scispace - formally typeset
Search or ask a question

Showing papers by "Carlo M. Croce published in 2001"


PatentDOI
TL;DR: The physical interaction between endogenous Akt1 and TCL1 occurs through the PH domain of the Akt 1 protein this article, and the identification of Tcl1 mimics and Tcl 1 antagonists that modifying this interaction, with the subsequent modification of apoptotic and proliferative signals.
Abstract: The TCL1 oncogene at 14q32.1 is involved in the development of human leukemia. This invention demonstrates the interaction between the Tcl1 and the Akt1 proteins. The physical interaction between endogenous Akt1 and Tcl1 occurs through the PH domain of the Akt1 protein. The present invention relates to the identification of Tcl1 mimics and Tcl1 antagonists that modifying this interaction, with the subsequent modification of apoptotic and proliferative signals.

320 citations


Journal ArticleDOI
09 Nov 2001-Science
TL;DR: In this paper, the trithorax acetylation complex (TAC1) was purified from Drosophila embryos, containing Trx, dCBP, and Sbf1.
Abstract: Trithorax (Trx) is a member of the trithorax group (trxG) of epigenetic regulators, which is required to maintain active states of Hox gene expression during development. We have purified from Drosophila embryos a trithorax acetylation complex (TAC1) that contains Trx, dCBP, and Sbf1. Like CBP, TAC1 acetylates core histones in nucleosomes, suggesting that this activity may be important for epigenetic maintenance of gene activity. dCBP and Sbf1 associate with specific sites on salivary gland polytene chromosomes, colocalizing with many Trx binding sites. One of these is the site of the Hox gene Ultrabithorax ( Ubx ). Mutations in either trx or the gene encoding dCBP reduce expression of the endogenous Ubx gene as well as of transgenes driven by the bxd regulatory region of Ubx . Thus Trx, dCBP, and Sbf1 are closely linked, physically and functionally, in the maintenance of Hox gene expression.

217 citations


Journal ArticleDOI
TL;DR: The analyses of precise interactions in the human REV proteins suggest the possibility that hREV7 might play an important role in regulating the enzymatic activities of h REV1 and hREv3 for mutagenesis in response to DNA damage.

214 citations


Journal ArticleDOI
15 Feb 2001-Oncogene
TL;DR: The present work suggests that upregulation of Meis1, HoxA9, and possibly H CoxA10 might also play a role in pathogenesis of acute lymphocytic and acute myeloid leukemias associated with ALL-1 fusions.
Abstract: Rearrangements of the human ALL-1 gene are frequently encountered in acute lymphocytic leukemias (ALL) and acute myeloid leukemias (AML). These rearrangements are mostly due to chromosome translocations and result in production of chimeric proteins composed of the N-terminal fragment of ALL-1 and the C-terminal segments of the partner proteins. The most common chromosome translocation involving ALL-1 is the t(4 : 11) associated with ALL. ALL-1 is the human homologue of Drosophila trithorax and directly activates transcription of multiple Hox genes. A preliminary DNA microarray screen indicated that the Meis1, HoxA9 and AC133 genes were overexpressed in ALLs with t(4 : 11), compared to ALLs with very similar phenotype but without the chromosomal abnormality. These genes, as well as additional five Hox genes, were subjected to comprehensive semi-quantitative or quantitative RT-PCR analysis in 57 primary ALL and AML tumors. Meis1 and HoxA9 were found expressed in 13/14 of ALLs with the t(4 : 11) and in 8/8 of AMLs with ALL-1 rearrangements. The two genes were not consistently transcribed in other types of ALL. AC133 was transcribed in 13/14 of ALLs with t(4 : 11), but in only 4/8 of AMLs with ALL-1 rearrangements. HoxA10 was expressed in most leukemias with ALL-1 alterations, but was also transcribed in PrePreB CD10(-) ALLs lacking the t(4 : 11). Expression of HoxA5, HoxA7, HoxC8 and HoxC10 did not correlate with ALL-1 rearrangements. Coexpression of Meis1 and HoxA9, overexpression of HoxA10, and overexpression or fusion of HoxA9 were previously implicated in certain acute myeloid leukemias in mice and humans. The present work suggests that upregulation of Meis1, HoxA9, and possibly HoxA10 might also play a role in pathogenesis of acute lymphocytic and acute myeloid leukemias associated with ALL-1 fusions.

