be predominately B-cell in origin. The development of hematologic malignancy in the mouse, even in the absence of a thymus, parallels the development of B- and T-cell lymphoma and leukemia in the few rare mismatch repair-null human patients that have been identified. The persistent development of hematologic malignancy both in the mouse and in human patients deficient in mismatch repair leads us to implicate mismatch repair as an important repair mechanism in normal B- and T-cell development. Thus, mismatch repair-deficient mice may prove to be a good model to study human hematologic malignancy.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/deficiency , Lymphoma, B-Cell/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/deficiency , Animals , Colon/pathology , DNA-Binding Proteins/genetics , Female , Forkhead Transcription Factors , Gene Rearrangement, B-Lymphocyte, Heavy Chain/genetics , Genetic Predisposition to Disease , Immunohistochemistry , Male , Mice , Mice, Knockout , Mice, Nude , Microsatellite Repeats/genetics , MutS Homolog 2 Protein , Proto-Oncogene Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
9.
Carcinogenesis ; 25(10): 1821-7, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15166087

ABSTRACT

DNA mismatch repair (MMR) proteins are integral to the maintenance of genomic stability and suppression of tumorigenesis due to their role in repair of post-replicative DNA errors. Recent data also support a role for MMR proteins in cellular responses to exogenous DNA damage that does not involve removal of DNA adducts. We have demonstrated previously that both Msh2- and Msh6-null primary mouse embryonic fibroblasts are significantly less sensitive to UVB (ultraviolet B)-induced cytotoxicity and apoptosis than wild-type control cells. In order to ascertain the physiological relevance of the data we have exposed MMR-deficient mice to acute and chronic UVB radiation. We found that MMR-deficiency was associated with reduced levels of apoptosis and increased residual UVB-induced DNA adducts in the epidermis 24-h following acute UVB exposure. Moreover, Msh2-null mice developed UVB-induced skin tumors at a lower level of cumulative UVB exposure and with a greater severity of onset than wild-type mice. The Msh2-null skin tumors did not display microsatellite instability, suggesting that these tumors develop via a different tumorigenic pathway than tumors that develop spontaneously. Therefore, we propose that dysfunctional MMR promotes UVB-induced tumorigenesis through reduced apoptotic elimination of damaged epidermal cells.


Subject(s)
Apoptosis/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/physiology , Epidermis/radiation effects , Neoplasms, Radiation-Induced/pathology , Proto-Oncogene Proteins/physiology , Skin Neoplasms/pathology , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , DNA Adducts , DNA-Binding Proteins/genetics , Epidermal Cells , Epidermis/physiology , Humans , Mice , Mice, Knockout , Microsatellite Repeats , MutS Homolog 2 Protein , Neoplasm Invasiveness/pathology , Neoplasms, Radiation-Induced/metabolism , Proto-Oncogene Proteins/genetics , Pyrimidine Dimers , Skin Neoplasms/metabolism , Ultraviolet Rays
10.
J Invest Dermatol ; 121(4): 876-80, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14632208

ABSTRACT

Recent data support a role for DNA mismatch repair in the cellular response to some forms of exogenous DNA damage beyond that of DNA repair; cells with defective DNA mismatch repair have partial or complete failure to undergo apoptosis and/or G2M arrest following specific types of damage. We propose that the DNA mismatch repair Msh2/Msh6 heterodimer, responsible for the detection of DNA damage, promotes apoptosis in normal cells, thus protecting mammals from ultraviolet-induced malignant transformation. Using primary mouse embryonic fibroblasts derived from Msh6+/+ and Msh6-/- mice, we compare the response of DNA-mismatch repair-proficient and -deficient cells to ultraviolet B radiation. In the wild-type mouse embryonic fibroblasts, ultraviolet-B-induced increases in Msh6 protein levels were not dependent on p53. Msh6-/- mouse embryonic fibroblasts were significantly less sensitive to the cytotoxic effects of ultraviolet B radiation. Further comparison of the Msh6+/+ and Msh6-/- mouse embryonic fibroblasts revealed that Msh6-/- mouse embryonic fibroblasts undergo significantly less apoptosis following ultraviolet B irradiation, thus indicating that ultraviolet-B-induced apoptosis is partially Msh6 dependent. These data support a role for Msh6 in protective cellular responses of primary cells to ultraviolet-B-induced mutagenesis and, hence, the prevention of skin cancer.


