Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
2.
Nat Genet ; 50(2): 219-228, 2018 02.
Article in English | MEDLINE | ID: mdl-29335542

ABSTRACT

The mechanisms by which mitochondrial metabolism supports cancer anabolism remain unclear. Here, we found that genetic and pharmacological inactivation of pyruvate dehydrogenase A1 (PDHA1), a subunit of the pyruvate dehydrogenase complex (PDC), inhibits prostate cancer development in mouse and human xenograft tumor models by affecting lipid biosynthesis. Mechanistically, we show that in prostate cancer, PDC localizes in both the mitochondria and the nucleus. Whereas nuclear PDC controls the expression of sterol regulatory element-binding transcription factor (SREBF)-target genes by mediating histone acetylation, mitochondrial PDC provides cytosolic citrate for lipid synthesis in a coordinated manner, thereby sustaining anabolism. Additionally, we found that PDHA1 and the PDC activator pyruvate dehydrogenase phosphatase 1 (PDP1) are frequently amplified and overexpressed at both the gene and protein levels in prostate tumors. Together, these findings demonstrate that both mitochondrial and nuclear PDC sustain prostate tumorigenesis by controlling lipid biosynthesis, thus suggesting this complex as a potential target for cancer therapy.


Subject(s)
Cell Compartmentation/physiology , Lipogenesis , Prostatic Neoplasms/metabolism , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase Complex/physiology , Animals , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cells, Cultured , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoplasm/pathology , Humans , Lipogenesis/genetics , Male , Mice , Mice, Knockout , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Pyruvate Dehydrogenase Complex/metabolism
3.
Swiss Med Wkly ; 147: w14412, 2017.
Article in English | MEDLINE | ID: mdl-28322421

ABSTRACT

QUESTIONS UNDER STUDY: This pilot study aimed to assess the feasibility, acceptance and costs of an ultrasound scan screening programme for abdominal aortic aneurysms (AAA) in the elderly male population resident in Canton Ticino, Switzerland. METHODS: The target population were male patients aged 65-80 years who attended the outpatient clinics of the Lugano Regional Hospital in 2013. The patients showing interest were contacted by phone to verify their eligibility and fix the appointment for the ultrasound scan of the abdominal aorta. Patients with recent examinations suitable for AAA detection were excluded. Aneurysm was defined as an abdominal aorta with sagittal and/or axial diameter  30 mm. Patients' characteristics and study results were presented as descriptive statistics. The chi-squared test was used to compare categorical variables with p <0.05 as a statistical significance threshold. RESULTS: 1634 patients received the screening information leaflet and 745 (45.6%) underwent the ultrasound scan. Among the 1091 eligible patients, the acceptance rate was 68.3%. A previously unknown AAA was diagnosed in 31 patients (4.2%, 95% confidence interval 2.8-5.9%). Age and area of residence had a statistically significant impact on patient's acceptance rate (p <0.05). The mean cost per screened patient was CHF 88. CONCLUSIONS: AAA screening of male patients aged 65-80 years is feasible with limited financial and organisational effort. Adherence might be improved by a larger community-based programme and involvement of general practitioners.


Subject(s)
Aortic Aneurysm, Abdominal/diagnosis , Mass Screening/methods , Ultrasonography/methods , Aged , Aged, 80 and over , Aortic Aneurysm, Abdominal/diagnostic imaging , Humans , Male , Pilot Projects , Prospective Studies , Risk Factors , Switzerland , Time Factors
4.
Nat Commun ; 6: 7227, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26085373

