Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
PLoS One ; 4(4): e4897, 2009.
Article in English | MEDLINE | ID: mdl-19340311

ABSTRACT

The von Hippel-Lindau (VHL) syndrome is a pleomorphic familial disease characterized by the development of highly vascularized tumors, such as hemangioblastomas of the central nervous system, pheochromocytomas, renal cell carcinomas, cysts and neuroendocrine tumors of the pancreas. Up to 75% of VHL patients are affected by VHL-associated pancreatic lesions; however, very few reports in the published literature have described the cellular origins and biological roles of VHL in the pancreas. Since homozygous loss of Vhl in mice resulted in embryonic lethality, this study aimed to characterize the functional significance of VHL in the pancreas by conditionally inactivating Vhl utilizing the Cre/LoxP system. Specifically, Vhl was inactivated in different pancreatic cell populations distinguished by their roles during embryonic organ development and their endocrine lineage commitment. With Cre recombinase expression directed by a glucagon promoter in alpha-cells or an insulin promoter in beta-cells, we showed that deletion of Vhl is dispensable for normal functions of the endocrine pancreas. In addition, deficiency of VHL protein (pVHL) in terminally differentiated alpha-cells or beta-cells is insufficient to induce pancreatic neuroendocrine tumorigenesis. Most significantly, we presented the first mouse model of VHL-associated pancreatic disease in mice lacking pVHL utilizing Pdx1-Cre transgenic mice to inactivate Vhl in pancreatic progenitor cells. The highly vascularized microcystic adenomas and hyperplastic islets that developed in Pdx1-Cre;Vhl f/f homozygous mice exhibited clinical features similar to VHL patients. Establishment of three different, cell-specific Vhl knockouts in the pancreas have allowed us to provide evidence suggesting that VHL is functionally important for postnatal ductal and exocrine pancreas, and that VHL-associated pancreatic lesions are likely to originate from progenitor cells, not mature endocrine cells. The novel model systems reported here will provide the basis for further functional and genetic studies to define molecular mechanisms involved in VHL-associated pancreatic diseases.


Subject(s)
Gene Silencing , Pancreas/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Animals , Cell Lineage , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mice , Pancreas/cytology , Up-Regulation , Von Hippel-Lindau Tumor Suppressor Protein/genetics
2.
Cancer Res ; 69(5): 1858-66, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19208834

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal syndrome caused by mutations in the MEN1 tumor suppressor gene. Whereas the protein product of MEN1, menin, is ubiquitously expressed, somatic loss of the remaining wild-type MEN1 allele results in tumors primarily in parathyroid, pituitary, and endocrine pancreas. To understand the endocrine specificity of the MEN1 syndrome, we evaluated biallelic loss of Men1 by inactivating Men1 in pancreatic progenitor cells using the Cre-lox system. Men1 deletion in progenitor cells that differentiate into exocrine and endocrine pancreas did not affect normal pancreas morphogenesis and development. However, mice having homozygous inactivation of the Men1 in pancreas developed endocrine tumors with no exocrine tumor manifestation, recapitulating phenotypes seen in the MEN1 patients. In the absence of menin, the endocrine pancreas showed increase in cell proliferation, vascularity, and abnormal vascular structures; such changes were lacking in exocrine pancreas. Further analysis revealed that these endocrine manifestations were associated with up-regulation in vascular endothelial growth factor expression in both human and mouse MEN1 pancreatic endocrine tumors. Together, these data suggest the presence of cell-specific factors for menin and a permissive endocrine environment for MEN1 tumorigenesis in endocrine pancreas. Based on our analysis, we propose that menin's ability to maintain cellular and microenvironment integrity might explain the endocrine- restrictive nature of the MEN1 syndrome.