Subject(s)
Apoptosis/physiology , DNA Repair/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/physiology , Animals , Apoptosis/radiation effects , Cell Survival/physiology , Cell Survival/radiation effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/radiation effects , Mice , Mice, Knockout , Skin Neoplasms/prevention & control , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays/adverse effects
11.
J Invest Dermatol ; 121(3): 435-40, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12925197

ABSTRACT

In addition to their established role in repairing post-replicative DNA errors, DNA mismatch repair proteins contribute to cell cycle arrest and apoptosis in response to a wide range of exogenous DNA damage (e.g., alkylation-induced lesions). The role of DNA mismatch repair in response to ultraviolet-induced DNA damage has been historically controversial. Recent data, however, suggest that DNA mismatch repair proteins probably do not contribute to the removal of ultraviolet-induced DNA damage, but may be important in suppressing mutagenesis, effecting apoptosis, and suppressing tumorigenesis following exposure to ultraviolet radiation.


Subject(s)
DNA Repair/radiation effects , DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic/radiation effects , Ultraviolet Rays/adverse effects , Animals , DNA Repair/physiology , Humans
12.
DNA Repair (Amst) ; 2(4): 427-35, 2003 Apr 02.
Article in English | MEDLINE | ID: mdl-12606123

ABSTRACT

DNA mismatch repair (MMR) is integral to the maintenance of genomic stability and more recently has been demonstrated to affect apoptosis and cell cycle arrest in response to a variety of adducts induced by exogenous agents. Comparing Msh2-null and wildtype mouse embryonic fibroblasts (MEFs), both primary and transformed, we show that Msh2 deficiency results in increased survival post-UVB, and that UVB-induced apoptosis is significantly reduced in Msh2-deficient cells. Furthermore, p53 phosphorylation at serine 15 is delayed or diminished in Msh2-deficient cells, suggesting that Msh2 may act upstream of p53 in a post-UVB apoptosis or growth arrest response pathway. Taken together, these data suggest that MMR heterodimers containing Msh2 may function as a sensor of UVB-induced DNA damage and influence the initiation of UVB-induced apoptosis, thus implicating MMR in protecting against UV-induced tumorigenesis.


Subject(s)
Apoptosis/physiology , DNA Damage/radiation effects , DNA Repair , DNA-Binding Proteins , Tumor Suppressor Protein p53/physiology , Animals , Mice , MutS Homolog 2 Protein , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Ultraviolet Rays/adverse effects
13.
Genes Chromosomes Cancer ; 35(4): 299-310, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12378524

ABSTRACT

Genomic instability is believed to play a significant role in cancer development by facilitating tumor progression and tumor heterogeneity. Inter-simple sequence repeat (inter-SSR) PCR has been proved to be a fast and reproducible technique for quantitation of genomic instability (amplifications, deletions, translocations, and insertions) in human sporadic tumors. However, the use of inter-SSR PCR in animal models of cancer has never been described. This new technique has been adapted in our laboratory for the analysis of spontaneous and induced mouse tumors. We established the best PCR conditions for each microsatellite-anchored primer and critically evaluated the reproducibility of the band patterns. We also studied the variation of the fingerprints between and within various inbred mouse strains, including wild-derived lines. Tumor-specific alterations were detected as gains, losses, or intensity changes in bands when compared with matched normal DNA. We quantitated the extent of alterations by dividing the number of altered bands in the tumor by the total number of bands in normal DNA (instability index). By means of inter-SSR PCR, we successfully analyzed genomic alterations in various mouse tumors, including spontaneous thymic lymphomas developed in Msh2 knockout mice as well as chemically induced squamous cell carcinomas and thymic lymphomas. Instability index values ranged between 0 and 9%, the highest levels observed in N-methyl-N-nitrosourea-induced thymic lymphomas generated in Trp53 (p53) nullizygote (-/-) mice. We report here, for the first time, the use of inter-SSR PCR to detect somatic mutations in mouse tumoral DNA, including laser-capture microdissected, methanol-fixed tissues. These PCR-based fingerprints provide a novel approach to assessing the number and onset of mutational events in mouse tumors and will help to understand better the mechanisms of carcinogenesis in mouse models.