ABSTRACT

Enhancement of cellular senescence in tumours triggers a stable cell growth arrest and activation of an antitumour immune response that can be exploited for cancer therapy. Currently, there are only a limited number of targeted therapies that act by increasing senescence in cancers, but the majority of them are not selective and also target healthy cells. Here we developed a chemogenomic screening to identify compounds that enhance senescence in PTEN-deficient cells without affecting normal cells. By using this approach, we identified casein kinase 2 (CK2) as a pro-senescent target. Mechanistically, we show that Pten loss increases CK2 levels by activating STAT3. CK2 upregulation in Pten null tumours affects the stability of Pml, an essential regulator of senescence. However, CK2 inhibition stabilizes Pml levels enhancing senescence in Pten null tumours. Taken together, our screening strategy has identified a novel STAT3-CK2-PML network that can be targeted for pro-senescence therapy for cancer.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Cellular Senescence/drug effects , Molecular Targeted Therapy , Naphthyridines/therapeutic use , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/drug therapy , Animals , Casein Kinase II/metabolism , Drug Evaluation, Preclinical , Female , HCT116 Cells , Humans , Male , Mice, Transgenic , Naphthyridines/pharmacology , Nuclear Proteins/metabolism , Phenazines , Promyelocytic Leukemia Protein , RNA, Small Interfering , STAT3 Transcription Factor/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
5.
Cell Rep ; 11(4): 564-76, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25892239

ABSTRACT

Understanding the molecular pathways that contribute to the aggressive behavior of HER2-positive breast cancers may aid in the development of novel therapeutic interventions. Here, we show that CDCP1 and HER2 are frequently co-overexpressed in metastatic breast tumors and associated with poor patient prognosis. HER2 and CDCP1 co-overexpression leads to increased transformation ability, cell migration, and tumor formation in vivo, and enhanced HER2 activation and downstream signaling in different breast cancer cell lines. Mechanistically, we demonstrate that CDCP1 binds to HER2 through its intracellular domain, thereby increasing HER2 interaction with the non-receptor tyrosine kinase c-SRC (SRC), leading to trastuzumab resistance. Taken together, our findings establish that CDCP1 is a modulator of HER2 signaling and a biomarker for the stratification of breast cancer patients with poor prognosis. Our results also provide a rationale for therapeutic targeting of CDCP1 in HER2-positive breast cancer patients.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/metabolism , Carcinogenesis/metabolism , Cell Adhesion Molecules/metabolism , Drug Resistance, Neoplasm , Neoplasm Proteins/metabolism , Receptor, ErbB-2/metabolism , Animals , Antigens, CD/genetics , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Carcinogenesis/genetics , Cell Adhesion Molecules/genetics , Female , Humans , MCF-7 Cells , Mice , Neoplasm Proteins/genetics , Protein Binding , Receptor, ErbB-2/genetics , Trastuzumab/pharmacology , src-Family Kinases/metabolism
6.
Cell Rep ; 9(1): 75-89, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25263564

ABSTRACT

Prosenescence therapy has recently emerged as a novel therapeutic approach for treating cancer. However, this concept is challenged by conflicting evidence showing that the senescence-associated secretory phenotype (SASP) of senescent tumor cells can have pro- as well as antitumorigenic effects. Herein, we report that, in Pten-null senescent tumors, activation of the Jak2/Stat3 pathway establishes an immunosuppressive tumor microenvironment that contributes to tumor growth and chemoresistance. Activation of the Jak2/Stat3 pathway in Pten-null tumors is sustained by the downregulation of the protein tyrosine phosphatase PTPN11/SHP2, providing evidence for the existence of a novel PTEN/SHP2 axis. Importantly, treatment with docetaxel in combination with a JAK2 inhibitor reprograms the SASP and improves the efficacy of docetaxel-induced senescence by triggering a strong antitumor immune response in Pten-null tumors. Altogether, these data demonstrate that immune surveillance of senescent tumor cells can be suppressed in specific genetic backgrounds but also evoked by pharmacological treatments.