Subject(s)
Homeodomain Proteins/physiology , Multiple Endocrine Neoplasia Type 1/etiology , Neuroendocrine Tumors/etiology , Pancreatic Neoplasms/etiology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Animals , Cell Proliferation , Humans , Islets of Langerhans/blood supply , Mice , Vascular Endothelial Growth Factor A/physiology
3.
Cancer ; 115(1): 128-39, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19090007

ABSTRACT

BACKGROUND: Recently, considerable efforts have been directed toward antivascular therapy as a new modality to treat human cancers. However, targeting a therapeutic gene of interest to the tumor vasculature with minimal toxicity to other tissues remains the objective of antivascular gene therapy. Tumor necrosis factor-alpha (TNF-alpha) is a potent antivascular agent but has limited clinical utility because of significant systemic toxicity. At the maximum tolerated doses of systemic TNF-alpha, there is no meaningful antitumor activity. Hence, the objective of this study was to deliver TNF-alpha targeted to tumor vasculature by systemic delivery to examine its antitumor activity. METHODS: A hybrid adeno-associated virus phage vector (AAVP) was used that targets tumor endothelium to express TNF-alpha (AAVP-TNF-alpha). The activity of AAVP-TNF-alpha was analyzed in various in vitro and in vivo settings using a human melanoma tumor model. RESULTS: In vitro, AAVP-TNF-alpha infection of human melanoma cells resulted in high levels of TNF-alpha expression. Systemic administration of targeted AAVP-TNF-alpha to melanoma xenografts in mice produced the specific delivery of virus to tumor vasculature. In contrast, the nontargeted vector did not target to tumor vasculature. Targeted AAVP delivery resulted in expression of TNF-alpha, induction of apoptosis in tumor vessels, and significant inhibition of tumor growth. No systemic toxicity to normal organs was observed. CONCLUSIONS: Targeted AAVP vectors can be used to deliver TNF-alpha specifically to tumor vasculature, potentially reducing its systemic toxicity. Because TNF-alpha is a promising antivascular agent that currently is limited by its toxicity, the current results suggest the potential for clinical translation of this strategy.


Subject(s)
Melanoma/blood supply , Melanoma/therapy , Skin Neoplasms/blood supply , Skin Neoplasms/therapy , Tumor Necrosis Factor-alpha/genetics , Animals , Cell Line, Tumor , Dependovirus/genetics , Gene Expression , Genetic Therapy/methods , Genetic Vectors , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/therapy , Mice , Mice, Nude , Neoplasm Transplantation , Transduction, Genetic , Tumor Necrosis Factor-alpha/metabolism
4.
Cancer Res ; 68(18): 7332-41, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794120

ABSTRACT

Inhibiting angiogenesis has become a major therapeutic strategy for cancer treatment. To identify common intracellular mediators, we previously analyzed gene expression profiles of endothelial cells after treatment with angiogenesis inhibitors. Filamin A interacting protein 1-like (FILIP1L; previously known as down-regulated in ovarian cancer 1) was identified as one of the genes up-regulated in endothelial cells in response to these inhibitors. However, the expression and function of FILIP1L protein is uncharacterized. Here, we provide the first description of the expression and specific subcellular localization of FILIP1L protein in human tissue. Overexpression of FILIP1L resulted in inhibition of cell proliferation and migration and increased apoptosis. In addition, overexpression of FILIP1L truncation mutants showed differential antiproliferative activity. A COOH terminal truncation mutant (FILIP1LDeltaC103) was more potent than wild-type FILIP1L in mediating this activity. Targeted expression of FILIP1LDeltaC103 in tumor vasculature inhibited tumor growth in vivo. Overall, these findings suggest that the novel protein FILIP1L may be an important mediator of the effects of angiogenesis inhibitors and that FILIP1L has the potential to be an antivascular reagent for cancer therapy.


