86 results on '"Huebner K"'
Search Results
2. Activation of MYC in a Masked T(8;17) Translocation Results in an Aggressive B-Cell Leukemia
- Author
-
Gauwerky, C. E., Huebner, K., Isobe, M., Nowell, P. C., and Croce, C. M.
- Published
- 1989
3. Cloning of Three Human Tyrosine Phosphatases Reveals a Multigene Family of Receptor-Linked Protein-Tyrosine-Phosphatases Expressed in Brain
- Author
-
Kaplan, R., Morse, B., Huebner, K., Croce, C., Howk, R., Ravera, M., Ricca, G., Jaye, M., and Schlessinger, J.
- Published
- 1990
4. Receptor Protein-Tyrosine Phosphatase γ is a Candidate Tumor Suppressor Gene at Human Chromosome Region 3p21
- Author
-
LaForgia, S., Morse, B., Levy, J., Barnea, G., Cannizzaro, L. A., Li, F., Nowell, P. C., Boghosian-Sell, L., Glick, J., Weston, A., Harris, C. C., Drabkin, H., Patterson, D., Croce, C. M., Schlessinger, J., and Huebner, K.
- Published
- 1991
5. DNA-transformed murine teratocarcinoma cells: regulation of expression of simian virus 40 tumor antigen in stem versus differentiated cells.
- Author
-
Linnenbach, A, Huebner, K, and Croce, C M
- Abstract
Thymidine kinase-deficient (TK-; ATP:thymidine 5'-phosphotransferase, EC 2.7.1.21)F9 teratocarcinoma stem cells have been transformed with a recombinant plasmid genome consisting of the pBR322 genome linked to a herpes simplex virus type 1 thymidine kinase gene (HSV-1 tk) and a simian virus 40 (SV40) genome. A clonal line of stem cells was obtained that contains only one copy of plasmid DNA, which is integrated into murine chromosomal DNA through a site on the pBRR322 genome. The HSV-1 tk gene, which is adjacent to the SV40 genome, is expressed in stem cells, whereas SV40 gene expression is not detectable. If differentiation of these stem cells is induced, the differentiated cells express SV40 early gene products. Thus, we have constructed a stem cell which contains a set of genes (SV40), the expression of which is regulated differently in stem and differentiated cells. This cell line could be used to determine the mechanism of suppression of expression of these genes in stem cells.
- Published
- 1980
- Full Text
- View/download PDF
6. Transcription of the simian virus 40 genome in DNA-transformed murine teratocarcinoma stem cells.
- Author
-
Linnenbach, A, Huebner, K, and Croce, C M
- Abstract
To study the molecular basis for lack of expression of the simian virus 40 (SV40) early region genes in murine teratocarcinoma-derived stem cells, we introduced a recombinant plasmid consisting of pBR322 linked to the herpes simplex virus type 1 thymidine kinase gene and SV40 genome into thymidine kinase-deficient F9 stem cells. The resulting stem cell clone, 12-1, and a retinoic acid-induced differentiated daughter cell clone, 12-1a, each contain one copy per cell of the entire recombinant plasmid integrated into the cellular genome through a site on the pBR322 genome. Restriction endonuclease analyses indicate that there is no difference in integration site or organization of the three component parts of the plasmid genome within cellular DNA of stem and differentiated cells; yet the differentiated cells, 12-1a, express SV40 large tumor antigen whereas the stem cells, 12-1, do not. Both stem and differentiated cells produce two size classes of polyadenylylated RNA, 2900 and 2600 bases in length, homologous to the early region of the SV40 genome, detectable by RNA blotting analysis. S1 nuclease analysis of the SV40 transcripts present in stem and differentiated cells indicate that the SV40 mRNAs were identically spliced in the two cell types, in a manner consistent with that observed for spliced large and small tumor antigen mRNAs in SV40-infected monkey kidney cells. Thus, the failure of 12-1 teratocarcinoma stem cells, containing an integrated SV40 genome, to express SV40 tumor antigen is not due to a lack of transcription of the SV40 early region or to an inability to splice primary transcripts.
- Published
- 1981
- Full Text
- View/download PDF
7. Papillomavirus sequences integrate near cellular oncogenes in some cervical carcinomas.
- Author
-
Dürst, M, Croce, C M, Gissmann, L, Schwarz, E, and Huebner, K
- Abstract
The chromosomal locations of cellular sequences flanking integrated papillomavirus DNA in four cervical carcinoma cell lines and a primary cervical carcinoma have been determined. The two human papillomavirus (HPV) 16 flanking sequences derived from the tumor were localized to chromosome regions 20pter----20q13 and 3p25----3qter, regions that also contain the protooncogenes c-src-1 and c-raf-1, respectively. The HPV 16 integration site in the SiHa cervical carcinoma-derived cell line is in chromosome region 13q14----13q32. The HPV 18 integration site in SW756 cervical carcinoma cells is in chromosome 12 but is not closely linked to the Ki-ras2 gene. Finally, in two cervical carcinoma cell lines, HeLa and C4-I, HPV 18 DNA is integrated in chromosome 8, 5' of the c-myc gene. The HeLa HPV 18 integration site is within 40 kilobases 5' of the c-myc gene, inside the HL60 amplification unit surrounding and including the c-myc gene. Additionally, steady-state levels of c-myc mRNA are elevated in HeLa and C4-I cells relative to other cervical carcinoma cell lines. Thus, in at least some genital tumors, cis-activation of cellular oncogenes by HPV may be involved in malignant transformation of cervical cells.
- Published
- 1987
- Full Text
- View/download PDF
8. Mapping of four distinct BCR-related loci to chromosome region 22q11: order of BCR loci relative to chronic myelogenous leukemia and acute lymphoblastic leukemia breakpoints.
- Author
-
Croce, C M, Huebner, K, Isobe, M, Fainstain, E, Lifshitz, B, Shtivelman, E, and Canaani, E
- Abstract
A probe derived from the 3' region of the BCR gene (breakpoint cluster region gene) detects four distinct loci in the human genome. One of the loci corresponds to the complete BCR gene, whereas the others contain a 3' segment of the gene. After HindIII cleavage of human DNA, these four loci are detected as 23-, 19-, 13-, and 9-kilobase-pair fragments, designated BCR4, BCR3, BCR2, and BCR1, respectively, with BCR1 deriving from the original complete BCR gene. All four BCR loci segregate 100% concordantly with human chromosome 22 in a rodent-human somatic cell hybrid panel and are located at chromosome region 22q11.2 by chromosomal in situ hybridization. The BCR2 and BCR4 loci are amplified in leukemia cell line K562 cells, indicating that they fall within the amplification unit that includes immunoglobulin lambda light chain locus (IGL) and ABL locus on the K562 Philadelphia chromosome (Ph1); additionally, in chronic myelogenous leukemia-derived mouse-human hybrids retaining a Ph1 chromosome in the absence of the 9q+ and normal chromosome 22, BCR2 and BCR4 loci are retained, whereas the 3' region of BCR1 and the BCR3 locus are lost, indicating that BCR3 is distal to BCR1 on chromosome 22. Similarly, in mouse-human hybrids retaining a Ph1 chromosome derived from an acute lymphoblastic leukemia-in the absence of the 9q+ and 22, only BCR2 and BCR4 loci are retained, indicating that the breakpoint in this acute lymphoblastic leukemia, as in chronic myelogenous leukemia, is proximal to the BCR1 3' region, but distal to the IGLC locus and the BCR2 and BCR4 3' loci. Thus, the order of loci on chromosome 22 is centromere----BCR2, BCR4, and IGL----BCR1----BCR3----SIS, possibly eliminating BCR2 and BCR4 loci as candidate targets for juxtaposition to the ABL gene in the acute lymphoblastic leukemia Ph1 chromosome.
- Published
- 1987
- Full Text
- View/download PDF
9. The alpha-spectrin gene is on chromosome 1 in mouse and man.
- Author
-
Huebner, K, Palumbo, A P, Isobe, M, Kozak, C A, Monaco, S, Rovera, G, Croce, C M, and Curtis, P J
- Abstract
By using alpha-spectrin cDNA clones of murine and human origin and somatic cell hybrids segregating either mouse or human chromosomes, the gene for alpha-spectrin has been mapped to chromosome 1 in both species. This assignment of the mouse alpha-spectrin gene to mouse chromosome 1 by DNA hybridization strengthens the previous identification of the alpha-spectrin locus in mouse with the sph locus, which previously was mapped by linkage analysis to mouse chromosome 1, distal to the Pep-3 locus. By in situ hybridization to human metaphase chromosomes, the human alpha-spectrin gene has been localized to 1q22-1q25; interestingly, the locus for a non-Rh-linked form of elliptocytosis has been provisionally mapped to band 1q2 by family linkage studies.