185 citations


Journal ArticleDOI
TL;DR: In 1979, the first chromosome alteration associated with familial cancer was reported, and five years later, a fragile site was observed in the same chromosome region, indicating that a 'one-hit' mechanism of tumorigenesis is operative.
Abstract: In 1979, the first chromosome alteration associated with familial cancer was reported. Five years later, a fragile site was observed in the same chromosome region. The product of the fragile histidine triad (FHIT) gene, which encompasses this fragile site, is partially or entirely lost in most human cancers, indicating that it has a tumour-suppressor function. Inactivation of only one FHIT allele compromises this suppressor function, indicating that a 'one-hit' mechanism of tumorigenesis is operative. Are genes disrupted at other fragile sites? And, are these genes also tumour suppressors?

178 citations


Journal ArticleDOI
TL;DR: 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.
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.

174 citations


Journal ArticleDOI
TL;DR: It is shown that Akt interacts with Nur77 and inactivates Nur77 by phosphorylated Ser-350 of the Nur77 protein within its DNA-binding domain in a phosphatidylinositol 3-kinase-dependent manner, connecting the phosph atidyl inositol3-kinases-dependent Akt pathway and a nuclear receptor pathway.
Abstract: The immediate early gene NUR77 (also called NGFI-B) is required for T cell antigen receptor-mediated cell death and is induced to very high levels in immature thymocytes and T cell hybridomas undergoing apoptosis. The Akt (PKB) kinase is a key player in transduction of anti-apoptotic and proliferative signals in T cells. Because Nur77 has a putative Akt phosphorylation site at Ser-350, and phosphorylation of this residue is critical for the transactivation activity of Nur77, we investigated whether Akt regulates Nur77. Coimmunoprecipitation experiments showed the detection of Nur77 in Akt immune complexes, suggesting that Nur77 and Akt physically interact. We further show that Akt specifically phosphorylates Ser-350 of the Nur77 protein within its DNA-binding domain in vitro and in vivo in 293 and NIH 3T3 cells. Because phosphorylation of Ser-350 of Nur77 is critical for its function as a transcription factor, we examined the effect of Akt on this function. By using luciferase assay experiments, we showed that phosphorylation of Nur77 by Akt decreased the transcriptional activity of Nur77 by 50--85%. Thus, we show that Akt interacts with Nur77 and inactivates Nur77 by phosphorylation at Ser-350 in a phosphatidylinositol 3-kinase-dependent manner, connecting the phosphatidylinositol 3-kinase-dependent Akt pathway and a nuclear receptor pathway.

172 citations


Journal ArticleDOI
TL;DR: An unexpected property of the proapoptotic gene caspase3 on cardiac contractility is documented, despite inducing ultrastructural damage, that does not trigger a full apoptotic response in the cardiomyocyte.
Abstract: Up-regulation of proapoptotic genes has been reported in heart failure and myocardial infarction. To determine whether caspase genes can affect cardiac function, a transgenic mouse was generated. Cardiac tissue-specific overexpression of the proapoptotic gene Caspase3 was induced by using the rat promoter of alpha-myosin heavy chain, a model that may represent a unique tool for investigating new molecules and antiapoptotic therapeutic strategies. Cardiac-specific Caspase3 expression induced transient depression of cardiac function and abnormal nuclear and myofibrillar ultrastructural damage. When subjected to myocardial ischemia-reperfusion injury, Caspase3 transgenic mice showed increased infarct size and a pronounced susceptibility to die. In this report, we document an unexpected property of the proapoptotic gene caspase3 on cardiac contractility. Despite inducing ultrastructural damage, Caspase3 does not trigger a full apoptotic response in the cardiomyocyte. We also implicate Caspase3 in determining myocardial infarct size after ischemia-reperfusion injury, because its cardiomyocyte-specific overexpression increases infarct size.