Subject(s)
Disease Models, Animal , Mutation/genetics , Neoplasms/genetics , Polymerase Chain Reaction/methods , Repetitive Sequences, Nucleic Acid/genetics , Animals , Crosses, Genetic , DNA Fingerprinting/methods , DNA, Neoplasm/analysis , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Mice, Inbred NZB , Mice, Inbred SENCAR , Mice, Inbred Strains , Mice, Mutant Strains , Mice, Nude , Neoplasm Transplantation/methods , Neoplasms/chemically induced , Neoplasms/etiology , Tumor Cells, Cultured
14.
Cancer Res ; 62(2): 359-62, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11809679

ABSTRACT

Individuals with a germ-line mutation in one of the DNA mismatch repair (MMR) genes are at significant risk for colorectal cancer and other tumors. Three families have previously been reported with individuals homozygous for mutations in the MMR gene MLH1 that are predicted to compromise MMR. These individuals develop hematological malignancies and/or neurofibromatosis type 1 at an early age. Here, in an individual, we demonstrate that a homozygous novel mutation in the MMR gene MSH2 is associated with leukemia and multiple café-au-lait spots, a feature of neurofibromatosis type 1. Because the hematological malignancies observed in the individuals homozygous for the loss of MMR are reflective of the lymphomas seen in mice lacking MMR, the mice may provide a useful model for human neoplasia.


Subject(s)
Cafe-au-Lait Spots/genetics , DNA-Binding Proteins , Germ-Line Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing , Base Pair Mismatch , Carrier Proteins , Child, Preschool , DNA Repair , Exons/genetics , Female , Genetic Predisposition to Disease , Humans , Male , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/genetics , Neurofibromatosis 1/genetics , Nuclear Proteins , Pedigree
15.
Genet Test ; 6(4): 319-22, 2002.
Article in English | MEDLINE | ID: mdl-12537657

ABSTRACT

Hereditary non-polyposis colorectal cancer (HNPCC) is a common hereditary cancer. Genetic testing is complicated by the multiple DNA mismatch repair genes that underlie the disorder. Many suspected HNPCC families have no germ-line mutation identified. We reassessed an unusual family that appeared to have 2 individuals homozygous for a germline mutation within exon 1 of the hMLH1 gene. A few rare individuals with two inherited mutations in one of the mismatch repair genes have been reported and appear to have a distinct clinical appearance. However, there were no clinical features in the family discussed here that were consistent with constitutive lack of hMLH1. Redesigning the intronic primers for exon 1 identified a common polymorphism located within the original intronic primer site. The polymorphism prevented amplification of the wild-type allele, giving the erroneous appearance of homozygous inheritance of the mutated allele. Likewise, common intronic polymorphisms, if located within primer sequences on the chromosome harboring the HNPCC germ-line mutation could restrict amplification to only the wild-type allele, which may contribute significantly to the low success rate of identifying mutations in HNPCC families.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Introns , Neoplasm Proteins/genetics , Polymorphism, Genetic , Adaptor Proteins, Signal Transducing , Base Sequence , Carrier Proteins , DNA Primers , Female , Humans , Male , Molecular Sequence Data , MutL Protein Homolog 1 , Nuclear Proteins , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL