Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Biol Chem ; 276(3): 1961-7, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11053428

ABSTRACT

We have demonstrated previously that Cdc42 induced MLK-3 homodimerization leads to both autophosphorylation and activation of MLK-3 and postulated that autophosphorylation is an intermediate step of MLK-3 activation following its dimerization. In this report we sought to refine further the mechanism of MLK-3 activation and study the role of the putative kinase activation loop in MLK-3 activation. First we mutated the three potential phosphorylation sites in MLK-3 putative activation loop to alanine in an effort to abrogate MLK-3 autophosphorylation. Mutant T277A displayed almost no autophosphorylation activity and was nearly nonfunctional; mutant S281A, that displayed a low level of autophosphorylation, only slightly activated its downstream targets, whereas the T278A mutant, that exhibited autophosphorylation comparable to that of the wild type, was almost fully functional. Thus, these residues within the activation loop are critical for MLK-3 autophosphorylation and activation. In addition, when the Thr277 and Ser281 residues were mutated to negatively charged glutamic acid to mimic phosphorylated serine/threonine residues, the resulting mutants were fully functional, implying that these two residues may serve as the autophosphorylation sites. Interestingly, HPK1 also phosphorylated MLK-3 activation loop in vitro, and Ser281 was found to be the major phosphorylation site, indicating that HPK1 also activates MLK-3 via phosphorylation of the kinase activation loop.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Base Sequence , DNA Primers , Enzyme Activation , MAP Kinase Kinase Kinases/chemistry , MAP Kinase Kinase Kinases/genetics , Molecular Sequence Data , Mutagenesis , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Sequence Homology, Amino Acid , Mitogen-Activated Protein Kinase Kinase Kinase 11
2.
Methods Mol Med ; 61: 109-22, 2001.
Article in English | MEDLINE | ID: mdl-22323255

ABSTRACT

The first description of familial melanoma in the English literature appeared in 1820, when Norris (1) reported: It is remarkable that this gentleman's father, about thirty years ago, died of a similar disease.... This tumour, I have remarked, originated in a mole, and it is worth mentioning, that not only my patient and his children had many moles on various parts of their bodies, but also his own father and brothers had many of them.... These facts, together with a case that has come under my notice, rather similar, would incline me to believe that this disease is hereditary. Since then, many families with a predisposition to melanoma have been described worldwide (2-5). For purposes of case definition, our laboratory curently defines familial melanoma (FMM) as a family containing >2 affected first-degree relatives with melanoma and/or pancreatic carcinoma. According to this definition, about 8-12% of melanoma is inherited as an autosomal dominant trait with variable penetrance. Affected members (AFM) of these FMM kindreds may develop multiple primary melanoma (6) and/or pancreatic cancer (7) and typically present at an earlier age than do patients with sporadic disease. In a subset of such individuals and kindreds, germline mutations of the CDKN2A gene (also known as p16INK4A and MTS1) cosegregate with cases of melanoma (2-5).We have hypothesized that the identification of mutation carriers may in the future allow us to direct resources to the prevention and surveillance of mela noma in high-risk individuals and families. This chapter provides an overview of melanoma genetics, as well as the indications, drawbacks, and methods of germline CDKN2A mutation screening by polymerase chain reaction (PCR) amplification and automated sequencing of genomic DNA.

3.
J Pediatr Hematol Oncol ; 22(5): 451-3, 2000.
Article in English | MEDLINE | ID: mdl-11037859

ABSTRACT

The concurrence of acute lymphoblastic leukemia (ALL) and an asymptomatic juvenile pilocytic astrocytoma is described. A 6-year-old boy without clinical evidence of neurofibromatosis had a juvenile pilocytic astrocytoma diagnosed on radiologic examination and before treatment of acute pre-B cell lymphoblastic leukemia. The patient has had a partial resection of the astrocytoma and is 9 months into treatment of his ALL, which is in complete remission. p53 gene mutation was not identified in this patient. The concurrent diagnosis before treatment of ALL and juvenile pilocytic astrocytoma, the latter normally an indolent tumor, suggests that some cases of astrocytoma previously ascribed to radiotherapy or other treatment may in fact be caused by other factors.