166 citations


Journal Article
TL;DR: A segment of 790 kb at 13q14 spanning the minimal region of loss in CLL is completely sequenced and characterized, finding that this feature of the sequence may favor the occurrence of chromosomal rearrangements and may confer instability to the region, resulting in deletions that may inactivate an as yet unidentified tumor suppressor.
Abstract: Chromosome 13q14 deletions constitute the most common genetic abnormality in chronic lymphocytic leukemia (CLL). To identify the putative tumor suppressor gene targeted by 13q14 genomic loss, we completely sequenced and characterized a segment of 790 kb at 13q14 spanning the minimal region of loss in CLL. Transcribed sequences in the region were identified through database homology searches and exon-prediction analysis. Two-hundred kb at the centromeric end of the sequence contain five CpG islands, three previously identified genes LEU5/RFP2, LEU2, and LEU1, seven of seven EST clusters composed of >10 ESTs, and a large number of predicted exons. Homology searches against the mouse EST database have allowed us to identify a highly conserved alternative first exon of the LEU2 gene, giving rise to a novel transcript, ALT1 (GenBank accession no. AF380424), which originates within a G+C region in the vicinity of the D13S272 marker. Two novel 3′ exons of LEU2 were also identified and are present in both LEU2 and ALT1 transcripts. However, we have not identified any mutations in leukemia cases, or alterations in expression of mRNAs in the region, that might directly implicate these mRNAs in the pathology of CLL. The centromeric end of the sequence, where all reported genes are located, contains twice the expected amount of ALU repeats, whereas the telomeric end is LINE1 rich and contains four LINE1 elements longer than 4 kb, including two full-length LINE1 sequences. This feature of the sequence may favor the occurrence of chromosomal rearrangements and may confer instability to the region, resulting in deletions that may inactivate an as yet unidentified tumor suppressor.

154 citations


Journal Article
TL;DR: Results showed suppression of cell growth in vitro in three of seven esophageal cancer cell lines, all seven of which showed abundant expression of the FHIT gene, suggesting that adenoviral-FHIT infection should be explored as a therapeutic strategy.
Abstract: Reintroduction of a tumor suppressor gene product in cancer cells is a promising strategy for cancer gene therapy. The fragile histidine triad (FHIT) gene has been identified in a region at chromosome 3p14.2, which is deleted in many tumors, including esophageal cancer. Previous studies have shown frequent biallelic alterations of the FHIT gene in numerous tumors, and have demonstrated a tumor suppressor function of Fhit. We have studied the biological effects of adenoviral-FHIT transduction in esophageal cancer cell lines. Results showed suppression of cell growth in vitro in three of seven esophageal cancer cell lines, all seven of which showed abundant expression of the transgene. Adenoviral-FHIT expression, but not control adenoviral infections, induced caspase-dependent apoptosis in two esophageal cancer cell lines, TE14 and TE4, which express no or very little Fhit, respectively. Treatment of TE14 cells with adenoviral-FHIT vectors resulted in abrogation of tumorigenicity in nude mice. A third esophageal cancer cell line, TE12, without detectable endogenous Fhit, showed accumulation of cells at S to G2-M and a small apoptotic cell fraction after adenoviral-FHIT transduction. Thus, adenoviral-FHIT expression can inhibit the growth of esophageal cancer cells, at least in part through caspase-dependent apoptosis, suggesting that adenoviral-FHIT infection should be explored as a therapeutic strategy.