Subject(s)
Colonic Neoplasms/blood supply , Colonic Neoplasms/therapy , Cytokines/biosynthesis , Cytokines/genetics , Melanoma/blood supply , Melanoma/therapy , Animals , Apoptosis/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cytokines/metabolism , DNA, Complementary/genetics , Endostatins/pharmacology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , Genetic Therapy/methods , Humans , Intracellular Signaling Peptides and Proteins , Male , Melanoma/genetics , Melanoma/metabolism , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Subcellular Fractions/metabolism , Transfection , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 14(1): 270-80, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18172279

ABSTRACT

PURPOSE: Over the past 60 years, cytotoxic chemotherapy has targeted the cancer cell. Despite this, there have been few cancer cures. A new approach to cancer therapy is to target the multicellular biological entity of the tumor microenvironment. EXPERIMENTAL DESIGN: Lenalidomide, an immunomodulatory drug, sunitinib, a tyrosine kinase inhibitor, and low-dose metronomic cyclophosphamide, were tested alone and in combination for their abilities to inhibit endothelial cell tube formation, rat aortic ring outgrowth, tumor growth, and metastatic development in mice. In addition, ectopic tumor lysates were evaluated for the presence of proangiogenic proteins. RESULTS: The three agents alone were shown to significantly inhibit endothelial cells' ability to form tubes and significantly inhibit the multicellular microenvironment in the rat aortic ring assay (P < 0.01 and P < 0.001). This effect was also significantly augmented when the agents were combined. Furthermore, the three-drug combination was able halt the progression of tumor growth almost completely in xenograft models of ocular melanoma, colon cancer, pancreatic cancer, and cutaneous melanoma. These agents significantly decrease the number of proliferating cells in tumors, significantly increase the number of cells undergoing active cell death in tumors, and significantly decrease the number of blood vessels in treated tumors (P < 0.05). Combination therapy shows a decrease in the compensatory up-regulation of proangiogenic proteins after treatment when compared with single-agent therapy. CONCLUSIONS: This combination of agents causes an inhospitable microenvironment for tumor cells and shows great promise for use in the clinic.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Proliferation/drug effects , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Endothelial Cells/drug effects , Female , Fluorescent Antibody Technique , Humans , Indoles/administration & dosage , Lenalidomide , Mice , Neoplasms, Experimental/blood supply , Pyrroles/administration & dosage , Rats , Sunitinib , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Xenograft Model Antitumor Assays
6.
J Transl Med ; 5: 38, 2007 Jul 18.
Article in English | MEDLINE | ID: mdl-17640369

ABSTRACT

BACKGROUND: Ocular melanoma is the leading intraocular malignancy. There is no effective treatment for metastatic ocular melanoma. We sought a treatment targeting the tumor microenvironment as well as the tumor cells. METHODS: Migration of HUVEC cells, the ability of HUVEC cells to form tubes, and proliferative capacity of a human ocular melanoma cell line were tested in the presence of lenalidomide and sorafenib alone and in combination. The compounds were also tested in a rat aortic ring assay and were tested in a highly aggressive human ocular melanoma xenograft model. RESULTS: Lenalidomide and Sorafenib inhibit HUVEC ability to migrate and form tubes and when used in combination the inhibition is increased. The agents alone and in combination inhibit outgrowth in the rat aortic ring model. The combination of the agents improved the inhibition over either single agent. In a xenograft model, combination therapy inhibited tumor growth over inhibition by single agent alone in a significant fashion (p < 0.004: lenalidomide and p < 0.0035: sorafenib). Furthermore, spontaneous lung metastasis development was completely inhibited in the combination treated animals. Sixty percent of vehicle treated animals developed lung metastases compared to 50% of lenalidomide treated animals, and 33% of sorafenib treated animals. CONCLUSION: Lenalidomide and sorafenib are effective at targeting endothelial cells, inhibiting growth of ocular melanoma cells and can inhibit growth of tumors in a xenograft model as well as inhibit development of metastases. Combining these agents works in an additive to synergistic way to inhibit the growth of tumors and development of metastases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Eye Neoplasms/drug therapy , Melanoma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzenesulfonates/therapeutic use , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Eye Neoplasms/blood supply , Eye Neoplasms/pathology , Humans , In Vitro Techniques , Lenalidomide , Melanoma/blood supply , Melanoma/pathology , Neovascularization, Pathologic/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley , Sorafenib , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Thalidomide/therapeutic use , Xenograft Model Antitumor Assays
7.
Ann Surg Oncol ; 12(5): 412-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15915376