- Published
- 1985
- Full Text
- View/download PDF
10. Characterization of a cDNA clone encoding human filaggrin and localization of the gene to chromosome region 1q21.
- Author
-
McKinley-Grant, L J, Idler, W W, Bernstein, I A, Parry, D A, Cannizzaro, L, Croce, C M, Huebner, K, Lessin, S R, and Steinert, P M
- Abstract
Filaggrins are an important class of intermediate filament-associated proteins that interact with keratin intermediate filaments of terminally differentiating mammalian epidermis. They show wide species variations and their aberrant expression has been implicated in a number of keratinizing disorders. We have isolated a cDNA clone encoding human filaggrin and used this to demonstrate that the human gene encodes a polyprotein precursor containing numerous tandem filaggrin repeats. This structure is similar to that of mouse; however, the human filaggrin repeat is much longer (972 base pairs; 324 amino acids) and shows little sequence homology to the mouse protein. Also, data presented here reveal that the human filaggrin repeats show considerable sequence variations; such polymorphism is not found in the mouse. Furthermore, chromosomal mapping data revealed that the human gene is located at 1q21, indicating that the polymorphism is confined to a single locus. By peptide mapping, we define a short linker sequence within the human filaggrin repeat that is excised by proteolysis to yield functional molecules. Finally, we show by in situ hybridization that human filaggrin precursor gene expression is tightly regulated at the transcriptional level in terminally differentiating epidermis and that this represents a useful system in which to study intermediate filament-intermediate filament-associated protein interactions as well as disorders of keratinization.
- Published
- 1989
- Full Text
- View/download PDF
11. Human DNA topoisomerase I is encoded by a single-copy gene that maps to chromosome region 20q12-13.2.
- Author
-
Juan, C C, Hwang, J L, Liu, A A, Whang-Peng, J, Knutsen, T, Huebner, K, Croce, C M, Zhang, H, Wang, J C, and Liu, L F
- Abstract
cDNA clones of the human TOP1 gene encoding DNA topoisomerase I (EC 5.99.1.2) have been obtained by immunochemical screening of phage lambda libraries expressing human cDNA segments, using rabbit antibodies raised against purified HeLa DNA topoisomerase I. Hybridization patterns between the cloned cDNA sequences and human cellular DNA and cytoplasmic mRNAs indicate that human TOP1 is a single-copy gene. The chromosomal location of the gene has been mapped to the long arm of chromosome 20, in the region q12-13.2, by hybridization of a radioactively labeled TOP1 cDNA probe to human metaphase chromosomes and to a panel of rodent-human somatic hybrids retaining overlapping subsets of human chromosomes.
- Published
- 1988
- Full Text
- View/download PDF
12. Deoxyribonuclease I sensitivity of plasmid genomes in teratocarcinoma-derived stem and differentiated cells.
- Author
-
Huebner, K, Linnenbach, A, Weidner, S, Glenn, G, and Croce, C M
- Abstract
The DNase I (EC 3.1.21.1) sensitivities of the simian virus 40 (SV40) genome, the pBR322 genome, and the herpes simplex virus type 1 thymidine kinase (HSV-1 tk) gene have been compared in teratocarcinoma-derived stem (12-1) and differentiated (12-1a) cell lines established by transfection of thymidine kinase (ATP:thymidine 5'-phosphotransferase, EC 2.7.1.21)-deficient F9 cells with DNA from a tripartite plasmid genome consisting of the pBR322 genome, the SV40 genome, and the HSV-1 tk gene. HSV-1 tk is present in both stem and differentiated cells; SV40 early proteins are present in differentiated cells but not in stem cells; the pBR322 genome is not expressed in either cell type. The SV40 and pBR322 genomes are more sensitive to DNase I digestion in stem cells than in differentiated cells, reflecting the DNase I-hypersensitivity of total stem-cell chromatin. The HSV-1 tk gene is the least sensitive to DNase I digestion in both cell types.
- Published
- 1981
- Full Text
- View/download PDF
13. Actively transcribed genes in the raf oncogene group, located on the X chromosome in mouse and human.
- Author
-
Huebner, K, ar-Rushdi, A, Griffin, C A, Isobe, M, Kozak, C, Emanuel, B S, Nagarajan, L, Cleveland, J L, Bonner, T I, and Goldsborough, M D
- Abstract
Murine and human cDNAs, related to but distinct from c-raf-1, have been isolated and designated mA-raf and hA-raf, respectively. The mA-raf and hA-raf cDNAs detect the same murine and human fragments in Southern blots of restriction enzyme-cleaved murine and human cellular DNA. The murine restriction enzyme fragments homologous to mA-raf cDNA cosegregate with mouse chromosome X in a panel of Chinese hamster-mouse hybrid cells, thus localizing the mA-raf locus to mouse chromosome X. Two independently segregating loci, detected by the hA-raf cDNA (or mA-raf cDNA), hA-raf-1 and hA-raf-2, are located on human chromosomes X and 7, respectively. The mA-raf locus and the hA-raf-1 locus are actively transcribed in several mouse and human cell lines.
- Published
- 1986
- Full Text
- View/download PDF
14. Molecular analysis of a t(14;14) translocation in leukemic T-cells of an ataxia telangiectasia patient.
- Author
-
Russo, G, Isobe, M, Gatti, R, Finan, J, Batuman, O, Huebner, K, Nowell, P C, and Croce, C M
- Abstract
We have detected and cloned two rearrangements in the T-cell receptor alpha locus from a clone of somatic cell hybrids carrying a t(14;14)(q11;q32) chromosomal translocation derived from an ataxia telangiectasia patient with T-cell chronic lymphocytic leukemia. The T-cell clone carrying the t(14;14) chromosomal translocation was known to be present for greater than 10 years before the onset of overt leukemia. One molecular rearrangement of the T-cell receptor alpha locus corresponded to a functional variable-joining region (V-J) joining, whereas the other derived from the breakpoint of the t(14;14)(q11;q32) translocation. Chromosomal in situ hybridization of the probe derived from the t(14;14) breakpoint localized the breakpoint region to 14q32.1, apparently the same region that is involved in another ataxia telangiectasia characteristic chromosome translocation, t(7;14)(q35;q32). The 14q32.1 breakpoint is at least 10,000 kilobase pairs (kbp) centromeric to the immunoglobulin heavy chain locus. Sequence analysis of the breakpoint indicates the involvement of a J alpha sequence during the translocation. Comigration of high-molecular weight DNA fragments involved with t(7;14) and t(14;14) translocations suggests the presence of a cluster of breakpoints in the 14q32.1 region, the site of a putative oncogene, TCL1.
- Published
- 1989
- Full Text
- View/download PDF
15. Chromosomal locations of mouse immunoglobulin genes.
- Author
-
Valbuena, O, Marcu, K B, Croce, C M, Huebner, K, Weigert, M, and Perry, R P
- Abstract
The chromosomal locations of the structural genes coding for the constant portions of mouse heavy (H) and light chain immunoglobulins were studied by molecular hybridization techniques. Complementary DNA probes containing the constant-region sequences of kappa and lambdaI light chain and alpha, gamma2b, and mu heavy chain mRNAs were annealed to a large excess of DNA from a series of eight mouse-human hybrid cell lines that are deficient for various mouse chromosomes. The lines were scored as positive when a high proportion of a probe annealed and negative when an insignificant proportion annealed. Some lines were clearly negative for H and lambda and clearly positive for kappa. Others were positive or intermediate for lambda, positive for kappa and negative for H. Still others, including a line that was selected for the absence of the mouse X chromosome, were positive for all immunoglobulin species. These results demonstrate that the Clambda, Ckappa, and CH genes are located on different autosomes in the mouse. In contrast, the three heavy-chain families exhibited consistently uniform hybridization results, suggesting that the genes for Calpha, Cgamma, and Cmu are located on the same chromosome. A comparison of karyotypic data with hybridization data has limited the possible locations of the Ig genes to only a few chromosomes.
- Published
- 1978
- Full Text
- View/download PDF
16. The gene encoding the T-cell surface protein T4 is located on human chromosome 12.
- Author
-
Isobe, M, Huebner, K, Maddon, P J, Littman, D R, Axel, R, and Croce, C M
- Abstract
The surface glycoproteins T4 and T8 define functionally distinct populations of T lymphocytes. We have obtained cDNA and genomic clones encoding the T4 molecule and used these as probes to determine the chromosomal location of this gene. Genomic blotting experiments, along with in situ hybridization analyses, indicate that the T4 gene resides on the short arm of human chromosome 12, at region p12-pter. Thus, the T4 gene is not linked to any known member of the immunoglobulin gene family, including its counterpart gene, T8, which resides on human chromosome 2 immediately distal to the immunoglobulin kappa locus.
- Published
- 1986
- Full Text
- View/download PDF
17. Chromosome localization of the gene for human terminal deoxynucleotidyltransferase to region 10q23-q25.
- Author
-
Isobe, M, Huebner, K, Erikson, J, Peterson, R C, Bollum, F J, Chang, L M, and Croce, C M
- Abstract
Complementary DNA clones representing the 3' half, the 5' half, and the entire coding region of the human terminal deoxynucleotidyltransferase gene (TdT; DNA nucleotidylexotransferase, nucleosidetriphosphate: DNA deoxynucleotidylexotransferase, EC 2.7.7.31) were used to screen a panel of mouse X human somatic cell hybrid DNAs to determine the chromosomal location of the human TdT gene. The results of the Southern transfer analysis of hybrid DNAs indicate that the gene for TdT is located on human chromosome 10. The in situ hybridization technique was then used to further localize the gene for TdT to region q23-q25 of human chromosome 10.