126 citations


Journal ArticleDOI
TL;DR: It is proposed that adipocytic cell growth derives from the balance of the HMGA1 and HMGA2 protein functions, which are involved in the regulation of chromatin structure and DNA-binding sites have been identified in functional regions of many gene promoters.
Abstract: The HMGA family is comprised of four proteins: HMGA1a, HMGA1b, HMGA1c and HMGA2. The first three proteins are products of the same gene, HMGA1, generated through an alternative splicing mechanism. The HMGA proteins are involved in the regulation of chromatin structure and HMGA DNA-binding sites have been identified in functional regions of many gene promoters. Rearrangements of the HMGA2 gene have been frequently detected in human benign tumors of mesenchymal origin including lipomas. 12q13-15 chromosomal translocations involving the HMGA2 gene locus, account for these rearrangements. The HMGA proteins have three AT-hook domains and an acidic C-terminal tail. The HMGA2 modifications consist in the loss of the C-terminal tail and fusion with ectopic sequences. A pivotal role of the HMGA2 rearrangements in the process of lipomagenesis is suggested by experiments showing that transgenic mice carrying a truncated HMGA2 gene showed a giant phenotype together with abdominal/pelvic lipomatosis. As HMGA2 null mice showed a great reduction in fat tissue, a positive role of the HMGA2 gene in adipocytic cell proliferation is proposed. More recently, similar alterations of the HMGA1 gene have been described. As the block of the HMGA1 protein synthesis induces an increase in growth rate of the pre-adipocytic cell line 3T3-L1, we suggest a negative role of the HMGA1 proteins in adipocytic cell growth and, therefore, we propose that adipocytic cell growth derives from the balance of the HMGA1 and HMGA2 protein functions.

Journal ArticleDOI
TL;DR: It is shown that introduction of FEZ1/LZTS1 into Fez1/ Lzts1-negative cancer cells results in suppression of tumorigenicity and reduced cell growth with accumulation of cells at late S–G2/M stage of the cell cycle.
Abstract: The FEZ1/LZTS1 gene maps to chromosome 8p22, a region that is frequently deleted in human tumors. Alterations in FEZ1/LZTS1 expression have been observed in esophageal, breast, and prostate cancers. Here, we show that introduction of FEZ1/LZTS1 into Fez1/Lzts1-negative cancer cells results in suppression of tumorigenicity and reduced cell growth with accumulation of cells at late S–G2/M stage of the cell cycle. Fez1/Lzts1 protein is hyperphosphorylated by cAMP-dependent kinase during cell-cycle progression. We found that Fez1/Lzts1 is associated with microtubule components and interacts with p34cdc2 at late S–G2/M stage in vivo. Present data show that FEZ1/LZTS1 inhibits cancer cell growth through regulation of mitosis, and that its alterations result in abnormal cell growth.

Journal Article
TL;DR: In vivo results show that FHit gene transfer delays tumor growth and prolongs survival in a murine model mimicking human disease, and viral FHIT gene delivery results in apoptosis by activation of the caspase pathway.
Abstract: The fragile histidine triad (FHIT) gene is a tumor suppressor gene that is altered by deletion in a large fraction of human tumors, including pancreatic cancer. To evaluate the potential of FHIT gene therapy, we developed recombinant adenoviral and adenoassociated viral (AAV) FHIT vectors and tested these vectors in vitro and in vivo for activity against human pancreatic cancer cells. Our data show that viral FHIT gene delivery results in apoptosis by activation of the caspase pathway. Furthermore, Fhit overexpression enhances the susceptibility of pancreatic cancer cells to exogenous inducers of apoptosis. In vivo results show that FHIT gene transfer delays tumor growth and prolongs survival in a murine model mimicking human disease.

Journal Article
TL;DR: The functional domains present within C LLD7 and CLLD8 suggest that the proteins may be involved in critical cellular processes such as cell cycle and transcriptional control and could therefore be directly or indirectly involved in leukemogenesis.
Abstract: Chromosome 13q14 deletions constitute the most common structural aberration in B-cell chronic lymphocytic leukemia (B-CLL). We constructed a high-resolution physical map covering the critical deleted region in B-CLL at 13q14 and flanking sequences. The order and position of both genomic markers and known genes were determined precisely. Three novel genes, CLLD6, CLLD7, and CLLD8, were isolated and characterized. The predicted protein sequence of CLLD6 revealed no homology with known proteins. However, both CLLD7 and CLLD8 predicted proteins contain known functional domains. CLLD7 has both an RCC1 and a BTB domain, and could thus be involved in cell cycle regulation by chromatin remodeling. CLLD8 contains a methyl-CpG binding, a preSET and a SET domain, suggesting that CLLD8 might be associated with methylation-mediated transcriptional repression. Mutation analysis of hematopoietic tumor cell lines and B-CLL tumor samples revealed no point mutations within the coding region of these three novel genes. The functional domains present within CLLD7 and CLLD8 suggest that the proteins may be involved in critical cellular processes such as cell cycle and transcriptional control and could therefore be directly or indirectly involved in leukemogenesis.