Subject(s)
Astrocytoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Child , Genes, p53 , Humans , Male , Neoplasms, Second Primary/etiology
4.
Int J Cancer ; 87(4): 463-7, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10918183

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN 1) is a familial cancer syndrome associated primarily with endocrine tumors of the parathyroids, enteropancreas and anterior pituitary. However, tumors of mesenchymal origin such as angiofibroma and collagenoma of the skin have also been associated with the syndrome. This highlights the possibility of an association between MEN 1 and some other types of tumors. Here we report 7 cases of primary malignant melanoma occurring in 7 MEN 1 families, all patients exhibiting classic features of MEN 1. Based on these findings and the previous implication of multiple melanoma tumor suppressor(s) in 11q, including the MEN1 region, we have investigated the involvement of the MEN1 gene in melanoma tumorigenesis. Mutation analysis was performed on a panel of 39 sporadic metastatic melanomas, 13 melanoma cell lines and 20 melanoma families without CDKN2A or CDK4 germline mutations. In addition, 19 sporadic metastatic tumors were screened for loss of heterozygosity (LOH) in 11q13. LOH was detected in 6 tumors (32%), and in 4 of the tumors the pattern of LOH suggested that the deletion included the MEN1 gene locus. A novel somatic nonsense mutation in exon 7 (Q349X) was identified in 1 sporadic tumor which also showed loss of the wild-type allele. We conclude that the MEN1 gene plays a role in the tumorigenesis of a small subgroup of melanoma.


Subject(s)
Melanoma/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Skin Neoplasms/genetics , Adult , Aged , Chromosomes, Human, Pair 11/genetics , DNA Mutational Analysis , Female , Humans , Karyotyping , Loss of Heterozygosity , Male , Melanoma/pathology , Middle Aged , Mutation , Skin Neoplasms/pathology , Tumor Cells, Cultured
5.
Eur J Hum Genet ; 8(8): 590-6, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10951521

ABSTRACT

Germline mutations in the p16 (CDKN2A) tumour suppressor gene have been linked to inherited predisposition to malignant melanoma (MM). Variable frequencies of p16 germline mutations were reported in different collections of melanoma families but it can be as high as 50%. Here we describe the results of p16 mutation screening in 30 melanoma kindreds in Israel. The entire coding region of the p16 gene, including exons 1, 2 and 3, flanking exon/intron junctions, and a portion of the 3' untranslated (UTR) region of the gene were examined by single-stranded conformation polymorphism (SSCP) analysis and direct sequencing. Two p16 germline mutations were identified: G101W, which has been previously observed in a number of melanoma kindreds, and G122V, a novel missense mutation. Thus, the frequency of mutations identified in this collection of Israeli families was 7%. Functional analysis indicated that the novel G122V variant retained some capacity to interact with cyclin dependent kinases (CDKs) in vitro, yet it was significantly impaired in its ability to cause a G1 cell cycle arrest in human diploid fibroblasts. This partial loss of function is consistent with the predicted impact of G122V substitution on the 3-dimensional structure of the p16 protein.


Subject(s)
Genes, p16/genetics , Germ-Line Mutation/genetics , Melanoma/genetics , Skin Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Substitution , Binding Sites , Electrophoresis, Polyacrylamide Gel , Female , Genetic Testing , Humans , Israel/epidemiology , Male , Melanoma/ethnology , Middle Aged , Models, Molecular , Pedigree , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Sequence Analysis, DNA , Skin Neoplasms/ethnology
6.
Oncogene ; 19(14): 1849-52, 2000 Mar 30.
Article in English | MEDLINE | ID: mdl-10777219