- Published
- 1985
- Full Text
- View/download PDF
18. The nerve growth factor receptor gene is at human chromosome region 17q12-17q22, distal to the chromosome 17 breakpoint in acute leukemias.
- Author
-
Huebner, K, Isobe, M, Chao, M, Bothwell, M, Ross, A H, Finan, J, Hoxie, J A, Sehgal, A, Buck, C R, and Lanahan, A
- Abstract
Genomic and cDNA clones for the human nerve growth factor receptor have been used in conjunction with somatic cell hybrid analysis and in situ hybridization to localize the nerve growth factor receptor locus to human chromosome region 17q12-q22. Additionally, part, if not all, of the nerve growth factor receptor locus is present on the translocated portion of 17q (17q21-qter) from a poorly differentiated acute leukemia in which the chromosome 17 breakpoint was indistinguishable cytogenetically from the 17 breakpoint observed in the t(15;17)(q22;q21) translocation associated with acute promyelocytic leukemia. Thus the nerve growth factor receptor locus may be closely distal to the acute promyelocytic leukemia-associated chromosome 17 breakpoint at 17q21.
- Published
- 1986
- Full Text
- View/download PDF
19. Localization of the human JUN protooncogene to chromosome region 1p31-32.
- Author
-
Haluska, F G, Huebner, K, Isobe, M, Nishimura, T, Croce, C M, and Vogt, P K
- Abstract
The oncogene jun is the putative transforming gene of avian sarcoma virus 17; jun appears to be derived from a gene of the chicken genome and has homologues in several other vertebrate species. Recent genetic and immunological data indicate that jun codes for a protein that is closely related and probably identical to the transcription factor AP-1. We have isolated a genomic DNA clone encompassing the human cellular counterpart of the gene, JUN, and used this DNA to determine the chromosomal location of the gene. A panel of DNA preparations derived from rodent-human somatic cell hybrids with defined chromosome complements was first screened with the JUN probe. This Southern blot analysis indicated that JUN is situated on the short arm of chromosome 1. In situ hybridization then assigned JUN to chromosome region 1p31-32, a chromosomal region involved in both translocations and deletions of chromosomes seen in human malignancies.
- Published
- 1988
- Full Text
- View/download PDF
20. The human c-ros gene (ROS) is located at chromosome region 6q16----6q22.
- Author
-
Nagarajan, L, Louie, E, Tsujimoto, Y, Balduzzi, P C, Huebner, K, and Croce, C M
- Abstract
The human homolog, c-ros, of the transforming gene, v-ros, of the avian sarcoma virus, UR2, has been isolated from a human genomic library. A single-copy fragment from the human c-ros genomic clone has been used to map the human c-ros homolog (ROS) to human chromosome region 6q16----6q22 by somatic cell hybrid analysis and chromosomal in situ hybridization. Thus, the c-ros gene joins the c-myb oncogene, which is distal to the c-ros gene on the long arm of human chromosome 6, as a candidate for involvement in chromosome 6q deletions and rearrangements seen in various malignancies.
- Published
- 1986
- Full Text
- View/download PDF
21. The translocated c-myc oncogene of Raji Burkitt lymphoma cells is not expressed in human lymphoblastoid cells.
- Author
-
Nishikura, K, Erikson, J, ar-Rushdi, A, Huebner, K, and Croce, C M
- Abstract
We hybridized Raji Burkitt lymphoma cells, which carry a t(8;14) chromosome translocation, with human lymphoblastoid cells to study the expression of the translocated cellular myc oncogene (c-myc) in the hybrid cells. In Raji cells the c-myc oncogene is translocated to a switch region of the gamma heavy chain locus (S gamma). Because of sequence alterations in the 5' exon of the translocated c-myc oncogene in this cell line, it is possible to distinguish the transcripts of the translocated c-myc gene and of the normal c-myc gene. S1 nuclease protection experiments with a c-myc first exon probe indicate that Raji cells express predominantly the translocated c-myc gene, while the level of expression of the normal c-myc gene is less than 2% of that of the translocated c-myc gene. Somatic cell hybrids between Raji and human lymphoblastoid cells retain the lymphoblastoid phenotype and express only the normal c-myc oncogene. This result indicates that the activation of a c-myc oncogene translocated to a S region depends on the stage of B-cell differentiation of the cells harboring the translocated c-myc gene and not on alterations in the structure of the translocated c-myc oncogene.
- Published
- 1985
- Full Text
- View/download PDF
22. Involvement of the ALL-1 gene in a solid tumor.
- Author
-
Baffa, R, Negrini, M, Schichman, S A, Huebner, K, and Croce, C M
- Abstract
Translocations involving chromosome band 11q23, found in 5-10% of human acute leukemias, disrupt the ALL-1 gene. This gene is fused by reciprocal translocation with a variety of other genes in acute lymphoblastic and myelogenous leukemias, and it undergoes self-fusion in acute myeloid leukemias with normal karyotype or trisomy 11. Here we report an alteration of the ALL-1 gene in a gastric carcinoma cell line (Mgc80-3). Characterization of this rearrangement revealed a three-way complex translocation, involving chromosomes 1 and 11, resulting in a partial duplication of the ALL-1 gene. Sequencing of reverse transcription-PCR products and Northern blot analysis showed that only the partially duplicated ALL-1 gene was transcribed, producing an mRNA with exon 8 fused to exon 2. This report of ALL-1 gene rearrangement in a solid tumor suggests that ALL-1 plays a role in the pathogenesis of some solid malignancies. The absence of the normal transcript in this cell line, in association with the loss-of-heterozygosity studies on chromosome 11q23 seen in solid tumors, suggests that ALL-1 is involved in tumorigenesis by a loss-of-function mechanism.
- Published
- 1995
- Full Text
- View/download PDF
23. Localization of the human pim oncogene (PIM) to a region of chromosome 6 involved in translocations in acute leukemias.
- Author
-
Nagarajan, L, Louie, E, Tsujimoto, Y, ar-Rushdi, A, Huebner, K, and Croce, C M
- Abstract
The human homolog, hpim, of the murine pim-1 gene, which is activated in murine T-cell lymphomas by insertion of retrovirus proviral genomes in the pim-1 region, has been molecularly cloned; the cloned probe has been used to map the hpim locus to human chromosome region 6p21 by somatic cell hybrid analysis and chromosomal in situ hybridization. The hpim gene is expressed as a 3.2-kilobase mRNA in various human cell lines of hematopoietic lineage, most dramatically in the K562 erythroleukemia cell line, which contains a cytogenetically demonstrable rearrangement in the 6p21 region. A characteristic chromosome anomaly, a reciprocal translocation t(6;9)(p21;q33), has been described in myeloid leukemias and could involve the hpim gene.
- Published
- 1986
- Full Text
- View/download PDF
24. Cloning and sequencing of cDNA encoding human DNA topoisomerase II and localization of the gene to chromosome region 17q21-22.
- Author
-
Tsai-Pflugfelder, M, Liu, L F, Liu, A A, Tewey, K M, Whang-Peng, J, Knutsen, T, Huebner, K, Croce, C M, and Wang, J C
- Abstract
Two overlapping cDNA clones encoding human DNA topoisomerase II were identified by two independent methods. In one, a human cDNA library in phage lambda was screened by hybridization with a mixed oligonucleotide probe encoding a stretch of seven amino acids found in yeast and Drosophila DNA topoisomerase II; in the other, a different human cDNA library in a lambda gt11 expression vector was screened for the expression of antigenic determinants that are recognized by rabbit antibodies specific to human DNA topoisomerase II. The entire coding sequences of the human DNA topoisomerase II gene were determined from these and several additional clones, identified through the use of the cloned human TOP2 gene sequences as probes. Hybridization between the cloned sequences and mRNA and genomic DNA indicates that the human enzyme is encoded by a single-copy gene. The location of the gene was mapped to chromosome 17q21-22 by in situ hybridization of a cloned fragment to metaphase chromosomes and by hybridization analysis with a panel of mouse-human hybrid cell lines, each retaining a subset of human chromosomes.