Journal ArticleDOI
TL;DR: It is reported that HMGI-C/T TG mice develop natural killer (NK)-T/NK cell lymphomas starting from 12 months of age, and it is demonstrated that HM GI-C-T protein positively regulates their expression in vitro.
Abstract: Rearrangements of the high mobility group protein I-C (HMGI-C) gene, consisting in the loss of the carboxyl-terminal tail, have been frequently detected in benign human tumors of mesenchymal origin. We have previously demonstrated that transgenic (TG) mice carrying a truncated HMGI-C construct (HMGI-C/T) exhibit a giant phenotype together with a predominantly abdominal/pelvic lipomatosis. Here, we report that HMGI-C/T TG mice develop natural killer (NK)-T/NK cell lymphomas starting from 12 months of age. We found an increased expression of IL-2 and IL-15 proteins and their receptors in these lymphomas, and we demonstrate that HMGI-C/T protein positively regulates their expression in vitro. Therefore, the HMGI-C/T-mediated chronic stimulation of the IL-2/IL-15 pathway could be responsible for the onset of NK-T/NK cell lymphomas in HMGI-C/T TG mice.

Journal ArticleDOI
10 Sep 2001-Oncogene
TL;DR: Functional analysis of TCL1 revealed its involvement in a PI3-kinase dependent Akt (PKB) pro-survival pathway through its interaction with the Akt kinase which increases Akt's enzymatic activity and promotes translocation of Akt to the nucleus.
Abstract: The TCL1 locus on human chromosome 14q32.1 is activated in T-cell leukemias by translocations and inversions that juxtapose it to regulatory elements of T-cell receptor genes. We isolated and characterized four genes at this locus, TCL1 and TCL1b (T-cell leukemia/lymphoma 1 and 1b), and TNG1 and TNG2 (TCL neighboring genes 1 and 2) all of which are overexpressed following rearrangements involving 14q32.1. TCL1 and TCL1b show 60% similarity and are represented in the mouse by a cluster of six homologous genes. In humans TCL1 and TCL1b show similar expression patterns: They are expressed mainly in CD4-/CD8- immature T-cells, pre B-cells and virgin B-cells. Expression decreases significantly at more mature stages of B-cell development. Activation of TCL1 and/or TCL1b in mature T-cells causes T-cell leukemia in humans. The oncogenic nature of TCL1 was confirmed by the analysis of a transgenic mouse model. Functional analysis of Tcl1 revealed its involvement in a PI3-kinase dependent Akt (PKB) pro-survival pathway through its interaction with the Akt kinase which increases Akt's enzymatic activity and promotes translocation of Akt to the nucleus.

Journal ArticleDOI
TL;DR: Comparison of orthologous fragile regions has identified highly conserved sequences with possible functional roles in maintenance of fragility in the human and mouse FRA3B and murine Fra14A2 locus.
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.

Journal ArticleDOI
14 Nov 2001-JAMA
TL;DR: Functional analysis of the Tcl1 protein revealed its involvement in an Akt (protein kinase B) prosurvival pathway through its interaction with the Akt kinase, which promotes translocation of Akt to the nucleus and increases Akt's enzymatic activity.
Abstract: T-cell chronic lymphocytic/prolymphocytic leukemia (T-CLL/T-PLL) is a lymphoproliferative disease derived from immunocompetent post-thymic T cells. Activation (initiation of expression) of the TCL1 locus at chromosome 14q32.1 appears to be the causal event in the pathogenesis of these mature T-cell leukemias. This activation occurs as a result of translocations or inversions that cause rearrangement of the TCL1 (T-cell leukemia/lymphoma 1) locus with regulatory elements of T-cell receptor genes. To describe the molecular events that take part in the leukemogenesis of mature T-cell leukemias, we reviewed the literature and our own data on the molecular basis of mature T-cell leukemia. This data search revealed that 4 genes have been identified at the TCL1 locus: TCL1, TCL1b, TNG1, and TNG2. The expression of these genes is substantially increased following rearrangements involving 14q32.1. Functional analysis of the Tcl1 protein revealed its involvement in an Akt (protein kinase B) prosurvival pathway through its interaction with the Akt kinase, which promotes translocation of Akt to the nucleus and increases Akt's enzymatic activity. The available data provide important insights into the molecular mechanisms of T-cell leukemogenesis that may lead to the development of new drugs for treatment of mature T-cell leukemia.