Subject(s)
Antineoplastic Agents/pharmacology , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/immunology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Animals , Cellular Senescence/immunology , Cytokines/immunology , Docetaxel , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Signal Transduction , Taxoids/pharmacology , Tumor Microenvironment
7.
Evid Based Med ; 19(5): 163-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25165158

ABSTRACT

UNLABELLED: To assess and quantify the impact of the literature in diagnostic decisions and treatment of patients admitted to an internal medicine service using the methodology of evidence-based medicine. From November 2012 to February 2013, patients who were hospitalised in the internal medicine service of Regional Hospital of Lugano (Switzerland) and generated questions on medical care were randomly assigned to two groups: an 'intervention group' (supported by the literature research) and a 'control group' (not supported by the literature research). The information obtained from the literature was submitted by email to all members of the medical team within 12 h after asking the question. Two hundred and one participants, from 866 patients hospitalised in the analysed period, divided into intervention (n=101) and control (n=100) groups, generated questions. In the intervention group, bibliographical research was possible for 98 participants. The medical team accepted the results and implemented the research for 90.8% of these participants (89/98). Statistical analyses were carried out on the intention-to-treat and on the per-protocol populations. Bibliographical research had a significant protective effect on transfer to an intensive care unit (relative risk (RR)=0.30; 95% CI 0.10 to 0.90; χ²=5.3, p=0.02) and hospital readmissions were also influenced by bibliographical research (RR=0.42; 95% CI 0.17 to 1.0; χ²=3.36, p=0.05) in the intention-to-treat population. Our results point out the importance of bibliographical support on the quality of medical care. In particular, they show its possible impact on clinical outcome. TRIAL REGISTRATION NUMBER: EOC Registry (registration number: 14-055).


Subject(s)
Evidence-Based Medicine , Hospitalization , Internal Medicine/standards , Outcome and Process Assessment, Health Care , Quality of Health Care , Biomedical Research , Humans , Information Dissemination , Prospective Studies
8.
Nature ; 515(7525): 134-7, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25156255

ABSTRACT

Aberrant activation of oncogenes or loss of tumour suppressor genes opposes malignant transformation by triggering a stable arrest in cell growth, which is termed cellular senescence. This process is finely tuned by both cell-autonomous and non-cell-autonomous mechanisms that regulate the entry of tumour cells to senescence. Whether tumour-infiltrating immune cells can oppose senescence is unknown. Here we show that at the onset of senescence, PTEN null prostate tumours in mice are massively infiltrated by a population of CD11b(+)Gr-1(+) myeloid cells that protect a fraction of proliferating tumour cells from senescence, thus sustaining tumour growth. Mechanistically, we found that Gr-1(+) cells antagonize senescence in a paracrine manner by interfering with the senescence-associated secretory phenotype of the tumour through the secretion of interleukin-1 receptor antagonist (IL-1RA). Strikingly, Pten-loss-induced cellular senescence was enhanced in vivo when Il1ra knockout myeloid cells were adoptively transferred to PTEN null mice. Therapeutically, docetaxel-induced senescence and efficacy were higher in PTEN null tumours when the percentage of tumour-infiltrating CD11b(+)Gr-1(+) myeloid cells was reduced using an antagonist of CXC chemokine receptor 2 (CXCR2). Taken together, our findings identify a novel non-cell-autonomous network, established by innate immunity, that controls senescence evasion and chemoresistance. Targeting this network provides novel opportunities for cancer therapy.


Subject(s)
Cell Movement , Cellular Senescence , Myeloid Cells/cytology , Myeloid Cells/metabolism , Prostatic Neoplasms/pathology , Receptors, Chemokine/metabolism , Animals , Cellular Senescence/drug effects , Disease Progression , Docetaxel , Drug Resistance, Neoplasm , Humans , Immunity, Innate , Interleukin 1 Receptor Antagonist Protein/deficiency , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1alpha/immunology , Interleukin-1alpha/metabolism , Male , Mice , Myeloid Cells/transplantation , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Receptors, Interleukin-8B/antagonists & inhibitors , Taxoids/pharmacology , Tumor Escape , Tumor Microenvironment
9.
Cancer Res ; 73(22): 6816-27, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24063893