- Published
- 1988
- Full Text
- View/download PDF
25. Cloning and characterization of two members of the vertebrate Dlx gene family.
- Author
-
Simeone, A, Acampora, D, Pannese, M, D'Esposito, M, Stornaiuolo, A, Gulisano, M, Mallamaci, A, Kastury, K, Druck, T, and Huebner, K
- Abstract
A number of vertebrate genes of the Dlx gene family have been cloned in mouse, frog, and zebrafish. These genes contain a homeobox related to that of Distalless, a gene expressed in the developing head and limbs of Drosophila embryos. We cloned and studied the expression of two members of this family, which we named Dlx5 and Dlx6, in human and mouse. The two human genes, DLX5 and DLX6, are closely linked in an inverted convergent configuration in a region of chromosome 7, at 7q22. Similarly, the two human genes DLX1 and DLX2 are closely linked in a convergent configuration at 2q32, near the HOXD (previously HOX4) locus. In situ hybridization experiments in mouse embryos revealed expression of Dlx5 and Dlx6 mRNA in restricted regions of ventral diencephalon and basal telencephalon, with a distribution very similar to that reported for Dlx1 and Dlx2 mRNA. A surprising feature of Dlx5 and Dlx6 is that they are also expressed in all skeletal structures of midgestation embryos after the first cartilage formation. The expression pattern of these genes, together with their chromosome localization, may provide useful cues for the study of congenital disorders in which there is a combination of craniofacial and limb defects.
- Published
- 1994
- Full Text
- View/download PDF
26. Control of expression of histocompatibility antigens (H-2) and beta 2-microglobulin in F9 teratocarcinoma stem cells.
- Author
-
Croce, C M, Linnenbach, A, Huebner, K, Parnes, J R, Margulies, D H, Appella, E, and Seidman, J G
- Abstract
Murine teratocarcinoma stem cells, unlike most other cell types, do not express major histocompatibility antigens. The steady-state levels of beta 2-microglobulin and H-2 mRNA from F9-derived teratocarcinoma stem and differentiated cells were examined by blot hybridization using cloned DNA probes specific for these mRNAs. No H-2- or beta 2-microglobulin-specific RNA was detected in F9 teratocarcinoma stem cells (clone 12-1); thus, F9 teratocarcinoma stem cells (clone 12-1) contain no more than 1/10 the H-2 and beta 2-microglobulin mRNAs of the differentiated daughter cells (clone 12-1a). We suggest that this regulation of major histocompatibility antigen expression is due to transcriptional control of the major histocompatibility antigen genes, H-2 and beta 2-microglobulin. The transcriptional regulation of these genes is accompanied by a change in their DNase I sensitivity. Normally, transcriptionally inactive genes are DNase I resistant, while active genes are DNase I sensitive. In contrast, the silent major histocompatibility antigen genes of teratocarcinoma stem cells are more DNase I sensitive than the active genes of the differentiated cells.
- Published
- 1981
- Full Text
- View/download PDF
27. Structural alteration in the MYB protooncogene and deletion within the gene encoding alpha-type protein kinase C in human melanoma cell lines.
- Author
-
Linnenbach, A J, Huebner, K, Reddy, E P, Herlyn, M, Parmiter, A H, Nowell, P C, and Koprowski, H
- Abstract
A correlative study was done to determine possible relationships between nonrandom aberrations in chromosomes 1, 6, and 7 occurring in human cutaneous malignant melanoma and the structure of oncogenes as well as specific genes encoding growth factors and growth factor receptors. Thirty cell lines derived from primary or metastatic melanomas of 28 patients were analyzed by Southern blotting with nick-translated probes for 28 different genes, some of which map near frequent chromosomal breakpoints observed in melanoma. An alteration in the MYB protooncogene was observed in a cell line derived from a primary melanoma in the vertical growth phase, which correlated with a 6q22 chromosomal abnormality. Another primary melanoma cell line had a cytogenetically undetected tumor-specific deletion within the gene for alpha-type protein kinase C. Polymorphic alleles for the genes encoding the epidermal growth factor receptor and alpha-type protein kinase C were also observed.
- Published
- 1988
- Full Text
- View/download PDF
28. Chromosomal translocation in T-cell leukemia line HUT 78 results in a MYC fusion transcript.
- Author
-
Finger, L R, Huebner, K, Cannizzaro, L A, McLeod, K, Nowell, P C, and Croce, C M
- Abstract
Primary cultures and established cell lines derived from human T-cell leukemias were analyzed for genomic rearrangements in the region 3' of the MYC locus. A T-cell leukemia line, HUT 78, whose 3' MYC region is rearranged, carries a chromosome t(2;8) juxtaposition; i.e., a locus derived from chromosome region 2q34 is attached to the 3' end of one MYC allele. The t(2;8) rearrangement in the HUT 78 cell line results in expression of a fused transcript encompassing the MYC gene and a locus designated TCL4 (T-cell leukemia/lymphoma 4), which normally resides on chromosome 2. The steady-state level of MYC-TCL4 fusion transcripts in HUT 78 cells is significantly higher than the MYC RNA level found in several other B- and T-cell lines. The production of fused MYC-TCL4 transcripts in a leukemic cell line raises the possibility that other B- and T-cell leukemias may express MYC fusion transcripts as an integral step in their pathogenesis.
- Published
- 1988
- Full Text
- View/download PDF
29. The human homologue of the retroviral oncogene qin maps to chromosome 14q13.
- Author
-
Kastury, K, Li, J, Druck, T, Su, H, Vogt, P K, Croce, C M, and Huebner, K
- Abstract
Chromosomal mapping of the human QIN gene (renamed FKH2 by the Human Genome Organization Nomenclature Committee) was initially accomplished by correlation of the presence of the QIN locus with specific chromosome regions in a rodent-human hybrid panel. This analysis revealed that the human QIN gene maps to chromosome region 14q11.2-->14q32, between the TCR and IGH loci. Further analysis by fluorescence in situ hybridization techniques with a human QIN genomic clone refined the human QIN gene localization to 14q13.
- Published
- 1994
- Full Text
- View/download PDF
30. Zinc treatment reverses and anti-Zn-regulated miRs suppress esophageal carcinomas in vivo.
- Author
-
Fong LY, Huebner K, Jing R, Smalley KJ, Brydges CR, Fiehn O, Farber JL, and Croce CM
- Subjects
- Humans, Rats, Animals, Antagomirs, Zinc metabolism, Apoptosis Regulatory Proteins metabolism, Inflammation complications, Cell Line, Tumor, Cell Proliferation, Gene Expression Regulation, Neoplastic, Cell Movement, RNA-Binding Proteins metabolism, Esophageal Neoplasms drug therapy, Esophageal Neoplasms genetics, Esophageal Neoplasms metabolism, Esophageal Squamous Cell Carcinoma drug therapy, Esophageal Squamous Cell Carcinoma genetics, Esophageal Squamous Cell Carcinoma pathology, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell metabolism, MicroRNAs genetics, MicroRNAs metabolism
- Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly disease with few prevention or treatment options. ESCC development in humans and rodents is associated with Zn deficiency (ZD), inflammation, and overexpression of oncogenic microRNAs: miR-31 and miR-21. In a ZD-promoted ESCC rat model with upregulation of these miRs, systemic antimiR-31 suppresses the miR-31-EGLN3/STK40-NF-κB-controlled inflammatory pathway and ESCC. In this model, systemic delivery of Zn-regulated antimiR-31, followed by antimiR-21, restored expression of tumor-suppressor proteins targeted by these specific miRs: STK40/EGLN3 (miR-31), PDCD4 (miR-21), suppressing inflammation, promoting apoptosis, and inhibiting ESCC development. Moreover, ESCC-bearing Zn-deficient (ZD) rats receiving Zn medication showed a 47% decrease in ESCC incidence vs. Zn-untreated controls. Zn treatment eliminated ESCCs by affecting a spectrum of biological processes that included downregulation of expression of the two miRs and miR-31-controlled inflammatory pathway, stimulation of miR-21-PDCD4 axis apoptosis, and reversal of the ESCC metabolome: with decrease in putrescine, increase in glucose, accompanied by downregulation of metabolite enzymes ODC and HK2. Thus, Zn treatment or miR-31/21 silencing are effective therapeutic strategies for ESCC in this rodent model and should be examined in the human counterpart exhibiting the same biological processes.