Journal ArticleDOI
21 Nov 2001-JAMA
TL;DR: Findings show that FHit gene therapy may potentially be clinically useful for treatment of cancer and also prevention of carcinogen-induced tumor development, suggesting a rationale for further research involving FHIT introduction.
Abstract: ContextThe fragile histidine triad gene (FHIT) encompasses a human common fragile site, FRA3B, that is susceptible to environmental carcinogens. Deletion and inactivation of FHIT have been seen in a number of human premalignant and malignant lesions.ObjectiveTo review and evaluate preclinical studies of cancer therapy using the FHIT tumor suppressor gene and related studies involving Fhit protein expression.Data SourcesA MEDLINE search of articles published from 1996 to June 2001 was performed; article reference lists were used to retrieve additional relevant articles.Study SelectionImmunohistochemical studies of primary tumors or relevant lesions were selected to evaluate Fhit expression in premalignant or malignant stages. Preclinical studies on antitumorigenic or therapeutic introduction of FHIT were reviewed for the effects of exogenous Fhit expression. For the immunohistochemical analyses, 26 studies were included that analyzed at least 15 cases of a single type of tumor. For precancerous lesions, 9 studies were included that analyzed at least 4 cases. For studies of FHIT introduction, 9 published studies were included.Data ExtractionUsing primary data from each of the studies, we assessed the rationale and potential contribution of FHIT cancer therapy. Data was independently abstracted by 2 authors and study quality was assessed by 2 other authors.Data SynthesisOverall, 60% (1162/1948 cases) of primary tumors showed absent or markedly reduced Fhit protein expression in cancer cells. Studies of preneoplastic lesions or early-stage cancer showed absence or marked reduction of Fhit protein expression in 0% to 93% of samples (overall, 31% [127/408 cases]). Preclinical studies using 26 cancer-derived cell lines from human lung, head and neck, esophageal, gastric, cervical, pancreatic, and kidney cancers, showed that reintroduction of FHIT resulted in inhibition of in vitro tumor cell growth or of in vivo tumorigenicity in 17 (57%) of 30 cell line experiments. Model systems for human preventive cancer therapy suggested that oral introduction of viral vector-mediated FHIT into Fhit-deficient mice may prevent carcinogen-induced tumor development in some cases.ConclusionThese findings show that FHIT gene therapy may potentially be clinically useful for treatment of cancer and also prevention of carcinogen-induced tumor development, suggesting a rationale for further research involving FHIT introduction.

Patent
15 May 2001
TL;DR: In this article, the structure of the NitFhit fusion protein and interactions between the Nit and Fhit polypeptides are defined, which relates to the identification of small molecules that interact with, and regulate, the Nit protein.
Abstract: In invertebrates, Fhit is encoded as a fusion protein with Nit. Outside of invertebrates, Nit homologs are found as separate polypeptides in organisms with Fhit homologs. Therefore, Nit and Fhit are expected to interact physically and function in the same cellular pathway. The structure of the NitFhit fusion protein and interactions between the Nit and Fhit polypeptides are defined. The present invention relates to the identification of small molecules that interact with, and regulate, the Nit protein. The present invention further relates to therapeutic compositions and their uses in regulating Nit activity, thereby modulating cellular proliferation.