ABSTRACT

Several studies link disease progression, recurrence, and treatment failures to the cancer stem-like cell (CSC) subpopulation within the heterogeneous tumor cell population. Myc is a transcription factor having a central function in stem cell biology and in human cancers. Hence, Myc represents an attractive target to develop CSC-specific therapies. Recent findings suggest that Myc transcription can be silenced using an RNA interference (RNAi)-based strategy that targets noncoding promoter-associated RNA (paRNA) overlapping the transcription start site. In this study, we investigated the effects of silencing Myc transcription on prostate CSC in cell culture and xenograft models of human prostate cancer. Treatment with an effective promoter-targeting siRNA reduced the fraction of CSCs, leading to reduced self-renewal, tumor-initiating, and metastatic capability. Combined analysis of stem-like cells and senescence markers indicated that Myc silencing triggered a phenotypic shift and senescence in the CSC subpopulation. Notably, systemic delivery of the promoter-targeting siRNA in the xenograft model produced a striking suppression in the development of prostate tumors. Our results support a pivotal role for Myc in CSC maintenance and show that Myc targeting via RNAi-based transcriptional silencing can trigger CSC senescence and loss of their tumor-initiating capability. More generally, our findings demonstrate the efficacy of RNAi-based transcriptional strategies and the potential to target regulatory noncoding paRNAs for therapeutic applications.


Subject(s)
Carcinoma/pathology , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/pathology , RNA, Small Interfering/pharmacology , Animals , Carcinoma/genetics , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cellular Senescence/drug effects , Cellular Senescence/genetics , Genes, myc/drug effects , Humans , Male , Mice , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/genetics , RNA Interference , Transcription, Genetic/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Oncol Rep ; 30(2): 824-32, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23715752

ABSTRACT

Testin (TES) is a putative tumour-suppressor gene downregulated in various types of cancers. Survivin is a nodal protein involved in multiple signalling pathways, tumour maintenance and inhibition of apoptosis. Previous studies indicate that TES and survivin can functionally interact and modulate cell death and proliferation in breast cancer cells. The aim of the present study was to investigate the expression and prognostic relevance of TES and survivin in breast cancer subtypes examining a large cohort of breast cancer patients. We determined the expression of TES and survivin by immunohistochemistry (IHC) in tissue samples from 242 breast cancer patients diagnosed between 1981 and 2009. The expression of these proteins was compared with clinical and pathological data. There was a significant association of nuclear survivin overexpression and TES downregulation with triple-negative tumours [P=0.009; univariate odds ratio (OR), 3.20; 95% CI, 1.34-7.66] (P=0.018; multivariate OR, 2.90; 95% CI, 1.20­6.97). A further significant correlation was observed between TES downregulation and the luminal B subtype (P=0.019, univariate OR: 2.90; 95% CI, 1.19­7.06) (P=0.032, multivariate OR, 2.67; 95% CI, 1.09-6.65), independent of survivin expression. Our results demonstrated a statistically significant association between TES downregulation and highly aggressive breast tumour subtypes, such as triple-negative and luminal B tumours, along with the prognostic relevance of nuclear expression of survivin. To our knowledge, this is the first demonstration of such an association.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cytoskeletal Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/biosynthesis , LIM Domain Proteins/biosynthesis , Breast Neoplasms/genetics , Cytoskeletal Proteins/genetics , Down-Regulation , Female , Gene Expression , Humans , Inhibitor of Apoptosis Proteins/genetics , LIM Domain Proteins/genetics , Middle Aged , Prognosis , RNA-Binding Proteins , Survivin
11.
Cancer Res ; 73(14): 4533-47, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23687337