ABSTRACT

Germline mutations in genes encoding several components of the retinoblastoma pathway have been linked with inherited predisposition to melanoma. Most commonly, such mutations involve CDKN2A, a cyclin-dependent kinase inhibitor of two kinases, CDK4 and CDK6, which phosphorylate the retinoblastoma protein (pRB) and thereby promote passage through the G1/S cell-cycle restriction point. Less frequently, germline mutations in the CDK4 gene have also been linked with an increased risk of melanoma. Despite the sequence and functional homology between CDK4 and CDK6, the role of germline mutations in CDK6 in melanoma predisposition is unknown. We detected no CDK6 mutations within the p16 (CDKN2A) binding domain in index cases from 60 melanoma-prone kindreds that lacked germline mutations in the coding regions of either CDKN2A or within the entire CDK4 coding region. We conclude that germline mutations in CDK6 do not make a significant contribution to melanoma predisposition.


Subject(s)
Germ-Line Mutation , Melanoma/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins , Amino Acid Sequence , Base Sequence , Binding Sites , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinases/genetics , Genetic Diseases, Inborn/genetics , Humans , Molecular Sequence Data
7.
Mol Cell Biol ; 20(7): 2556-68, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10713178

ABSTRACT

The phosphorylation of IkappaB by the multiprotein IkappaB kinase complex (IKC) precedes the activation of transcription factor NF-kappaB, a key regulator of the inflammatory response. Here we identified the mixed-lineage group kinase 3 (MLK3) as an activator of NF-kappaB. Expression of the wild-type form of this mitogen-activated protein kinase kinase kinase (MAPKKK) induced nuclear immigration, DNA binding, and transcriptional activity of NF-kappaB. MLK3 directly phosphorylated and thus activated IkappaB kinase alpha (IKKalpha) and IKKbeta, revealing its function as an IkappaB kinase kinase (IKKK). MLK3 cooperated with the other two IKKKs, MEKK1 and NF-kappaB-inducing kinase, in the induction of IKK activity. MLK3 bound to components of the IKC in vivo. This protein-protein interaction was dependent on the central leucine zipper region of MLK3. A kinase-deficient version of MLK3 strongly impaired NF-kappaB-dependent transcription induced by T-cell costimulation but not in response to tumor necrosis factor alpha or interleukin-1. Accordingly, endogenous MLK3 was phosphorylated and activated by T-cell costimulation but not by treatment of cells with tumor necrosis factor alpha or interleukin-1. A dominant negative version of MLK3 inhibited NF-kappaB- and CD28RE/AP-dependent transcription elicited by the Rho family GTPases Rac and Cdc42, thereby providing a novel link between these GTPases and the IKC.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , CD28 Antigens/metabolism , Cell Line , Enzyme Activation , Fluorescent Antibody Technique , Humans , I-kappa B Kinase , Leucine Zippers , NF-kappa B/metabolism , Phosphorylation , T-Lymphocytes/metabolism , Transcriptional Activation , Transfection , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinase Kinase Kinase 11
8.
Genes Chromosomes Cancer ; 27(4): 358-61, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10719365

ABSTRACT

Germline mutations of the CDKN2A tumor suppressor gene have been identified in melanoma kindreds linked to 9p21, and pancreatic adenocarcinoma is the second most common malignancy in some of these families. We hypothesized that unselected patients with both primary cancers, i.e., pancreatic cancer and malignant melanoma, have a genetic predisposition to tumor development, and that this susceptibility may be due to germline CDKN2A mutations. Fourteen patients, with both pathologically verified pancreatic adenocarcinoma and melanoma, were assessed for germline CDKN2A mutations by polymerase chain reaction amplification and sequencing of six overlapping fragments encompassing exons 1alpha and 2. A yeast two-hybrid assay was used to assess the functional consequences of CDKN2A variants. Germline CDKN2A mutations were identified in 2/14 patients: I49S, a novel substitution in exon 1alpha, and M53I, a previously reported missense mutation in exon 2. Both variants lead to compromised CDKN2A function. We conclude that the occurrence of both pancreatic cancer and melanoma, in the same patient, signals an inherited susceptibility to cancer, and that this predisposition is, in some cases, due to germline CDKN2A mutations. This finding has important implications not only for the proband, but also for other family members.