- Published
- 2023
- Full Text
- View/download PDF
31. Abrogation of esophageal carcinoma development in miR-31 knockout rats.
- Author
-
Fong LY, Taccioli C, Palamarchuk A, Tagliazucchi GM, Jing R, Smalley KJ, Fan S, Altemus J, Fiehn O, Huebner K, Farber JL, and Croce CM
- Subjects
- Animals, Carcinogens toxicity, Cell Line, Tumor, Dietary Supplements, Esophageal Neoplasms chemically induced, Esophageal Neoplasms pathology, Esophageal Neoplasms prevention & control, Esophageal Squamous Cell Carcinoma chemically induced, Esophageal Squamous Cell Carcinoma pathology, Esophageal Squamous Cell Carcinoma prevention & control, Esophagus pathology, Gene Expression Regulation, Neoplastic, Gene Knockout Techniques, Humans, Male, MicroRNAs antagonists & inhibitors, MicroRNAs genetics, NF-kappa B metabolism, Neoplasms, Experimental chemically induced, Neoplasms, Experimental pathology, Neoplasms, Experimental prevention & control, Nitrosamines toxicity, Rats, Rats, Transgenic, Signal Transduction genetics, Zinc administration & dosage, Zinc deficiency, Esophageal Neoplasms genetics, Esophageal Squamous Cell Carcinoma genetics, Hypoxia-Inducible Factor-Proline Dioxygenases genetics, MicroRNAs metabolism, Neoplasms, Experimental genetics
- Abstract
MicroRNA-31 (miR-31) is overexpressed in esophageal squamous cell carcinoma (ESCC), a deadly disease associated with dietary Zn deficiency and inflammation. In a Zn deficiency-promoted rat ESCC model with miR-31 up-regulation, cancer-associated inflammation, and a high ESCC burden following N -nitrosomethylbenzylamine (NMBA) exposure, systemic antimiR-31 delivery reduced ESCC incidence from 85 to 45% ( P = 0.038) and miR-31 gene knockout abrogated development of ESCC ( P = 1 × 10
-6 ). Transcriptomics, genome sequencing, and metabolomics analyses in these Zn-deficient rats revealed the molecular basis of ESCC abrogation by miR-31 knockout. Our identification of EGLN3, a known negative regulator of nuclear factor κB (NF-κB), as a direct target of miR-31 establishes a functional link between oncomiR-31, tumor suppressor target EGLN3, and up-regulated NF-κB-controlled inflammation signaling. Interaction among oncogenic miR-31, EGLN3 down-regulation, and inflammation was also documented in human ESCCs. miR-31 deletion resulted in suppression of miR-31-associated EGLN3/NF-κB-controlled inflammatory pathways. ESCC-free, Zn-deficient miR-31-/- rat esophagus displayed no genome instability and limited metabolic activity changes vs. the pronounced mutational burden and ESCC-associated metabolic changes of Zn-deficient wild-type rats. These results provide conclusive evidence that miR-31 expression is necessary for ESCC development., Competing Interests: The authors declare no competing interest., (Copyright © 2020 the Author(s). Published by PNAS.)- Published
- 2020
- Full Text
- View/download PDF
32. Human-like hyperplastic prostate with low ZIP1 induced solely by Zn deficiency in rats.
- Author
-
Fong LY, Jing R, Smalley KJ, Wang ZX, Taccioli C, Fan S, Chen H, Alder H, Huebner K, Farber JL, Fiehn O, and Croce CM
- Subjects
- Adenocarcinoma genetics, Animals, Cell Proliferation, Citric Acid metabolism, Diet, Disease Models, Animal, Gene Expression Regulation, Neoplastic, Humans, Male, MicroRNAs biosynthesis, Prostatic Hyperplasia genetics, Prostatic Neoplasms genetics, Rats, Rats, Sprague-Dawley, Rats, Wistar, Signal Transduction genetics, Transcription, Genetic genetics, Tumor Cells, Cultured, Zinc metabolism, Adenocarcinoma pathology, Cation Transport Proteins metabolism, Prostate pathology, Prostatic Hyperplasia pathology, Prostatic Neoplasms pathology, Zinc deficiency
- Abstract
Prostate cancer is a leading cause of cancer death in men over 50 years of age, and there is a characteristic marked decrease in Zn content in the malignant prostate cells. The cause and consequences of this loss have thus far been unknown. We found that in middle-aged rats a Zn-deficient diet reduces prostatic Zn levels ( P = 0.025), increases cellular proliferation, and induces an inflammatory phenotype with COX-2 overexpression. This hyperplastic/inflammatory prostate has a human prostate cancer-like microRNA profile, with up-regulation of the Zn-homeostasis-regulating miR-183-96-182 cluster (fold change = 1.41-2.38; P = 0.029-0.0003) and down-regulation of the Zn importer ZIP1 (target of miR-182), leading to a reduction of prostatic Zn. This inverse relationship between miR-182 and ZIP1 also occurs in human prostate cancer tissue, which is known for Zn loss. The discovery that the Zn-depleted middle-aged rat prostate has a metabolic phenotype resembling that of human prostate cancer, with a 10-fold down-regulation of citric acid ( P = 0.0003), links citrate reduction directly to prostatic Zn loss, providing the underlying mechanism linking dietary Zn deficiency with miR-183-96-182 overexpression, ZIP1 down-regulation, prostatic Zn loss, and the resultant citrate down-regulation, changes mimicking features of human prostate cancer. Thus, dietary Zn deficiency during rat middle age produces changes that mimic those of human prostate carcinoma and may increase the risk for prostate cancer, supporting the need for assessment of Zn supplementation in its prevention., Competing Interests: The authors declare no conflict of interest., (Copyright © 2018 the Author(s). Published by PNAS.)
- Published
- 2018
- Full Text
- View/download PDF
33. Nit1 is a metabolite repair enzyme that hydrolyzes deaminated glutathione.
- Author
-
Peracchi A, Veiga-da-Cunha M, Kuhara T, Ellens KW, Paczia N, Stroobant V, Seliga AK, Marlaire S, Jaisson S, Bommer GT, Sun J, Huebner K, Linster CL, Cooper AJL, and Van Schaftingen E
- Subjects
- Aminohydrolases physiology, Animals, Deamination, Humans, Hydrolysis, Mice, Mice, Knockout, Substrate Specificity, Aminohydrolases metabolism, Glutathione metabolism, Saccharomyces cerevisiae Proteins metabolism, Transaminases metabolism
- Abstract
The mammalian gene Nit1 (nitrilase-like protein 1) encodes a protein that is highly conserved in eukaryotes and is thought to act as a tumor suppressor. Despite being ∼35% sequence identical to ω-amidase (Nit2), the Nit1 protein does not hydrolyze efficiently α-ketoglutaramate (a known physiological substrate of Nit2), and its actual enzymatic function has so far remained a puzzle. In the present study, we demonstrate that both the mammalian Nit1 and its yeast ortholog are amidases highly active toward deaminated glutathione (dGSH; i.e., a form of glutathione in which the free amino group has been replaced by a carbonyl group). We further show that Nit1 -KO mutants of both human and yeast cells accumulate dGSH and the same compound is excreted in large amounts in the urine of Nit1 -KO mice. Finally, we show that several mammalian aminotransferases (transaminases), both cytosolic and mitochondrial, can form dGSH via a common (if slow) side-reaction and provide indirect evidence that transaminases are mainly responsible for dGSH formation in cultured mammalian cells. Altogether, these findings delineate a typical instance of metabolite repair, whereby the promiscuous activity of some abundant enzymes of primary metabolism leads to the formation of a useless and potentially harmful compound, which needs a suitable "repair enzyme" to be destroyed or reconverted into a useful metabolite. The need for a dGSH repair reaction does not appear to be limited to eukaryotes: We demonstrate that Nit1 homologs acting as excellent dGSH amidases also occur in Escherichia coli and other glutathione-producing bacteria., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
34. Protective role of miR-155 in breast cancer through RAD51 targeting impairs homologous recombination after irradiation.
- Author
-
Gasparini P, Lovat F, Fassan M, Casadei L, Cascione L, Jacob NK, Carasi S, Palmieri D, Costinean S, Shapiro CL, Huebner K, and Croce CM
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms radiotherapy, Female, Humans, MCF-7 Cells, Models, Biological, Prognosis, Radiation Tolerance, Breast Neoplasms prevention & control, Homologous Recombination radiation effects, MicroRNAs physiology, Rad51 Recombinase genetics
- Abstract
Cell survival after DNA damage relies on DNA repair, the abrogation of which causes genomic instability and development of cancer. However, defective DNA repair in cancer cells can be exploited for cancer therapy using DNA-damaging agents. DNA double-strand breaks are the major lethal lesions induced by ionizing radiation (IR) and can be efficiently repaired by DNA homologous recombination, a system that requires numerous factors including the recombinase RAD51 (RAD51). Therapies combined with adjuvant radiotherapy have been demonstrated to improve the survival of triple-negative breast cancer patients; however, such therapy is challenged by the emergence of resistance in tumor cells. It is, therefore, essential to develop novel therapeutic strategies to overcome radioresistance and improve radiosensitivity. In this study we show that overexpression of microRNA 155 (miR-155) in human breast cancer cells reduces the levels of RAD51 and affects the cellular response to IR. miR-155 directly targets the 3'-untranslated region of RAD51. Overexpression of miR-155 decreased the efficiency of homologous recombination repair and enhanced sensitivity to IR in vitro and in vivo. High miR-155 levels were associated with lower RAD51 expression and with better overall survival of patients in a large series of triple-negative breast cancers. Taken together, our findings indicate that miR-155 regulates DNA repair activity and sensitivity to IR by repressing RAD51 in breast cancer. Testing for expression levels of miR-155 may be useful in the identification of breast cancer patients who will benefit from an IR-based therapeutic approach.