ABSTRACT

Chromosomal translocations leading to deregulated expression of ETS transcription factors are frequent in prostate tumors. Here, we report a novel mechanism leading to oncogenic activation of the ETS factor ESE1/ELF3 in prostate tumors. ESE1/ELF3 was overexpressed in human primary and metastatic tumors. It mediated transforming phenotypes in vitro and in vivo and induced an inflammatory transcriptome with changes in relevant oncogenic pathways. ESE1/ELF3 was induced by interleukin (IL)-1ß through NF-κB and was a crucial mediator of the phenotypic and transcriptional changes induced by IL-1ß in prostate cancer cells. This linkage was mediated by interaction of ESE1/ELF3 with the NF-κB subunits p65 and p50, acting by enhancing their nuclear translocation and transcriptional activity and by inducing p50 transcription. Supporting these findings, gene expression profiling revealed an enrichment of NF-κB effector functions in prostate cancer cells or tumors expressing high levels of ESE1/ELF3. We observed concordant upregulation of ESE1/ELF3 and NF-κB in human prostate tumors that was associated with adverse prognosis. Collectively, our results define an important new mechanistic link between inflammatory signaling and the progression of prostate cancer.


Subject(s)
DNA-Binding Proteins/metabolism , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Disease Progression , Gene Expression , Gene Knockdown Techniques , Heterografts , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/genetics , Prognosis , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation , Transcriptome
12.
Cancer Res ; 72(11): 2889-900, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22505649

ABSTRACT

Cancer stem cells (CSC) play a significant role in tumor progression, disease recurrence, and treatment failure. Here, we show that the endogenously expressed ETS transcription factor ESE3/EHF controls prostate epithelial cell differentiation and stem-like potential. We found that loss of ESE3/EHF induced epithelial-to-mesenchymal transition (EMT), stem-like features, and tumor-initiating and metastatic properties in prostate epithelial cells, and reexpression of ESE3/EHF inhibited the stem-like properties and tumorigenic potential of prostate cancer cells. Mechanistically, ESE3/EHF repressed the expression of key EMT and CSC genes, including TWIST1, ZEB2, BMI1, and POU5F1. Analysis of human tissue microarrays showed that reduced ESE3/EHF expression is an early event in tumorigenesis, frequently occurring independently of other ETS gene alterations. Additional analyses linked loss of ESE3/EHF expression to a distinct group of prostate tumors with distinctive molecular and biologic characteristics, including increased expression of EMT and CSC genes. Low ESE3/EHF expression was also associated with increased biochemical recurrence of prostate cancer and reduced overall survival after prostatectomy. Collectively, our findings define a key role for ESE3/EHF in the development of a subset of prostate tumors and highlight the clinical importance of identifying molecularly defined tumor subgroups.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Transcription Factors/physiology , Animals , Gene Expression Regulation , Humans , Male , Mice , Trans-Activators/analysis , Transcriptional Regulator ERG
13.
Clin Cancer Res ; 11(2 Pt 1): 806-13, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15701871

ABSTRACT

PURPOSE: The human TESTIN (TES) gene is a putative tumor suppressor gene in the fragile chromosomal region FRA7G at 7q31.1/2 that was reported to be altered in leukemia and lymphoma cell lines. In this report, we investigated the effect of TES gene expression in vivo to evaluate a possible role of TES gene in human cancer. EXPERIMENTAL DESIGN: We have analyzed the expression of TES gene in a panel of 25 breast tumors and 17 cell lines of breast, colon, and uterine cancers. Furthermore, to evaluate the potential of TES gene therapy, we studied the effects of adenoviral TES transduction (Ad-TES) in cell lines with undetectable TES expression (T47D and MES-SA) as well as in MCF-7 cell line where TES expression is normal. RESULTS: Twenty-five percent of primary breast tumor samples as well as the breast cancer cell line T47D and the uterine sarcoma cell line MES-SA were negative or displayed low levels of TES. After TES restoration by Ad-TES transduction, T47D and MES-SA cell lines underwent apoptosis. Furthermore, TES expression significantly reduced the tumorigenic potential of both T47D and MES-SA in nude mice, whereas the untreated cells and Ad-GFP-infected cells showed tumor growth in vivo. The TES-positive cell line control (MCF-7) was not affected by TES expression and did not show a reduction of tumorigenicity in nude mice after infection with Ad-TES. CONCLUSIONS: Ad-TES expression inhibit the growth of breast and uterine cancer cells lacking of TES expression through caspase-dependent and caspase-independent apoptosis, respectively, suggesting that Ad-TES infection should be explored as a therapeutic strategy.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/therapy , Colonic Neoplasms/therapy , Homeodomain Proteins/therapeutic use , Transduction, Genetic , Tumor Suppressor Proteins/therapeutic use , Uterine Neoplasms/therapy , Adenoviridae/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Caspases/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cytoskeletal Proteins , Female , Genetic Vectors , Green Fluorescent Proteins , Humans , LIM Domain Proteins , Mice , Mice, Nude , RNA-Binding Proteins , Sarcoma, Experimental/genetics , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/therapy , Tumor Cells, Cultured , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Xenograft Model Antitumor Assays
14.
Cancer Gene Ther ; 11(9): 633-41, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15272314