ABSTRACT

BACKGROUND: Sporadic parathyroid adenomas (SPAs) are benign neoplasms responsible for most cases of primary hyperparathyroidism (pHPT). The molecular pathways responsible for the variations in clinical severity of pHPT are unknown. We studied gene expression profiles in patients with SPAs and pHPT to determine associations between these changes and clinical parameters. METHODS: We selected 10 patients with solitary SPAs and nonfamilial, non-MEN1 pHPT treated with surgery from 2001 to 2003. Pathologic and clinical data were reviewed. At operation, tissues from SPAs were frozen in liquid nitrogen; total RNA was obtained from sections, and the diagnosis was confirmed with hematoxylin and eosin staining. Control normal parathyroid RNA was age- and sex-matched. RNA was amplified, labeled, and hybridized to a microarray of 22,272 human oligonucleotides. Cluster analysis of gene expression, analysis of expression ratios, and comparison of clinical parameters were performed. RESULTS: All patients were cured; all specimens were consistent with SPAs. K means clustering divided the 10 patients into 2 distinct 5-patient gene expression groups by using uncentered correlation based on gene subgrouping. Of the clinical parameters, only the mean gland volume was significantly different between group 1 (390 +/- 160 mm(3)) and group 2 (1080 +/- 615 mm(3); P = .032 by Mann-Whitney test). Seventy-five genes were significantly upregulated or downregulated (with a ratio of <.33 or >3) compared with controls. These genes included the v-fos viral oncogene homolog and six calcium ion-binding signaling proteins. CONCLUSIONS: Differential expression of a few critical genes may contribute to differences in gland volume in SPAs. A better understanding of these pathways may help to define the pathophysiology of pHPT.


Subject(s)
Adenoma/genetics , Gene Expression Profiling , Parathyroid Glands/pathology , Parathyroid Neoplasms/genetics , Adenoma/pathology , Adenoma/physiopathology , Cluster Analysis , Down-Regulation , Female , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Parathyroid Neoplasms/pathology , Parathyroid Neoplasms/physiopathology , Retrospective Studies , Up-Regulation
8.
Genome Res ; 14(8): 1585-93, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15289477

ABSTRACT

A tumor needs to initiate angiogenesis in order to develop its own blood supply, to grow, to invade, and to spread. Angiogenesis, under normal conditions, is a tightly regulated balance between endogenous pro- and antiangiogenic factors. In this study, we investigated, by microarray analysis, the effects of two known antiangiogenic agents (endostatin and fumagillin) on the gene expression profiles of human umbilical vein endothelial cells (HUVEC) in order to elucidate pathways common to the effects of these agents. We observed a majority of gene expression changes within 1 and 2 h of treatment. The genes demonstrating these early expression changes are involved in cell proliferation, gene transcription, and a number have unknown functions. We selected four genes (DOC1, KLF4, TC-1, ID1) from the microarray profile that showed a similar pattern of expression for both of the antiangiogenic agents we tested. We then used small interfering RNAs (siRNA) in an attempt to better understand the role of these selected genes in the inhibitory activity of these agents. Because the gene expression changes occurred within 1 and 2 h of treatment, these genes might be involved in the initial pathways of angiogenesis inhibition.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endostatins/pharmacology , Endothelium, Vascular/drug effects , Fatty Acids, Unsaturated/pharmacology , Gene Expression/drug effects , Cell Division/drug effects , Cells, Cultured , Cyclohexanes , Gene Expression Profiling , Humans , Kruppel-Like Factor 4 , Oligonucleotide Array Sequence Analysis/methods , RNA, Small Interfering/genetics , Sesquiterpenes , Umbilical Veins
9.
Cancer Res ; 64(13): 4481-6, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15231657

