Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Chem ; 56(4): 603-12, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20093556

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) has the poorest overall prognosis among gastrointestinal cancers; however, curative resection in early-stage PDAC greatly improves survival rates, indicating the importance of early detection. Because abnormal microRNA production is commonly detected in cancer, we investigated noninvasive precursor pancreatic intraepithelial neoplasia (PanIN) lesions for microRNA production as a potential early biomarker of PDAC. METHODS: Pathologists identified and classified ductal lesions. We extracted total RNA from laser-capture microdissected PanIN tissue samples from a conditional KRAS(G12D) mouse model (n = 29) or of human origin (n = 38) (KRAS is v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog). MicroRNA production was quantified by quantitative real-time PCR. Internal controls included 5S and U6 RNAs. RESULTS: Production of microRNAs miR-21, miR-205, and miR-200 paralleled PanIN progression in the KRAS(G12D) mouse model, compared with microRNA production in samples of nonpathologic ducts. miR-21 demonstrated the highest relative concentrations in the precursor lesions. Interestingly, miR-205 and miR-21 up-regulation preceded phenotypic changes in the ducts. The production of microRNAs miR-21, miR-221, miR-222, and let-7a increased with human PanIN grade, with peak production occurring in hyperplastic PanIN-2/3 lesions. In situ hybridization analysis indicated miR-21 production to be concentrated in pathologic ductal cells. miR-21 production was regulated by KRAS(G12D) and epidermal growth factor receptor in PDAC-derived cell lines. CONCLUSIONS: Aberrant microRNA production is an early event in the development of PanIN. Our findings indicate that miR-21 warrants further investigation as a marker for early detection of PDAC.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Animals , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/surgery , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Knockout , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
2.
Hum Gene Ther ; 20(8): 831-44, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19323605

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is still the fourth leading cause of cancer-related deaths in Western countries, with increasing incidence. Neither effective prognostic markers nor therapies exist for this cancer. MicroRNAs are potent inhibitors of protein translation, and aberrantly expressed in many cancers. Because let-7 microRNA targets the K-ras oncogene, we aimed to characterize let-7 expression and function in PDAC in vitro and in vivo. Let-7 expression was quantified by real-time RT-PCR from resected tumors and matching adjacent tissue, and in endoscopic ultrasound-guided fine needle aspiration material from patients with PDAC. Let-7 is detected by reverse transcription in situ PCR in a PDAC tissue microarray. PDAC-derived cells were transfected with plasmid-based synthetic microRNAs or by lentiviral transduction, in vitro and in vivo. Let-7 microRNA expression is strongly reduced in PDAC samples, as compared with adjacent tissue. Let-7 is present in normal acinar pancreatic cells, and lost in poorly differentiated cancer samples. In addition, let-7 expression was repressed in patients with PDAC not eligible for surgery. Restoring let-7 levels in cancer-derived cell lines strongly inhibits cell proliferation, K-ras expression, and mitogen-activated protein kinase activation, but fails to impede tumor growth progression after intratumoral gene transfer or after implantation of Capan-1 cells stably overexpressing let-7 microRNA. We describe here for the first time the extensive loss of expression of let-7 in PDAC. In addition, this study provides the initial steps for a microRNA replacement therapy for this cancer.


Subject(s)
MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Aged , Aged, 80 and over , Animals , Biopsy, Fine-Needle , Cell Line, Tumor , Cell Proliferation , Disease Progression , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Genes, ras , Humans , Male , Mice , MicroRNAs/metabolism , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/enzymology , Transfection , Ultrasonography
3.
Endocrinology ; 149(6): 3137-47, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18325993

ABSTRACT

Somatostatin is a neuropeptide that inhibits exocrine and endocrine secretions of several hormones and negatively regulates cell proliferation. These events are mediated through somatostatin engagement on one of five G protein-coupled receptors named SSTR1 to STTR5. Somatostatin binding to SSTR2 mediates predominantly antisecretory and antiproliferative effects; two important biological activities in the gastroenteropancreatic endocrine and exocrine system. Herein we demonstrate novel regulatory sequences for human (h) SSTR2 transcription. By genomic DNA sequence analysis, we reveal two CpG islands located 3.8 kb upstream from the transcription start site. We identify a novel transcription start site and a promoter region within one of these CpG islands. We demonstrate that two epigenetic modifications, DNA methylation and histone acetylation, regulate the activation of hSSTR2 upstream promoter. Furthermore, we show that the transcription from this upstream promoter region directly correlates to hSSTR2 mRNA expression in various human cell lines. A combined treatment of a demethylating agent, 5-aza-2-deoxycytidine and a histone deacetylase inhibitor, trichostatin A, leads to increased expression of hSSTR2 mRNA in cell lines in which the CpG island is methylated. The epigenetic regulation of this promoter region results in differential expression of hSSTR2 mRNA in human cell lines. This study reveals the existence of a novel upstream promoter for the hSSTR2 gene that is regulated by epigenetic modifications, suggesting for complex control of the hSSTR2 transcription.