ABSTRACT

Pancreatic carcinoma is one of the most aggressive tumors, and, being refractory to conventional therapies, is an excellent target for new therapeutic approaches. Based on our previous finding of high HMGA1 expression in pancreatic cancer cells compared to normal pancreatic tissue, we evaluated whether suppression of HMGA1 protein expression could be a treatment option for patients affected by pancreatic cancer. Here we report that HMGA1 proteins are overexpressed in pancreatic carcinoma cell lines, and their downregulation through an adenovirus carrying the HMGA1 gene in an antisense orientation (Ad Yas-GFP) results in the death of three human pancreatic carcinoma cell lines (PANC1, Hs766T and PSN1). Pretreatment of PANC1 and PSN1 cells with Ad Yas-GFP suppressed and reduced, respectively, their ability to form xenograft tumors in nude mice. To further verify the role of HMGA1 in pancreatic tumorigenesis, we used a HMGA1 antisense phosphorothioate oligodeoxynucleotide (ODN); its addition induced a decrease in HMGA1 protein levels and a significant reduction of the proliferation rate of PANC1-, Hs766T- and PSN1-treated cells. Therefore, suppression of HMGA1 protein synthesis by an HMGA1 antisense approach seems to be a feasible treatment strategy in pancreatic carcinomas.


Subject(s)
Apoptosis , Cell Proliferation/drug effects , HMGA1a Protein/antagonists & inhibitors , Models, Animal , Pancreatic Neoplasms/therapy , Adenoviridae/genetics , Animals , Down-Regulation , Green Fluorescent Proteins/metabolism , HMGA1a Protein/genetics , HMGA1a Protein/metabolism , Humans , Male , Mice , Mice, Nude , Oligonucleotides, Antisense/pharmacology , Pancreatic Neoplasms/genetics , Transplantation, Heterologous , Tumor Cells, Cultured
15.
Clin Cancer Res ; 10(8): 2720-4, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15102676

ABSTRACT

PURPOSE: Parkin, a gene mutated in autosomal recessive juvenile Parkinsonism and mapped to the common fragile site FRA6E on human chromosome 6q25-q27, is associated with a frequent loss of heterozygosity and altered expression in breast and ovarian carcinomas. In addition, homozygous deletions of exon 2 creating deleterious truncations of the Parkin transcript were observed in the lung adenocarcinoma cell lines Calu-3 and H-1573, suggesting that the loss of this locus and the resulting changes in its expression are involved in the development of these tumors. EXPERIMENTAL DESIGN: We examined 20 paired normal and non-small cell lung cancer samples for the presence of Parkin alterations in the coding sequence and changes in gene expression. We also restored gene expression in the Parkin-deficient lung carcinoma cell line H460 by use of a recombinant lentivirus containing the wild-type Parkin cDNA. RESULTS: Loss of heterozygosity analysis identified a common region of loss in the Parkin/FRA6E locus with the highest frequency for the intragenic marker D6S1599 (45%), and semi-quantitative reverse transcription-PCR revealed reduced expression in 3 of 9 (33%) lung tumors. Although we did not observe any in vitro changes in cell proliferation or cell cycle, ectopic Parkin expression had the ability to reduce in vivo tumorigenicity in nude mice. CONCLUSION: These data suggest that Parkin is a tumor suppressor gene whose inactivation may play an important role in non-small cell lung cancer tumorigenesis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle , Cell Division , Cell Line, Tumor , DNA Methylation , DNA Mutational Analysis , DNA, Complementary/metabolism , Exons , Gene Deletion , Gene Expression Regulation, Neoplastic , Homozygote , Humans , Lentivirus/genetics , Loss of Heterozygosity , Lung Neoplasms/metabolism , Mice , Mice, Nude , Microsatellite Repeats , Models, Genetic , Mutation , Neoplasm Transplantation , Polymorphism, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
16.
FASEB J ; 17(12): 1768-70, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12958204