Journal Article
TL;DR: The expression of the Fhit protein in relation to human papillomavirus (HPV) subtype, the type of the intraepithelial lesion, HIV-induced immunodeficiency, and the DNA content correlates with the biological behavior of the lesions.
Abstract: Purpose: The tumor suppressor gene, FHIT , has been cloned and mapped at chromosome region 3p14.2, one of the regions most frequently deleted in cervical carcinoma. In this report, we show that the expression of the Fhit protein in relation to human papillomavirus (HPV) subtype, the type of the intraepithelial lesion, HIV-induced immunodeficiency, and the DNA content (ploidy) correlates with the biological behavior of the lesions. Experimental Design: To investigate involvement of the FHIT gene in squamous intraepithelial lesions of low and high grade (LGSILs and HGSILs, respectively ) of the uterine cervix, we examined the Fhit protein expression by immunocytochemistry in 131 cervical smears of 96 HIV-seropositive patients (42 with LGSILs and 10 with HGSILs) and 35 HIV-seronegative (5 with LGSILs) persons. Results: Fhit protein was detected in normal cells, whereas dysplastic cells (independently of HPV infection and HPV subtypes) showed reduced expression of Fhit ( P = 0.00001). Lesions from 52 HIV-seropositive patients, 42 LGSILs and 10 HGSILs, showed diploid DNA content in 63.5%, aneuploid in 32.7%, and polyploid in 3.8%, but 90% of the HGSILs showed an aneuploid DNA content, and all were infected by HPV 16/18 subtypes. 23.8% of LGSIL cases were associated with HPV 16. Conclusions: These data clearly suggest that loss of Fhit expression occurs in the early stages of cervical carcinogenesis. Pap test represents one of the most convenient and rapid procedures available in identification of cellular changes; hence, Fhit staining might be used as an useful tool in larger population screening to detect early alteration in cellular behaviors.

Journal ArticleDOI
TL;DR: It is proposed that TCL1 expression especially in high-grade B cell non-Hodgkin’s lymphomas might interfere with B cell differentiation and promote the transition from low- to high- grade lymphoma.
Abstract: Objective:TCL1, MTCP1 and TCL1b are three members of a new family of oncogenes that are expressed in T cell leukemias of ataxia telangiectasia patients (T-PLL, T-CLL). TCL1 is located at 14q32.1 and activated by juxtaposition to the α/δ-locus at 14q11 or β-locus at 7q35 of the T cell receptor during the reciprocal translocations t(14;14)(q11;q32), t(7;14)(q35;q32), or inversion inv(14)(q11;q32). TCL1 encodes a predominantly cytoplasmic protein of 114 aa (14 kD) of unknown function. Recent studies suggest that TCL1 promotes cell survival rather than stimulating cell proliferation, as previously proposed. Methods: In an attempt to clarify the contexts in which TCL1 is expressed, we investigated TCL1 expression in 114 lymphoma and leukemia patients by Northern blot, RT-PCR and immunohistochemistry. Results:TCL1 expression is restricted to lymphoid cells, and is found in neoplastic (T and B cell neoplasms, and Hodgkin’s disease) and nonneoplastic proliferations (reactive lesions). Out of 114 cases, 18 neoplasms of myeloid and 4 cases of epithelial origin were TCL1-negative. In lesions of the lymphoid system, both low- and high-grade lymphomas were found to express TCL1. Conclusions: We propose that TCL1 expression especially in high-grade B cell non-Hodgkin’s lymphomas might interfere with B cell differentiation and promote the transition from low- to high-grade lymphoma.

Journal ArticleDOI
TL;DR: Most human tumors carry alterations in multiple genes affecting cell proliferation, cell survival, and genomic stability, so the process often is initiated by the loss of function of a tumor suppressor gene.
Abstract: Cancer is a genetic disease caused by a multistep process involving activation of oncogenes, loss of function of tumor suppressor genes, and alterations of modifier genes, for example, genes involved in DNA repair and genomic stability. Thus, most human tumors carry alterations in multiple genes affecting cell proliferation, cell survival, and genomic stability. In malignant diseases of the hematopoietic system and in soft tissue sarcomas, the process often is initiated by the activation of an oncogene, for example c-MYC in Burkitt lymphoma (1) or BCL2 in follicular lymphoma (2). In solid tumors the process often is initiated by the loss of function of a tumor suppressor gene (3).