ABSTRACT

Solid tumors depend on angiogenesis for sustained growth. Tissue inhibitor of metalloproteinase 2 (TIMP-2) is an angiogenesis inhibitor initially characterized for its ability to block matrix metalloproteinases; however, recent data suggest that the antiangiogenic action of TIMP-2 may rely on matrix metalloproteinase-independent mechanisms. The aim of this study was to identify molecular pathways involved in the effects of TIMP-2 on processes dependent on tumor-host interactions such as angiogenesis. Using in vitro cell culture and a syngeneic murine tumor model, we compared the effects of TIMP-2 overexpression on gene expression profiles in vitro to those observed in vivo. Validating these findings by real-time quantitative PCR and layered protein scanning, we identified up-regulation of mitogen-activated protein kinase phosphatase 1 as an effector of the antiangiogenic function of TIMP-2. Up-regulation of mitogen-activated protein kinase phosphatase 1 in tumors overexpressing TIMP-2 leads to dephosphorylation of p38 mitogen-activated protein kinase and inhibition of tumor growth and angiogenesis. Phosphatase activity appears important in regulating tumor angiogenesis, offering a promising direction for the identification of novel molecular targets and antiangiogenic compounds for the treatment of cancer.


Subject(s)
Cell Cycle Proteins , Colonic Neoplasms/pathology , Neovascularization, Pathologic/pathology , Phosphoprotein Phosphatases , Tissue Inhibitor of Metalloproteinase-2/physiology , Animals , Cell Division/physiology , Colonic Neoplasms/blood supply , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Dual Specificity Phosphatase 1 , Enzyme Induction , Female , Gene Expression Profiling , Immediate-Early Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phosphorylation , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/biosynthesis , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/genetics , Transduction, Genetic , p38 Mitogen-Activated Protein Kinases
10.
Lancet ; 363(9406): 363-7, 2004 Jan 31.
Article in English | MEDLINE | ID: mdl-15070567

ABSTRACT

BACKGROUND: Insulinomas are beta-cell tumours characterised by uncontrolled insulin secretion even in the presence of hypoglycaemia. However, the mechanisms allowing such excessive insulin secretion are not known. Insulin secretion can occur only when the beta-cell insulin stores have been replenished by insulin biosynthesis, which is mainly controlled by translation. Such specific translational regulation often involves the 5' untranslated region. We have identified an insulin splice variant in isolated human pancreatic islets of non-diabetic donors that retains 26 bp of intron 1 and thereby changes the 5' untranslated region, but leaves the coding region unchanged. This splice variant has increased translation efficiency in vitro and in vivo compared with native insulin mRNA. However, splice variant expression is less than 1% of native insulin mRNA in normal islets. METHODS: To test whether this splice variant is involved in insulin production by human insulinomas, we extracted RNA from nine laser-captured surgical insulinoma samples and from isolated islets of nine donors who did not have diabetes. We then determined the ratio of splice variant to native insulin mRNA by quantitative real-time RT-PCR. FINDINGS: The mean ratio of the splice variant to native insulin mRNA was increased more than 50-fold in insulinomas compared with normal islets, and this difference was present in all nine human insulinomas. Overexpression of the splice variant therefore seems to be a general characteristic of insulinomas and is estimated to contribute about 90% to insulin synthesis by these tumours. INTERPRETATION: Overexpression of the insulin splice variant with increased translation efficiency in insulinomas might explain how these tumours maintain high levels of insulin synthesis and secretion leading to hyperinsulinaemia-the hallmark of this disease.


Subject(s)
Alternative Splicing , Insulin/genetics , Insulinoma/genetics , Pancreatic Neoplasms/genetics , Animals , Cell Line, Tumor , Female , Genetic Variation , Humans , Insulin/biosynthesis , Insulinoma/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pancreatic Neoplasms/metabolism , RNA, Messenger/genetics
11.
J Transl Med ; 2(1): 4, 2004 Jan 30.
Article in English | MEDLINE | ID: mdl-14754458

ABSTRACT

Reliable quantitative evaluation of molecular pathways is critical for both drug discovery and treatment monitoring. We have modified the CAM assay to quantitatively measure vascular density, endothelial proliferation, and changes in protein expression in response to anti-angiogenic and pro-angiogenic agents. This improved CAM assay can correlate changes in vascular density with changes seen on a molecular level. We expect that these described modifications will result in a single in vivo assay system, which will improve the ability to investigate molecular mechanisms underlying the angiogenic response.