ABSTRACT

Fhit expression is reduced in most cancers, and Fhit replacement by FHIT expression viruses in lung, esophageal, pancreatic, and cervical cancers induces apoptosis in the cancer cells. Mice carrying one or two inactivated Fhit alleles are hypersensitive to development of N-nitrosomethylbenzylamine (NMBA)-induced forestomach tumors. In the present study, we investigated the kinetics and mechanism of tumor reversal and intervention by oral delivery of FHIT expression viruses. Tumor analysis showed that: a) by 37 days post-NMBA, control mice showed approximately 7 tumors and by 84 days approximately 10 tumors/forestomach; b) mice receiving FHIT virus at 2 or 42 days post-NMBA showed significantly reduced tumor burdens; c) Fhit was still expressed at 82 days postinfection; d) control viral infection had no effect on tumor development; and e) reduced Bcl2, increased Bax expression, and increased TUNEL-positive apoptotic nuclei characterized the restored epithelia of FHIT transduced forestomachs. Thus, FHIT viral gene delivery prevents or retards development of carcinogen-induced forestomach tumors and reverses development of established tumors by 60-70% through an apoptotic pathway. This dramatic reduction in tumor burden emphasizes the efficacy of targeting the FHIT apoptotic pathway for tumor eradication.


Subject(s)
Acid Anhydride Hydrolases , Dimethylnitrosamine/analogs & derivatives , Neoplasm Proteins/genetics , Stomach Neoplasms/therapy , Administration, Oral , Animals , Apoptosis , Cell Division , Gastric Mucosa/pathology , Genetic Therapy , Genetic Vectors/administration & dosage , Mice , Models, Biological , Stomach/anatomy & histology , Stomach/pathology , Stomach Neoplasms/chemically induced , Stomach Neoplasms/pathology , Viruses/genetics
17.
Cancer Res ; 63(6): 1183-7, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12649173

ABSTRACT

The fragile histidine triad (FHIT) gene at chromosome 3p14.2 is a tumor suppressor gene that is altered mainly by deletion in a large fraction of human tumors, including breast cancers. To evaluate the potential of FHIT gene therapy in this type of cancer, we have studied the biological effects of adenoviral FHIT transduction (Ad-FHIT) in breast cancer cell lines. The results showed that, after FHIT restoration in BT-549, MDA-MB-436, and HCC1806 cells, they underwent apoptosis by activation of the intrinsic pathway. In all three cell lines infected with Ad-FHIT, we have found activation of caspase-2, which is required for permeabilization of mitochondria, release of cytochrome c, and apoptosis. Furthermore, Fhit overexpression produces alteration in cell cycling properties, as well as reduction of the tumorigenic potential in nude mice.


Subject(s)
Acid Anhydride Hydrolases , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Neoplasm Proteins/genetics , Adenoviridae/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Cycle/genetics , Cell Division/genetics , Female , Genetic Therapy/methods , Humans , Mice , Mice, Nude , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/physiology , Transduction, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...