- Published
- 2014
- Full Text
- View/download PDF
35. Breast cancer signatures for invasiveness and prognosis defined by deep sequencing of microRNA.
- Author
-
Volinia S, Galasso M, Sana ME, Wise TF, Palatini J, Huebner K, and Croce CM
- Subjects
- Carcinoma in Situ genetics, Carcinoma in Situ pathology, Carcinoma, Intraductal, Noninfiltrating genetics, Carcinoma, Intraductal, Noninfiltrating pathology, Disease Progression, Female, Gene Expression Regulation, Neoplastic, Genes, Neoplasm genetics, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Kaplan-Meier Estimate, MicroRNAs metabolism, Neoplasm Invasiveness, Neoplasm Metastasis, Prognosis, Breast Neoplasms genetics, Breast Neoplasms pathology, Gene Expression Profiling, High-Throughput Nucleotide Sequencing methods, MicroRNAs genetics
- Abstract
The transition from ductal carcinoma in situ to invasive ductal carcinoma is a key event in breast cancer progression that is still not well understood. To discover the microRNAs regulating this critical transition, we used 80 biopsies from invasive ductal carcinoma, 8 from ductal carcinoma in situ, and 6 from normal breast. We selected them from a recently published deep-sequencing dataset [Farazi TA, et al. (2011) Cancer Res 71:4443-4453]. The microRNA profile established for the normal breast to ductal carcinoma in situ transition was largely maintained in the in situ to invasive ductal carcinoma transition. Nevertheless, a nine-microRNA signature was identified that differentiated invasive from in situ carcinoma. Specifically, let-7d, miR-210, and -221 were down-regulated in the in situ and up-regulated in the invasive transition, thus featuring an expression reversal along the cancer progression path. Additionally, we identified microRNAs for overall survival and time to metastasis. Five noncoding genes were associated with both prognostic signatures--miR-210, -21, -106b*, -197, and let-7i, with miR-210 the only one also involved in the invasive transition. To pinpoint critical cellular functions affected in the invasive transition, we identified the protein coding genes with inversely related profiles to miR-210: BRCA1, FANCD, FANCF, PARP1, E-cadherin, and Rb1 were all activated in the in situ and down-regulated in the invasive carcinoma. Additionally, we detected differential splicing isoforms with special features, including a truncated EGFR lacking the kinase domain and overexpressed only in ductal carcinoma in situ.
- Published
- 2012
- Full Text
- View/download PDF
36. Intramitochondrial calcium regulation by the FHIT gene product sensitizes to apoptosis.
- Author
-
Rimessi A, Marchi S, Fotino C, Romagnoli A, Huebner K, Croce CM, Pinton P, and Rizzuto R
- Subjects
- Acid Anhydride Hydrolases analysis, Calcium Signaling, HeLa Cells, Homeostasis, Humans, Neoplasm Proteins analysis, Vitamin K 3 pharmacology, Acid Anhydride Hydrolases physiology, Apoptosis drug effects, Calcium metabolism, Mitochondria metabolism, Neoplasm Proteins physiology
- Abstract
Despite the growing interest in the Fhit tumor suppressor protein, frequently deleted in human cancers, the mechanism of its powerful proapoptotic activity has remained elusive. We here demonstrate that Fhit sensitizes the low-affinity Ca(2+) transporters of mitochondria, enhancing Ca(2+) uptake into the organelle both in intact and in permabilized cells, and potentiating the effect of apoptotic agents. This effect can be attributed to the fraction of Fhit sorted to mitochondria, as a fully mitochondrial Fhit (a chimeric protein including a mitochondrial targeting sequence) retains the Ca(2+) signaling properties of Fhit and the proapoptotic activity of the native protein (whereas the effects on the cell cycle are lost). Thus, the partial sorting of Fhit to mitochondria allows to finely tune the sensitivity of the organelle to the highly pleiomorphic Ca(2+) signals, synergizing with apoptotic challenges. This concept, and the identification of the molecular machinery, may provide ways to act on apoptotic cell death and its derangement in cancer.
- Published
- 2009
- Full Text
- View/download PDF
37. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B.
- Author
-
Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E, Liu S, Alder H, Costinean S, Fernandez-Cymering C, Volinia S, Guler G, Morrison CD, Chan KK, Marcucci G, Calin GA, Huebner K, and Croce CM
- Subjects
- Carcinoma, Non-Small-Cell Lung genetics, DNA Methyltransferase 3A, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Humans, Lung enzymology, Lung Neoplasms enzymology, Lung Neoplasms mortality, Prognosis, RNA, Neoplasm genetics, Survival Analysis, DNA Methyltransferase 3B, Carcinoma, Non-Small-Cell Lung enzymology, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methylation, DNA, Neoplasm genetics, Lung Neoplasms genetics, MicroRNAs genetics
- Abstract
MicroRNAs (miRNAs) are small, noncoding RNAs that regulate expression of many genes. Recent studies suggest roles of miRNAs in carcinogenesis. We and others have shown that expression profiles of miRNAs are different in lung cancer vs. normal lung, although the significance of this aberrant expression is poorly understood. Among the reported down-regulated miRNAs in lung cancer, the miRNA (miR)-29 family (29a, 29b, and 29c) has intriguing complementarities to the 3'-UTRs of DNA methyltransferase (DNMT)3A and -3B (de novo methyltransferases), two key enzymes involved in DNA methylation, that are frequently up-regulated in lung cancer and associated with poor prognosis. We investigated whether miR-29s could target DNMT3A and -B and whether restoration of miR-29s could normalize aberrant patterns of methylation in non-small-cell lung cancer. Here we show that expression of miR-29s is inversely correlated to DNMT3A and -3B in lung cancer tissues, and that miR-29s directly target both DNMT3A and -3B. The enforced expression of miR-29s in lung cancer cell lines restores normal patterns of DNA methylation, induces reexpression of methylation-silenced tumor suppressor genes, such as FHIT and WWOX, and inhibits tumorigenicity in vitro and in vivo. These findings support a role of miR-29s in epigenetic normalization of NSCLC, providing a rationale for the development of miRNA-based strategies for the treatment of lung cancer.
- Published
- 2007
- Full Text
- View/download PDF
38. WWOX gene restoration prevents lung cancer growth in vitro and in vivo.
- Author
-
Fabbri M, Iliopoulos D, Trapasso F, Aqeilan RI, Cimmino A, Zanesi N, Yendamuri S, Han SY, Amadori D, Huebner K, and Croce CM
- Subjects
- Adenoviridae genetics, Animals, Apoptosis, Cell Cycle, Cell Line, Tumor, Female, Humans, Lung Neoplasms pathology, Mice, Tumor Suppressor Proteins, WW Domain-Containing Oxidoreductase, Lung Neoplasms prevention & control, Oxidoreductases genetics
- Abstract
The WWOX (WW domain containing oxidoreductase) gene at the common fragile site, FRA16D, is altered in many types of cancer, including lung cancer. We have examined the tumor suppressor function of WWOX in preclinical lung cancer models. The WWOX gene was expressed in lung cancer cell lines through recombinant adenovirus (Ad) infection (Ad-WWOX), and through a drug [ponasterone A, (ponA)]-inducible system. After WWOX restoration in vitro, endogenous Wwox protein-negative cell lines (A549, H460, and H1299) underwent apoptosis through activation of the intrinsic apoptotic caspase cascade in A549 and H460 cells. Ectopic expression of Wwox caused dramatic suppression of tumorigenicity of A549, H460, and H1299 cells in nude mice after Ad-WWOX infection and after ponA induction of Wwox expression in H1299 lung cancer cells. Tumorigenicity and in vitro growth of U2020 (Wwox-positive) lung cancer cells was unaffected by Wwox overexpression. This study confirms that WWOX is a tumor suppressor gene and is highly effective in preventing growth of lung cancer xenografts, whether introduced through viral infection or by induction of a silent WWOX transgene.
- Published
- 2005
- Full Text
- View/download PDF
39. Frag1, a homolog of alternative replication factor C subunits, links replication stress surveillance with apoptosis.
- Author
-
Ishii H, Inageta T, Mimori K, Saito T, Sasaki H, Isobe M, Mori M, Croce CM, Huebner K, Ozawa K, and Furukawa Y
- Subjects
- Amino Acid Sequence, Animals, Ataxia Telangiectasia Mutated Proteins, Base Sequence, Blotting, Northern, Bromodeoxyuridine, Cell Cycle Proteins metabolism, Cells, Cultured, Chromatin metabolism, Flow Cytometry, Humans, Immunoprecipitation, Mice, Molecular Sequence Data, Nuclear Proteins metabolism, Plasmids genetics, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, RNA, Small Interfering genetics, S Phase physiology, Sequence Analysis, DNA, bcl-2-Associated X Protein, Apoptosis genetics, DNA Damage genetics, Gene Expression Regulation, Genomic Instability genetics, Nuclear Proteins genetics, Signal Transduction genetics
- Abstract
We report the identification and characterization of a potent regulator of genomic integrity, mouse and human FRAG1 gene, a conserved homolog of replication factor C large subunit that is homologous to the alternative replication factor C subunits Elg1, Ctf18/Chl12, and Rad24 of budding yeast. FRAG1 was identified in a search for key caretaker genes involved in the regulation of genomic stability under conditions of replicative stress. In response to stress, Atr participates in the down-regulation of FRAG1 expression, leading to the induction of apoptosis through the release of Rad9 from damaged chromatin during the S phase of the cell cycle, allowing Rad9-Bcl2 association and induction of proapoptotic Bax protein. We propose that the Frag1 signal pathway, by linking replication stress surveillance with apoptosis induction, plays a central role in determining whether DNA damage is compatible with cell survival or whether it requires cell elimination by apoptosis.