12.
Oncogene ; 23(8): 1575-85, 2004 Feb 26.
Article in English | MEDLINE | ID: mdl-14767469

ABSTRACT

Corticotropin (ACTH)-independent macronodular adrenal hyperplasia (AIMAH) is a heterogeneous condition in which cortisol secretion may be mediated by gastrointestinal peptide (GIP), vasopressin, catecholamines and other hormones. We studied the expression profile of AIMAH by genomic cDNA microarray analysis. Total RNA was extracted from eight tissues (three GIP-dependent) and compared to total RNA obtained from adrenal glands from 62 normal subjects. Genes had to be altered in 75% of the patients, and be up- or downregulated at a cutoff ratio of at least 2.0; 82 and 31 genes were found to be consistently up- and downregulated, respectively. Among the former were regulators of transcription, chromatin remodeling, and cell cycle and adhesion. Downregulated sequences included genes involved in immune responses and insulin signaling. Hierarchical clustering correlated with the two main AIMAH diagnostic groups: GIP-dependent and non-GIP-dependent. The genes encoding the 7B2 protein (SGNE1) and WNT1-inducible signaling pathway protein 2 (WISP2) were specifically overexpressed in the GIP-dependent AIMAH. For these, and six more genes, the data were validated by semiquantitative amplification in samples from a total of 32 patients (the original eight, six more cases of AIMAH, and 18 other adrenocortical hyperplasias and tumors) and the H295R adrenocortical cancer cell line. In conclusion, our data confirmed AIMAH's clinical heterogeneity by identifying molecularly distinct diagnostic subgroups. Several candidate genes that may be responsible for AIMAH formation and/or progression were also identified, suggesting pathways that affect the cell cycle, adhesion and transcription as possible mediators of adrenocortical hyperplasia.


Subject(s)
Adrenal Cortex Neoplasms/pathology , Adrenal Glands/pathology , Cushing Syndrome/metabolism , Genetic Heterogeneity , Hyperplasia/genetics , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/metabolism , Adult , Cell Line, Tumor , Chromosome Mapping , Cushing Syndrome/diagnosis , Cushing Syndrome/genetics , Cushing Syndrome/pathology , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , RNA/analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
13.
Surgery ; 134(6): 982-7; discussion 987-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14668731

ABSTRACT

BACKGROUND: The molecular pathways that are responsible for pathologic insulin secretion by insulinomas have not been characterized. We studied gene expression profiles from insulinomas and determined associations between these changes and preoperative peak serum insulin levels. METHODS: Ten patients with insulinomas underwent calcium-stimulated arteriography and surgical resection. Tumor RNA was isolated; corresponding complementary DNA was hybridized to 10K human complementary DNA arrays. Pooled human islet cell complementary DNA served as the control. Cluster analysis of gene expression and analysis of expression ratios was performed. RESULTS: Nineteen genes were up-regulated at least 3-fold in insulinomas compared with controls, which included the genes for islet amyloid polypeptide and proprotein convertase type 2. Cluster analysis revealed 2 groups of patients with insulinoma and with distinct patterns of gene expression. Mean peak serum insulin values between groups were 196 and 1100 (U/mL (P<.05), which demonstrates a significant difference in insulin response to calcium stimulation between these 2 groups. CONCLUSION: We show that genes that are relevant to the pathogenesis of hyperinsulinemia are expressed preferentially in insulinomas. In addition, patients with a distinct and common pattern of gene expression had significantly higher stimulated insulin secretion levels. The study of these genes may help to identify the biochemical pathways that are responsible for pathologic insulin secretion.