Subject(s)
Chromosomes, Human, Pair 17 , Promoter Regions, Genetic , RNA, Messenger/genetics , Receptors, Somatostatin/genetics , Base Sequence , Chromatin/genetics , Chromatin/ultrastructure , DNA Methylation , Dinucleoside Phosphates , Exons , Genes, Reporter , Humans , Luciferases/genetics , Molecular Sequence Data , Plasmids , Sequence Deletion , Sequence Homology, Nucleic Acid
4.
Mol Ther ; 14(6): 758-67, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17000136

ABSTRACT

Excepting surgical resection, there is no efficient treatment against pancreatic cancer. The chemotherapeutic agent gemcitabine improves the patient's clinical status but survival is not prolonged. The aim of this study was to design a new strategy to render gemcitabine more efficient in the treatment of pancreatic cancer using gene therapy. We have generated a fusion gene (DCK::UMK) combining deoxycytidine kinase (DCK) and uridine monophosphate kinase (UMK), which converts gemcitabine into its toxic phosphorylated metabolite. Antitumor effects of DCK::UMK gene expression were tested in vitro and in vivo in an orthotopic transplantable model of pancreatic cancer established in hamsters. DCK::UMK sensitizes pancreatic cancer cells to gemcitabine by reducing dramatically both in vitro cell viability and in vivo tumor volume. We found that in vivo expression of DCK::UMK resulted in an antitumor bystander effect due to apoptosis of untransduced cells. In vivo intratumoral gene transfer of DCK::UMK using the synthetic carrier PEI induced a potent tumor regression. Taken together, the results show that the fusion gene DCK::UMK sensitizes pancreatic cancer cells to gemcitabine treatment to induce cell death by apoptosis and tumor regression. Intratumoral delivery of the DCK::UMK gene in combination with gemcitabine might be of high interest for pancreatic cancer management.


Subject(s)
Deoxycytidine Kinase/genetics , Deoxycytidine/analogs & derivatives , Genetic Therapy/methods , Nucleoside-Phosphate Kinase/genetics , Pancreatic Neoplasms/therapy , Animals , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cricetinae , Deoxycytidine/metabolism , Deoxycytidine/pharmacology , Deoxycytidine Kinase/metabolism , Genetic Vectors/genetics , Male , Mesocricetus , Neoplasms, Experimental/therapy , Nucleoside-Phosphate Kinase/metabolism , Pancreatic Neoplasms/pathology , Gemcitabine
5.
Hum Gene Ther ; 16(10): 1175-93, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16218779

ABSTRACT

Pancreatic cancer is one of the most aggressive and devastating human malignancies. The present study was conducted to determine whether in vivo sst2 gene transfer into human pancreatic tumors would impair tumor progression, and to characterize sst2 antitumoral bystander mechanisms. sst2 administration, using the synthetic vector PEI, strongly inhibited tumor progression of human pancreatic adenocarcinoma, in vivo. sst2 gene transfer induced intratumoral production of its ligand somatostatin. Disruption of this autocrine loop by RNA interference completely reversed sst2 antitumoral activity. Mice depleted of natural killer (NK) cells did not hamper sst2 tumor growth inhibition. However, microvessel density and vascular endothelial growth factor (VEGF) expression were markedly reduced in sst2-transfected tumors, whereas sst3 somatostatin receptor was upregulated. Depleting somatostatin by RNA interference completely abolished the sst2 inhibitory effect on VEGF expression and tumor angiogenesis, and sst2-induced sst3 expression in peripheral tumor vessels. We conclude that in vivo sst2 gene transfer elicited intratumoral somatostatin production and strongly impaired human pancreatic tumor growth. NK cells were not involved in this antitumoral bystander effect. VEGF and tumor vascularization were identified as novel targets for sst2-mediated antitumoral bystander effect. sst3 somatostatin receptor was upregulated in sst2-transfected tumors. Therefore, in vivo gene delivery of sst2 receptor to target the angiogenic process in pancreatic ductal adenocarcinoma might be a new therapeutic approach for treatment of pancreatic cancer in patients with unresectable disease.