- Published
- 2005
- Full Text
- View/download PDF
40. Functional association between Wwox tumor suppressor protein and p73, a p53 homolog.
- Author
-
Aqeilan RI, Pekarsky Y, Herrero JJ, Palamarchuk A, Letofsky J, Druck T, Trapasso F, Han SY, Melino G, Huebner K, and Croce CM
- Subjects
- Cell Line, Cell Line, Tumor, Gene Expression Regulation, Genes, Reporter, Genes, Tumor Suppressor, Green Fluorescent Proteins, Humans, Luminescent Proteins genetics, Luminescent Proteins metabolism, Phosphorylation, Plasmids, Protein Binding, RNA, Small Interfering genetics, Recombinant Fusion Proteins metabolism, Transcription, Genetic, Transcriptional Activation, Transfection, Tumor Protein p73, Tumor Suppressor Proteins, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism
- Abstract
The WWOX gene is a recently cloned tumor suppressor gene that spans the FRA16D fragile region. Wwox protein contains two WW domains that are generally known to mediate protein-protein interaction. Here we show that Wwox physically interacts via its first WW domain with the p53 homolog, p73. The tyrosine kinase, Src, phosphorylates Wwox at tyrosine 33 in the first WW domain and enhances its binding to p73. Our results further demonstrate that Wwox expression triggers redistribution of nuclear p73 to the cytoplasm and, hence, suppresses its transcriptional activity. In addition, we show that cytoplasmic p73 contributes to the proapoptotic activity of Wwox. Our findings reveal a functional cross-talk between p73 and Wwox tumor suppressor protein.
- Published
- 2004
- Full Text
- View/download PDF
41. Fhit is a physiological target of the protein kinase Src.
- Author
-
Pekarsky Y, Garrison PN, Palamarchuk A, Zanesi N, Aqeilan RI, Huebner K, Barnes LD, and Croce CM
- Subjects
- 3T3 Cells, Animals, Blotting, Western, Chromatography, High Pressure Liquid, Humans, Mass Spectrometry, Mice, Phosphorylation, Acid Anhydride Hydrolases, Neoplasm Proteins metabolism, src-Family Kinases metabolism
- Abstract
The FHIT gene is a tumor suppressor that is frequently inactivated by genomic alterations at chromosomal region 3p14.2. In the last few years, a considerable amount of data describing inactivation of FHIT in a variety of human malignancies and demonstrating the tumor suppressor potential of Fhit have been reported. Despite the demonstration that FHIT functions as a tumor suppressor, the pathway through which Fhit induces apoptosis and inhibits growth of cancer cells is not known. Our data demonstrate that Fhit is a target of tyrosine phosphorylation by the Src protein kinase. We show that Src phosphorylates Y114 of Fhit in vitro and in vivo, providing insight into a biochemical pathway involved in Fhit signaling.
- Published
- 2004
- Full Text
- View/download PDF
42. Fragile site orthologs FHIT/FRA3B and Fhit/Fra14A2: evolutionarily conserved but highly recombinogenic.
- Author
-
Matsuyama A, Shiraishi T, Trapasso F, Kuroki T, Alder H, Mori M, Huebner K, and Croce CM
- Subjects
- Animals, Conserved Sequence, DNA genetics, DNA Damage, Databases, Genetic, Exons, Humans, Mice, Molecular Sequence Data, Repetitive Sequences, Nucleic Acid, Acid Anhydride Hydrolases, Chromosome Fragile Sites, Evolution, Molecular, Genes, Tumor Suppressor, Neoplasm Proteins genetics, Recombination, Genetic
- Abstract
Common fragile sites are regions that show elevated susceptibility to DNA damage, leading to alterations that can contribute to cancer development. FRA3B, located at chromosome region 3p14.2, is the most frequently expressed human common fragile site, and allelic losses at FRA3B have been observed in many types of cancer. The FHIT gene, encompassing the FRA3B region, is a tumor-suppressor gene. To identify the features of FHIT/FRA3B that might contribute to fragility, sequences of the human FHIT and the flanking PTPRG gene were compared with those of murine Fhit and Ptprg. Human and mouse orthologous genes, FHIT and Fhit, are more highly conserved through evolution than PTPRG/Ptprg and yet contain more sequence elements that are exquisitely sensitive to genomic rearrangements, such as high-flexibility regions and long interspersed nuclear element 1s, suggesting that common fragile sites serve a function. The conserved AT-rich high-flexibility regions are the most characteristic of common fragile sites.
- Published
- 2003
- Full Text
- View/download PDF
43. Designed FHIT alleles establish that Fhit-induced apoptosis in cancer cells is limited by substrate binding.
- Author
-
Trapasso F, Krakowiak A, Cesari R, Arkles J, Yendamuri S, Ishii H, Vecchione A, Kuroki T, Bieganowski P, Pace HC, Huebner K, Croce CM, and Brenner C
- Subjects
- Cell Line, Flow Cytometry, Humans, Kinetics, Mutation, Neoplasm Proteins isolation & purification, Neoplasm Proteins metabolism, Neoplasm Proteins physiology, Neoplasms genetics, Substrate Specificity, Tumor Cells, Cultured, Acid Anhydride Hydrolases, Alleles, Apoptosis physiology, Neoplasm Proteins genetics, Neoplasms pathology
- Abstract
The FHIT gene is inactivated early in the development of many human tumors, and Fhit-deficient mice have increased cancer incidence. Viral reexpression of Fhit kills Fhit-deficient cells by induction of apoptosis. Fhit, a member of branch 2 of the histidine-triad superfamily of nucleoside monophosphate hydrolases and transferases, is a diadenosine polyphosphate hydrolase, the active-site histidine of which is not required for tumor suppression. To provide a rigorous test of the hypothesis that Fhit function depends on forming a complex with substrates, we designed a series of alleles of Fhit intended to reduce substrate-binding andor hydrolytic rates, characterized these mutants biochemically, and then performed quantitative cell-death assays on cancer cells virally infected with each allele. The allele series covered defects as great as 100,000-fold in k(cat) and increases as large as 30-fold in K(M). Nonetheless, when mutant FHIT genes were expressed in two human cancer cell lines containing FHIT deletions, reductions in apoptotic activity correlated exclusively with K(M). Mutants with 2- and 7-fold increases in K(M) significantly reduced apoptotic indices, whereas the mutant with a 30-fold increase in K(M) retained little cellular function. These data indicate that the proapoptotic function of Fhit is limited by substrate binding and is unrelated to substrate hydrolysis.
- Published
- 2003
- Full Text
- View/download PDF
44. Tumor suppressors on 3p: a neoclassic quartet.
- Author
-
Huebner K
- Subjects
- Animals, Carcinogens pharmacology, Chromosome Mapping, Humans, Loss of Heterozygosity, Mice, Mice, Knockout, Chromosomes, Human, Pair 3, Genes, Tumor Suppressor
- Published
- 2001
- Full Text
- View/download PDF
45. The tumor spectrum in FHIT-deficient mice.
- Author
-
Zanesi N, Fidanza V, Fong LY, Mancini R, Druck T, Valtieri M, Rüdiger T, McCue PA, Croce CM, and Huebner K
- Subjects
- Animals, Carcinogens toxicity, Dimethylnitrosamine analogs & derivatives, Dimethylnitrosamine toxicity, Female, Genotype, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Neoplasms, Experimental chemically induced, Neoplasms, Experimental pathology, Phenotype, Stomach Neoplasms chemically induced, Stomach Neoplasms genetics, Stomach Neoplasms pathology, Acid Anhydride Hydrolases, Neoplasm Proteins, Neoplasms, Experimental genetics, Proteins genetics
- Abstract
Mice carrying one inactivated Fhit allele (Fhit +/- mice) are highly susceptible to tumor induction by N-nitrosomethylbenzylamine, with 100% of Fhit +/- mice exhibiting tumors of the forestomach/squamocolumnar junction vs. 25% of Fhit +/+ controls. In the current study a single N-nitrosomethylbenzylamine dose was administered to Fhit +/+, +/-, and -/- mice to compare carcinogen susceptibility in +/- and -/- Fhit-deficient mice. At 29 weeks after treatment, 7.7% of wild-type mice had tumors. Of the Fhit -/- mice 89.5% exhibited tumors (average 3.3 tumors/mouse) of the forestomach and squamocolumnar junction; half of the -/- mice had medium (2 mm diameter) to large (>2 mm) tumors. Of the Fhit +/- mice 78% exhibited tumors (average 2.4 tumors/mouse) and 22% showed medium to large tumors. Untreated Fhit-deficient mice have been observed for up to 2 years for spontaneous tumors. Fhit +/- mice (average age 21 mo) exhibit an average of 0.94 tumors of different types; Fhit -/- mice (average age 16 mo) also showed an array of tumors (average 0.76 tumor/mouse). The similar spontaneous and induced tumor spectra observed in mice with one or both Fhit alleles inactivated suggests that Fhit may be a one-hit tumor suppressor gene in some tissues.