Journal ArticleDOI
TL;DR: It is confirmed that FHIT is aberrantly expressed in ALL, but suggests it does not have a role as a prognostic factor, and studies with large numbers of patients and evaluation of the mechanisms of FHit function in ALL are needed.

Journal Article
TL;DR: After N-nitrosomethylbenzylamine treatment, the spectrum of tumors developed by the Fhit-deficient mice was similar to those observed in a familial skin cancer condition, the Muir-Torre syndrome, although there is no clear evidence yet for a relationship of FHIT and the human syndrome.
Abstract: Five years ago the fragile histidine triad (FHIT) gene including the most common fragile site locus of the human genome, FRA3B, was identified. The gene is altered in many types of cancer and several data support the idea that FHIT has to be considered a tumor suppressor. FHIT abnormalities were investigated in some skin tumors. Fifty-seven per cent of Merkel cell carcinomas displayed abnormal FHIT products but the involvement of FHIT in human non-melanoma skin cancer is still unclear. Because the murine Fhit locus is similar to its human homologue and is altered in cancer cell lines, we have established a strain of Fhit-deficient mice. After N-nitrosomethylbenzylamine treatment, the spectrum of tumors developed by the Fhit-deficient mice was similar to those observed in a familial skin cancer condition, the Muir-Torre syndrome, although there is no clear evidence yet for a relationship of FHIT and the human syndrome. Because cancer cells lacking in FHIT are defective in apoptosis, we propose the Fhit-deficient mouse as a model to understand a possible proapoptotic mechanism deficiency in the human syndrome.

Journal ArticleDOI
TL;DR: The crystal structure of a dimer of murine Tcl1 has been determined and the molecular surfaces of all three structures showed a common planar region which is likely to be involved in protein-protein interactions.
Abstract: Tcl1 and Mtcp1, members of the Tcl1 family, are implicated in T-cell prolymphocytic leukemia. The crystal structure of a dimer of murine Tcl1 has been determined at 2.5 A resolution with an R factor of 0.225. Murine Tcl1, human Tcl1 and Mtcp1 share very similar subunit structures, with RMS differences of 0.6 and 1.4 A for Cα atoms, respectively, while the sequences share 50 and 36% identity, respectively. These structures fold into an eight-stranded β-barrel of unique topology and high internal symmetry of 1.1–1.3 A for the two halves of human and murine Tcl1 and 1.7 A for Mtcp1, despite the low 12–13% sequence identity. The molecular surfaces of all three structures showed a common planar region which is likely to be involved in protein–protein interactions.

Journal ArticleDOI
TL;DR: To elucidate the possible role of FHIT as a tumor suppressor in breast cancer and to identify its mechanisms, Fhit protein-negative breast cancer cell lines lacking endogenous protein expression were stable transfected with Fhit cDNA and showed no alteration in cell morphology and in in vitro anchorage-dependent and independent proliferation.
Abstract: The gene FHIT, encompassing the FRA3B fragile site, is located in a region of chromosome 3p14.2 that is often deleted in several types of epithelial cancers and, therefore, it has been investigated as a candidate tumor suppressor. In breast cancer inactivation of FHIT occurred in 70% of the patients, and it is caused by both alterations in the regulation of Fhit expression and by deletions of the gene. Moreover, analysis of 500 cases of breast carcinomas with 20 years of follow up demonstrated that loss of Fhit protein is associated with high proliferative, large and undifferentiated tumors, even though Fhit is not a prognostic factor. In order to elucidate the possible role of FHIT as a tumor suppressor in breast cancer and to identify its mechanisms, Fhit protein-negative breast cancer cell lines lacking endogenous protein expression were stable transfected with FHIT cDNA. Stable transfectant clones showed no alteration in cell morphology and in in vitro anchorage-dependent and independent proliferation. A significant delay in the tumor growth in nude mice was observed for some Fhit-positive clones; in an additional case the outgrowing of the tumor was due to loss of Fhit expression in vivo. Interestingly, some Fhit-positive clones are able to develop tumors in vivo despite high stable Fhit expression, prompting us to investigate the different mechanisms between the two types of Fhit-expressing clones. Supported by AIRC.