Subject(s)
Adenocarcinoma/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Germ-Line Mutation/genetics , Melanoma/genetics , Neoplasms, Multiple Primary/genetics , Pancreatic Neoplasms/genetics , Adult , Aged , DNA Mutational Analysis , DNA, Neoplasm/analysis , Female , Genetic Carrier Screening , Genetic Predisposition to Disease , Humans , Male , Middle Aged
9.
Blood ; 95(5): 1869-71, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10688850

ABSTRACT

Germline mutations of the CDKN2A (p16(INK4A)) tumor suppressor gene predispose patients to melanoma and pancreatic carcinoma. In contrast, mutations of the murine CDKN2A gene predispose BALB/c mice to pristane-induced plasmacytoma. We describe here a family in which a germline mutation of CDKN2A is present in 4 individuals who developed melanoma as well as in a fifth family member who is suffering from multiple myeloma. To determine whether the CDKN2A mutation predisposed the myeloma patient to her disease, we carried out loss of heterozygosity studies on sorted bone marrow from this individual and observed loss of the wild type CDKN2A allele in the malignant plasma cells. We suggest that germline mutations of CDKN2A may predispose individuals to a wider variety of malignancy than has been hitherto reported, but that the expression of these cancers may depend heavily on the genetic background of the patient. (Blood. 2000;95:1869-1871)


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/physiology , Genes, p16 , Germ-Line Mutation , Melanoma/genetics , Multiple Myeloma/genetics , Neoplastic Syndromes, Hereditary/genetics , Animals , Brain Neoplasms/genetics , Exons , Female , Genetic Predisposition to Disease , Humans , Loss of Heterozygosity , Male , Mice , Mice, Inbred BALB C , Pedigree , Species Specificity
10.
Gene Ther ; 6(3): 350-63, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10435085

ABSTRACT

We conducted a phase 1 trial of direct injection of an E1, E3-deleted adenovirus encoding interleukin-2 (AdCAIL-2) into subcutaneous deposits of melanoma or breast cancer. Twenty-three patients were injected at seven dose levels (10(7)-10(10) p.f.u). Local inflammation was observed at the site of injection in 60% of patients, but side-effects were otherwise minor. Incomplete local tumor regression occurred at the site of injection in 24% of patients, but no conventional clinical responses were seen. Circulating CD4 and CD8 counts fell significantly 24 h after injection. Post-injection biopsies demonstrated tumor necrosis and lymphocytic infiltration with the predominant tumor-infiltrating cells both CD3- and CD8-positive. Vector-derived sequences were detected in 14 of 18 biopsies examined 7 days after injection and vector-derived hIL-2 mRNA was detected in 80% of 7-day biopsies processed after injection of 10(8) p.f.u. of AdCAIL-2 or higher. While IL-2 was detectable by ELISA in tumor biopsies at 48 h, no protein was detectable in injected tumors after 7 days and no circulating IL-2 was detectable at any time-point. No Ad5E1 sequences were detected either before or after injection indicating absence of replication-competent virus or endogenous E1-like sequence; furthermore, only rare vector shedding was detected. Anti-adenovirus and neutralizing antibody titers were elevated 1 month after injection in all patients. This trial therefore confirms the safety of use of adenoviral vectors for gene delivery in humans and demonstrates successful transgene expression even in the face of pre-existing immunity to adenovirus.