- Published
- 2001
- Full Text
- View/download PDF
46. Sequence conservation at human and mouse orthologous common fragile regions, FRA3B/FHIT and Fra14A2/Fhit.
- Author
-
Shiraishi T, Druck T, Mimori K, Flomenberg J, Berk L, Alder H, Miller W, Huebner K, and Croce CM
- Subjects
- Animals, Chromosome Fragile Sites, Exons, Humans, Mice, Molecular Sequence Data, Polymerase Chain Reaction, Repetitive Sequences, Nucleic Acid, Acid Anhydride Hydrolases, Chromosome Fragility, Neoplasm Proteins, Proteins genetics
- Abstract
It has been suggested that delayed DNA replication underlies fragility at common human fragile sites, but specific sequences responsible for expression of these inducible fragile sites have not been identified. One approach to identify such cis-acting sequences within the large nonexonic regions of fragile sites would be to identify conserved functional elements within orthologous fragile sites by interspecies sequence comparison. This study describes a comparison of orthologous fragile regions, the human FRA3B/FHIT and the murine Fra14A2/Fhit locus. We sequenced over 600 kbp of the mouse Fra14A2, covering the region orthologous to the fragile epicenter of FRA3B, and determined the Fhit deletion break points in a mouse kidney cancer cell line (RENCA). The murine Fra14A2 locus, like the human FRA3B, was characterized by a high AT content. Alignment of the two sequences showed that this fragile region was stable in evolution despite its susceptibility to mitotic recombination on inhibition of DNA replication. There were also several unusual highly conserved regions (HCRs). The positions of predicted matrix attachment regions (MARs), possibly related to replication origins, were not conserved. Of known fragile region landmarks, five cancer cell break points, one viral integration site, and one aphidicolin break cluster were located within or near HCRs. Thus, comparison of orthologous fragile regions has identified highly conserved sequences with possible functional roles in maintenance of fragility.
- Published
- 2001
- Full Text
- View/download PDF
47. FHIT gene therapy prevents tumor development in Fhit-deficient mice.
- Author
-
Dumon KR, Ishii H, Fong LY, Zanesi N, Fidanza V, Mancini R, Vecchione A, Baffa R, Trapasso F, During MJ, Huebner K, and Croce CM
- Subjects
- Adenoviridae genetics, Animals, Carcinogens adverse effects, Dependovirus genetics, Dimethylnitrosamine adverse effects, Genetic Vectors genetics, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Neoplasm Proteins metabolism, Neoplasm Proteins physiology, Neoplasms, Experimental chemically induced, Neoplasms, Experimental pathology, Acid Anhydride Hydrolases, Dimethylnitrosamine analogs & derivatives, Genes, Tumor Suppressor physiology, Genetic Therapy methods, Neoplasm Proteins genetics, Neoplasms, Experimental prevention & control
- Abstract
The tumor suppressor gene FHIT spans a common fragile site and is highly susceptible to environmental carcinogens. FHIT inactivation and loss of expression is found in a large fraction of premaligant and malignant lesions. In this study, we were able to inhibit tumor development by oral gene transfer, using adenoviral or adenoassociated viral vectors expressing the human FHIT gene, in heterozygous Fhit(+/-) knockout mice, that are prone to tumor development after carcinogen exposure. We therefore suggest that FHIT gene therapy could be a novel clinical approach not only in treatment of early stages of cancer, but also in prevention of human cancer.
- Published
- 2001
- Full Text
- View/download PDF
48. Muir-Torre-like syndrome in Fhit-deficient mice.
- Author
-
Fong LY, Fidanza V, Zanesi N, Lock LF, Siracusa LD, Mancini R, Siprashvili Z, Ottey M, Martin SE, Druck T, McCue PA, Croce CM, and Huebner K
- Subjects
- Adenoma genetics, Animals, Carcinogens, Dimethylnitrosamine analogs & derivatives, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Inbred Strains, Mice, Knockout, Neoplasm Proteins deficiency, Neoplasm Proteins genetics, Neoplasms, Multiple Primary chemically induced, Neoplasms, Multiple Primary pathology, Papilloma genetics, Proteins metabolism, Restriction Mapping, Sebaceous Gland Neoplasms genetics, Stomach Neoplasms chemically induced, Stomach Neoplasms genetics, Stomach Neoplasms pathology, Syndrome, Acid Anhydride Hydrolases, Neoplasms, Multiple Primary genetics, Proteins genetics
- Abstract
To investigate the role of the Fhit gene in carcinogen induction of neoplasia, we have inactivated one Fhit allele in mouse embryonic stem cells and produced (129/SvJ x C57BL/6J) F(1) mice with a Fhit allele inactivated (+/-). Fhit +/+ and +/- mice were treated intragastrically with nitrosomethylbenzylamine and observed for 10 wk posttreatment. A total of 25% of the +/+ mice developed adenoma or papilloma of the forestomach, whereas 100% of the +/- mice developed multiple tumors that were a mixture of adenomas, squamous papillomas, invasive carcinomas of the forestomach, as well as tumors of sebaceous glands. The visceral and sebaceous tumors, which lacked Fhit protein, were similar to those characteristic of Muir-Torre familial cancer syndrome.
- Published
- 2000
- Full Text
- View/download PDF
49. Cancer-specific chromosome alterations in the constitutive fragile region FRA3B.
- Author
-
Mimori K, Druck T, Inoue H, Alder H, Berk L, Mori M, Huebner K, and Croce CM
- Subjects
- Humans, Molecular Sequence Data, Neoplasm Proteins genetics, Sequence Deletion, Translocation, Genetic, Tumor Cells, Cultured, Acid Anhydride Hydrolases, Biomarkers, Tumor, Chromosome Fragility genetics, Genetic Markers, Neoplasms genetics, Proteins genetics
- Abstract
We have sequenced 870 kilobases of the FHIT/FRA3B locus, from FHIT intron 3 to intron 7. The locus is AT rich (61.5%) and Alu poor (6. 2%), and it apparently does not harbor other genes. In a detailed analysis of the 308-kilobase region between FHIT exon 5 and the telomeric end of intron 3, a region known to encompass a human papillomavirus-16 integration site and two clusters of aphidicolin-induced chromosome 3p14.2 breakpoints, we have precisely mapped 10 deletion and translocation endpoints in cancer-derived cell lines relative to positions of specific repetitive elements, regions of high genome flexibility and aphidicolin-induced breakpoints. Conclusions are (i) that aphidicolin-induced breakpoint clusters fall close to high-flexibility sequences, suggesting that these sequences contribute directly to aphidicolin-induced fragility; (ii) that 9 of the 10 FHIT allelic deletions in cancer cell lines resulted in loss of exons, with 7 deletion endpoints near long interspersed nuclear elements or long terminal repeat elements; and (iii) that cancer-specific deletions encompass multiple high-flexibility genomic regions, suggesting that fragile breaks may occur at these regions, whereas repair of the breaks involves homologous pairing of flanking sequences with concomitant deletion of the damaged fragile sequence.
- Published
- 1999
- Full Text
- View/download PDF
50. Nitrilase and Fhit homologs are encoded as fusion proteins in Drosophila melanogaster and Caenorhabditis elegans.
- Author
-
Pekarsky Y, Campiglio M, Siprashvili Z, Druck T, Sedkov Y, Tillib S, Draganescu A, Wermuth P, Rothman JH, Huebner K, Buchberg AM, Mazo A, Brenner C, and Croce CM
- Subjects
- Alternative Splicing, Amino Acid Sequence, Animals, Base Sequence, Cloning, Molecular, DNA, Complementary, Humans, Mice, Molecular Sequence Data, Sequence Homology, Amino Acid, Acid Anhydride Hydrolases, Aminohydrolases genetics, Caenorhabditis elegans genetics, Drosophila melanogaster genetics, Neoplasm Proteins, Proteins genetics, Recombinant Fusion Proteins genetics
- Abstract
The tumor suppressor gene FHIT encompasses the common human chromosomal fragile site at 3p14.2 and numerous cancer cell biallelic deletions. To study Fhit function we cloned and characterized FHIT genes from Drosophila melanogaster and Caenorhabditis elegans. Both genes code for fusion proteins in which the Fhit domain is fused with a novel domain showing homology to bacterial and plant nitrilases; the D. melanogaster fusion protein exhibited diadenosine triphosphate (ApppA) hydrolase activity expected of an authentic Fhit homolog. In human and mouse, the nitrilase homologs and Fhit are encoded by two different genes: FHIT and NIT1, localized on chromosomes 3 and 1 in human, and 14 and 1 in mouse, respectively. We cloned and characterized human and murine NIT1 genes and determined their exon-intron structure, patterns of expression, and alternative processing of their mRNAs. The tissue specificity of expression of murine Fhit and Nit1 genes was nearly identical. Because fusion proteins with dual or triple enzymatic activities have been found to carry out specific steps in a given biochemical or biosynthetic pathway, we postulate that Fhit and Nit1 likewise collaborate in a biochemical or cellular pathway in mammalian cells.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.