Subject(s)
Bystander Effect , Carcinoma , Genetic Therapy , Neoplasm Transplantation , Pancreatic Neoplasms , Receptors, Somatostatin/metabolism , Animals , Autocrine Communication/genetics , Bystander Effect/genetics , Carcinoma/blood supply , Carcinoma/metabolism , Carcinoma/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Mice , Neoplasm Transplantation/methods , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Receptors, Somatostatin/genetics
6.
Mol Cell Endocrinol ; 217(1-2): 181-8, 2004 Mar 31.
Article in English | MEDLINE | ID: mdl-15134816

ABSTRACT

The ability of steroid ligands to inactivate the human mineralocorticoid receptor (MR(WT)) has been shown to be due to their inability to contact Asn770, a residue of the H3 helix involved in stabilizing contacts with the H11-H12 loop region. However, all steroid ligands that display antagonist properties when bound to MR(WT), have been shown to activate a mutant receptor (MR(L810)) associated with a severe form of hypertension. Biochemical studies revealed that S810L mutation induces a change in the receptor conformation and increases the steroid-receptor complexes stability. From a three-dimensional model of the MR ligand-binding domain, it is likely that the S810L mutation causes a steric hindrance between the side chains of Leu810 (H5) and Gln776 (H3) that provokes a bending of the H3 helix. As a consequence, the positioning of MR(WT) antagonists within the ligand-binding cavity is modified in such a way that they can activate the mutant MR(L810). The results from biochemical studies also revealed that 5alpha-pregnan-20-one, 4,9-androstadiene-3,17-dione and RU486, unable to bind MR(WT), acted as potent MR(L810) antagonists.


Subject(s)
Hypertension/metabolism , Mineralocorticoid Receptor Antagonists , Steroids/chemistry , Steroids/pharmacology , Amino Acid Substitution , Crystallography, X-Ray , Humans , Hypertension/congenital , Hypertension/genetics , Leucine/genetics , Ligands , Mutation , Protein Binding/genetics , Protein Conformation , Receptors, Mineralocorticoid/chemistry , Receptors, Mineralocorticoid/genetics , Serine/genetics , Steroids/metabolism , Structure-Activity Relationship
7.
J Gastroenterol Hepatol ; 19(4): 388-96, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15012775

ABSTRACT

BACKGROUND AND AIM: The majority of patients cannot benefit from the conventional curative treatments that are currently used for hepatocellular carcinoma (HCC), which remains a world health problem. Interleukin (IL)-12 is one of the most potent anti-tumor cytokines. The aim of the present study was to examine the anti-tumor effect and toxicity of intrahepatic delivery of IL-12 using an ex vivo gene therapy approach in a murine model of HCC. METHODS: Syngenic fibroblasts or MM45T-Li HCC tumor cells were genetically modified in vitro to express IL-12 using a polycistronic TFG murine IL-12 retroviral vector (TFGmIL-12) coding for both p35 and p40 murine IL-12 subunits. Hepatocellular carcinoma was generated using direct intrahepatic inoculation of the tumor cell line into the left liver lobe of BALB/c mice. RESULTS: Direct liver expression of IL-12 by the injected genetically modified tumor cells induced a marked inhibition of tumor growth. This effect was associated with an early infiltration of macrophages, and lymphocytes forming numerous intralobular foci. There was no significant liver toxicity, as shown by normal biochemical liver tests. At a later time, the intralobular foci were rare and consisted mainly of CD4+ T cells, while CD8+ T cells were present in the lobule. Intrahepatic expression of IL-12 did not modify circulating or splenic B lymphocytes or natural killer (NK) cells. The inhibition of tumor growth was maintained in nude mice even when depleted in NK cells. Importantly, in a second model, treatment of established day 7 liver tumors in BALB/c mice using direct intra-tumor injection of syngenic fibroblasts that were genetically modified to express IL-12 significantly reduced tumor size. CONCLUSION: In conclusion, these data provide evidence that experimental HCC can be efficiently and safely treated using ex vivo IL-12 gene therapy, which seems promising for future clinical studies.