Subject(s)
Adenoviridae/genetics , Breast Neoplasms/secondary , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Interleukin-2/genetics , Melanoma/therapy , Antibodies, Viral/blood , Breast Neoplasms/immunology , Breast Neoplasms/therapy , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Immunologic , Female , Gene Expression , Humans , Immunohistochemistry , Injections, Intralesional , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Polymerase Chain Reaction , Skin Neoplasms/immunology , Skin Neoplasms/therapy
11.
Nat Genet ; 21(1): 128-32, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9916806

ABSTRACT

Approximately 8-12% of melanoma is inherited in an autosomal dominant fashion with variable penetrance. A chromosome 9p21 locus has been linked to this disease in 50-80% of affected families. CDKN2A (also known as P16, INK4, p16INK4A and MTS1) is allelic to this locus and encodes a cdk4/cdk6 kinase inhibitor that constrains cells from progressing through the G1 restriction point. Although germline CDKN2A coding mutations cosegregate with melanoma in 25-60% of families predisposed to the disease, there remains a number of mutation-negative families that demonstrate linkage of inherited melanoma to 9p21 markers. We show here that a subset of these kindreds possess a G-->T transversion at base -34 of CDKN2A, designated G-34T. This mutation gives rise to a novel AUG translation initiation codon that decreases translation from the wild-type AUG. The G-34T mutation is not seen in controls, segregates with melanoma in families and, on the basis of haplotyping studies, probably arose from a common founder in the United Kingdom. Characterization of this and other CDKN2A non-coding mutations should have an impact on current efforts to identify susceptible melanoma-prone families and individuals.


Subject(s)
5' Untranslated Regions , Codon, Initiator , Cyclin-Dependent Kinase Inhibitor p16/genetics , Genetic Predisposition to Disease , Guanine , Melanoma/genetics , Mutation , Thymine , Alleles , Cell Line, Transformed , Female , Humans , Male , Pedigree , Protein Biosynthesis , Transcription, Genetic
12.
J Biol Chem ; 273(49): 32408-15, 1998 Dec 04.
Article in English | MEDLINE | ID: mdl-9829970

ABSTRACT

Mixed lineage kinase-3 (MLK-3) is a mitogen-activated kinase kinase kinase that mediates stress-activating protein kinase (SAPK)/c-Jun NH2-terminal kinase activation. MLK-3 and other MLK family kinases are characterized by the presence of multiple protein-protein interaction domains including a tandem leucine/isoleucine zipper (LZs) motif. Leucine zippers are known to mediate protein dimerization raising the possibility that the tandem leucine/isoleucine zippers may function as a dimerization motif of MLK-3. Using both co-immunoprecipitation and nonreducing SDS-polyacrylamide gel electrophoresis, we demonstrated that MLK-3 forms disulfide bridged homo-dimers and that the LZs motif is sufficient for MLK-3 homodimerization. We next asked whether MLK-3 utilizes a dimerization-based activation mechanism analogous to that of receptor tyrosine kinases. We found that dimerization via the LZs motif is a prerequisite for MLK-3 autophosphorylation. We then demonstrated that co-expression of Cdc42 lead to a substantial increase in MLK-3 dimerization, indicating that binding by this GTPase may induce MLK-3 dimerization. Moreover, the LZs minus form of MLK-3 failed to activate the downstream target SAPK, and expression of a MLK-3 LZs polypeptide was found to block SAPK activation by wild type MLK-3. Taken together, these findings indicate that dimerization plays a pivotal role in MLK-3 activation.


Subject(s)
Leucine Zippers , MAP Kinase Kinase Kinases , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/metabolism , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins/metabolism , DNA Primers , Dimerization , Disulfides/metabolism , JNK Mitogen-Activated Protein Kinases , Phosphorylation , Mitogen-Activated Protein Kinase Kinase Kinase 11
13.
J Cutan Med Surg ; 2(3): 172-9, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9479083

ABSTRACT

BACKGROUND: Approximately 8 to 12% of melanoma appears to be inherited in an autosomal dominant form. Although most early stage melanomas can be treated successfully by simple surgical excision, patients with advanced disease are rarely cured even with aggressive chemotherapy and/or immunotherapy. OBJECTIVE: There is now compelling evidence that germline mutations of the CDKN2A gene on chromosome 9p21 predispose to melanoma in a subset of melanoma-prone families. In this article the evidence for the role of CDKN2A in the genesis of familial melanoma is reviewed and the implications of genetic testing in families with this disease are discussed. CONCLUSION: The identification and subsequent surveillance of unaffected individuals who have a genetic predisposition to melanoma may lead to the detection of early (curable) melanomas and to a reduction in mortality.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Genes, p16 , Melanoma/genetics , Skin Neoplasms/genetics , Cell Cycle , Genetic Linkage , Humans , Karyotyping , Mutation
14.
N Engl J Med ; 338(13): 879-87, 1998 Mar 26.
Article in English | MEDLINE | ID: mdl-9516223

ABSTRACT

BACKGROUND: Germ-line mutations in the CDKN2A tumor-suppressor gene (also known as p16, p16INK4a, and MTS1) have been linked to the development of melanoma in some families with inherited melanoma. Whether mutations in CDKN2A confer a predisposition to sporadic (nonfamilial) melanoma is not known. In some patients with sporadic melanoma, one or more additional primary lesions develop, suggesting that some of these patients have an underlying genetic susceptibility to the cancer. We hypothesized that this predisposition might be due to germ-line CDKN2A mutations. METHODS: We used the polymerase chain reaction, single-strand conformation polymorphism analysis, and direct DNA sequencing to identify germ-line mutations in the CDKN2A gene in patients with multiple primary melanomas who did not have family histories of the disease. A quantitative yeast two-hybrid assay was used to evaluate the functional importance of the CDKN2A variants. RESULTS: Of 33 patients with multiple primary melanomas, 5 (15 percent; 95 percent confidence interval, 4 percent to 27 percent) had germ-line CDKN2A mutations. These included a 24-bp insertion at the 5' end of the coding sequence, three missense mutations (Arg24Pro, Met53Ile, and Ser56Ile), and a 2-bp deletion at position 307 to 308 (resulting in a truncated CDKN2A protein). In three families, CDKN2A mutations identical to those in the probands were found in other family members. In two families with mutations, we uncovered previously unknown evidence of family histories of melanoma. CONCLUSIONS: Some patients with multiple primary melanomas but without family histories of the disease have germ-line mutations of the CDKN2A gene. The presence of multiple primary melanomas may signal a genetic susceptibility to melanoma not only in the index patient but also in family members, who may benefit from melanoma-surveillance programs.


Subject(s)
Genes, p16/genetics , Germ-Line Mutation , Melanoma/genetics , Neoplasms, Multiple Primary/genetics , Skin Neoplasms/genetics , Adult , Aged , DNA Mutational Analysis/methods , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Pedigree
15.
Int J Cancer ; 73(4): 531-6, 1997 Nov 14.
Article in English | MEDLINE | ID: mdl-9389568

ABSTRACT

The CDKN2A gene encodes p16 (CDKN2A), a cell-cycle inhibitor protein which prevents inappropriate cell cycling and, hence, proliferation. Germ-line mutations in CDKN2A predispose to the familial atypical multiple-mole melanoma (FAMMM) syndrome but also have been seen in rare families in which only 1 or 2 individuals are affected by cutaneous malignant melanoma (CMM). We therefore sequenced exons 1alpha and 2 of CDKN2A using lymphocyte DNA isolated from index cases from 67 families with cancers at multiple sites, where the patterns of cancer did not resemble those attributable to known genes such as hMLH1, hMLH2, BRCA1, BRCA2, TP53 or other cancer susceptibility genes. We found one mutation, a mis-sense mutation resulting in a methionine to isoleucine change at codon 53 (M531) of exon 2. The individual tested had developed 2 CMMs but had no dysplastic nevi and lacked a family history of dysplastic nevi or CMM. Other family members had been diagnosed with oral cancer (2 persons), bladder cancer (1 person) and possibly gall-bladder cancer. While this mutation has been reported in Australian and North American melanoma kindreds, we did not observe it in 618 chromosomes from Scottish and Canadian controls. Functional studies revealed that the CDKN2A variant carrying the M531 change was unable to bind effectively to CDK4, showing that this mutation is of pathological significance. Our results have confirmed that CDKN2A mutations are not limited to FAMMM kindreds but also demonstrate that multi-site cancer families without melanoma are very unlikely to contain CDKN2A mutations.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Genes, Tumor Suppressor/genetics , Germ-Line Mutation/genetics , Neoplasms, Multiple Primary/genetics , Adolescent , Adult , Aged , Female , Humans , Male , Melanoma/genetics , Middle Aged , Pedigree , Syndrome
18.
Hum Genet ; 99(6): 776-80, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9187672