Subject(s)
Genetic Therapy , Interleukin-12/genetics , Liver Neoplasms, Experimental/therapy , Animals , Biomedical Engineering , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Cell Movement , Female , Fibroblasts/metabolism , Fibroblasts/transplantation , Injections, Intralesional , Interleukin-12/metabolism , Interleukin-12/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Liver/enzymology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/physiopathology , Liver Neoplasms, Experimental/prevention & control , Macrophages , Mice , Mice, Inbred BALB C , Mice, Nude
8.
Endocrinology ; 144(2): 528-33, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538613

ABSTRACT

A gain of function mutation resulting in the substitution of leucine for serine at codon 810 (S810L) in the human mineralocorticoid receptor (MR) is responsible for early-onset hypertension that is exacerbated in pregnancy. All steroids, including progesterone, that display antagonist properties when bound to the wild-type MR are able to activate the mutant receptor (MR(L810)). These findings suggest that progesterone may contribute to the dramatic aggravation of hypertension in MR(L810) carriers during pregnancy. However, the steroid(s) responsible for hypertension in MR(L810) carriers (men and nonpregnant women) has not yet been identified. Here we show that cortisone and 11-dehydrocorticosterone, the main cortisol and corticosterone metabolites produced in the distal nephron, where sodium reabsorption stimulated by aldosterone takes place, bind with high affinity to MR(L810). The potency with which cortisone and 11-dehydrocorticosterone bind to the mutant MR contrasts sharply with their low wild-type MR-binding capacity. In addition, cotransfection assays demonstrate that cortisone and 11-dehydrocorticosterone are potent activators of the MR(L810) trans-activation function. Because the plasma concentration of cortisol in humans is about 30-fold higher than that of corticosterone, these findings strongly suggest that cortisone is one of the endogenous steroids responsible for early-onset hypertension in men and nonpregnant women carrying the MR(L810) mutation.


Subject(s)
Corticosterone/analogs & derivatives , Cortisone/metabolism , Hypertension/genetics , Hypertension/physiopathology , Point Mutation , Receptors, Mineralocorticoid/genetics , Adult , Aldosterone/metabolism , Aldosterone/pharmacology , Animals , Binding, Competitive , COS Cells , Corticosterone/metabolism , Corticosterone/pharmacology , Cortisone/chemistry , Cortisone/pharmacology , Female , Humans , Hydrocortisone/chemistry , Hydrocortisone/metabolism , Hydrocortisone/pharmacology , Male , Pregnancy , Pregnancy Complications, Cardiovascular/physiopathology , Receptors, Mineralocorticoid/metabolism
9.
Mol Pharmacol ; 62(6): 1306-13, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12435797

ABSTRACT

The binding of mineralocorticoid hormones to the mineralocorticoid receptor is the first step in a cascade of events leading to the stimulation of Na(+) reabsorption by renal cortical collecting duct (CCD) principal cells. The agonist properties of mineralocorticoid hormones are linked to contacts between their 21-hydroxyl group and Asn770, a residue of the ligand-binding domain of the human mineralocorticoid receptor (hMR). Here, we investigate whether the presence of a hydroxyl group at position 11, 17, or 20 could also alter the activity of progesterone (P), a mineralocorticoid antagonist without the 21-hydroxyl group. Both 17 alpha-hydroxyprogesterone (17OHP) and 20 alpha-hydroxyprogesterone (20OHP) antagonized the aldosterone-induced trans-activation activity (IC(50): 17OHP, 10(-7) M; 20OHP, 10(-8) M) of the hMR transiently expressed in COS-7 cells lacking steroid receptors. In cultured mouse mpkCCD(cl4) principal cells, 17OHP and 20OHP also prevented the aldosterone-stimulated amiloride-sensitive component of the short-circuit current (Ams I(sc)), reflecting Na(+) absorption mediated by the epithelial Na(+) channel (ENaC). In contrast, 11 beta-hydroxyprogesterone (11OHP) activated the transiently expressed hMR in COS-7 cells in a dose-dependent manner (ED(50): 10(-8) M) and, like aldosterone, stimulated Ams I(sc) in mpkCCD(cl4) cells. Docking 11OHP within the hMR-ligand-binding domain homology model revealed that the agonist activity of 11OHP is caused by contacts between its 11 beta-hydroxyl group and Asn770. Furthermore, 11OHP was unable to activate the mutant hMR/N770A, in which Ala is substituted for Asn at position 770. These findings demonstrate that in the absence of the 21-hydroxyl group, the 11 beta-hydroxyl group can produce the contact with the hMR-Asn770 required for the hMR activation leading to stimulated Na(+) absorption.


Subject(s)
Hydroxyprogesterones/pharmacology , Kidney Tubules, Collecting/cytology , Potassium/metabolism , Receptors, Mineralocorticoid/agonists , Absorption/drug effects , Aldosterone/pharmacology , Animals , COS Cells , Cell-Free System , Chlorocebus aethiops , Humans , Mice , Progesterone/pharmacology , Receptors, Mineralocorticoid/metabolism , Transcriptional Activation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...