ABSTRACT

MLK-3 kinase is a widely expressed serine/ threonine kinase that bears multiple protein interaction domains and regulates signals mediated by the stress-responsive pathway. Thus, MLK-3 signaling affects numerous cellular processes, raising the possibility that MLK-3 might play a role in oncogenesis. In this report, we describe the fine mapping of the MLK-3 gene within the 11q13.1 chromosomal region. By integrating data from somatic cell hybrids and double color fluorescence in situ hybridization on metaphase chromosomes and DNA fibers. MLK-3 has been assigned approximately 1 Mb telomeric of PYGM, close to the D11S546 locus. Since the MEN1 susceptibility locus is also located within the 11q13.1 region, we have carried out Southern and Northern blot analyses, as well as protein truncation assays to establish whether abnormalities in MLK-3 lead to the development of this familial cancer syndrome. Our observations exclude MLK-3 as the MEN1 gene.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 11/genetics , MAP Kinase Kinase Kinases , Multiple Endocrine Neoplasia Type 1/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Cricetinae , Cricetulus , Genetic Predisposition to Disease , Humans , Hybrid Cells/chemistry , Hybrid Cells/metabolism , In Situ Hybridization, Fluorescence , Mitogen-Activated Protein Kinase Kinase Kinase 11
20.
Clin Cancer Res ; 3(6): 977-81, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9815774

ABSTRACT

Gliomas are tumors of the central nervous system that may be inherited in some patients. The gene(s) responsible for the clustering of gliomas in families have not yet been identified. Molecular studies of sporadic high-grade gliomas have revealed mutations or deletions of the genes encoding the protein kinase inhibitors p16(INK4A) and p15(INK4B) in a large proportion of tumors. Moreover, those tumors without deletions frequently display gene amplification and/or over-expression of mRNA encoding the protein kinase cdk4. We hypothesized that germ-line mutations in the p16(INK4A), p15(INK4B), or CDK4 genes might contribute to some cases of familial gliomas. To address this issue, we analyzed 36 kindreds with a predisposition to glial tumors. Genomic DNA from index members of these families was screened by PCR-single-strand conformational polymorphism analysis. We did not detect any functional mutations in the p16(INK4A), p15(INK4B), or CDK4 genes, although two individuals did have a previously described A140T polymorphism in p16(INK4A). Thus, despite the association between the sporadic forms of high-grade glioma and abnormalities of p16(INK4A), p15(INK4B), or CDK4, we found no evidence that germ-line mutations in the coding region of these three genes predispose to inherited glial tumors.


Subject(s)
Brain Neoplasms/genetics , Carrier Proteins/genetics , Cell Cycle Proteins , Cyclin-Dependent Kinases/genetics , Germ-Line Mutation , Glioma/genetics , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Adult , Aged , Base Sequence , Brain Neoplasms/epidemiology , Cluster Analysis , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16 , DNA Primers , Exons , Family , Female , Gene Amplification , Genes, Tumor Suppressor , Genetic Predisposition to Disease , Glioma/epidemiology , Humans , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...