Subject(s)
Gene Expression/genetics , Insulin/biosynthesis , Insulin/genetics , Insulinoma/genetics , Pancreatic Neoplasms/genetics , Adult , Aged , Female , Humans , Insulinoma/metabolism , Insulinoma/surgery , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Pancreatectomy/methods , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/surgery , RNA, Neoplasm/analysis , Up-Regulation/genetics
14.
Cancer Res ; 63(22): 8022-8, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14633735

ABSTRACT

The inactivation of the MEN1 tumor suppressor gene in patients leads to a constellation of changes in endocrine tissues, including parathyroid neoplasia, pituitary adenomas, pancreatic neuroendocrine tumors, and carcinoids. To study the pathophysiological consequences of the deletion of the MEN1 gene, we set out to create a mouse model of hyperparathyroidism resulting from the deletion of the Men1 gene in parathyroid tissue. We introduced a Men1 gene flanked by loxP sites into the mouse germ line and then used a parathyroid cell-specific promoter to drive the expression of Cre recombinase, resulting in the deletion of the Men1 gene. Here, we show that loss of Men1 gene function in the parathyroid glands of mice results in histological changes consistent with parathyroid neoplasia as well as systemic hypercalcemia. This model provides a means for dissecting the molecular basis of this familial cancer syndrome and may allow for the development of new strategies to treat related forms of hypercalcemia.


Subject(s)
Hypercalcemia/genetics , Hyperparathyroidism/genetics , Parathyroid Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Animals , Calcium/blood , Crosses, Genetic , Female , Gene Deletion , Humans , Hypercalcemia/blood , Hyperparathyroidism/blood , Integrases/genetics , Male , Mice , Mice, Transgenic , Parathyroid Neoplasms/blood , Viral Proteins/genetics
15.
J Cell Physiol ; 197(1): 139-48, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12942550

ABSTRACT

Anti-angiogenic agents regulate tumor growth by inhibiting endothelial cell proliferation and invasion. Carboxyamido-triazole (CAI), an inhibitor of non-voltage-operated calcium entry and calcium influx-mediated pathways, has angiogenesis and invasion inhibitory activity. We hypothesized that CAI may express its anti-angiogenic effects through negative regulation of pro-angiogenic cytokine production and/or function. In vivo, orally administered CAI prevented A2058 human melanoma xenograft growth and concomitantly resulted in a marked reduction in circulating vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8). In vitro, A2058 cell secretion of VEGF was inhibited by CAI treatment under limiting micronutrient conditions that approximate the tumor microenvironment, media restriction, and acidification to pH 6.8 (P=0.0003 and P=0.0006, respectively). VEGF and HIF-1alpha message and protein were also reduced by CAI treatment. Oral CAI treatment reduced vascular ingrowth in vivo into VEGF-containing Matrigel plugs. Commensurate with those findings, human umbilical vein endothelial cell (HUVEC) migration towards VEGF was reduced below background by exposure to CAI in the migration chamber (P<0.0001). An 88% reduction in circulating IL-8 concentration was measured in CAI-treated animals. However, IL-8 protein secretion and gene expression were increased by CAI treatment in culture (P< or =0.01), where CAI caused a dose-dependent acidification of the culture milieu (P< or =0.005). This paradox suggests that IL-8 production in vitro may be more sensitive to ambient pH than cytosolic calcium. These observations suggest that CAI inhibition of tumor cell VEGF production and endothelial cell response to VEGF results in disruption of signaling between the tumor and its microenvironment, causing a net anti-angiogenic effect.


Subject(s)
Antineoplastic Agents/pharmacology , Lymphokines/drug effects , Neovascularization, Pathologic , Triazoles/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Cell Movement/drug effects , Cell Transplantation , Culture Media/chemistry , Endothelial Growth Factors/analysis , Endothelial Growth Factors/blood , Endothelium, Vascular , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit , Immunoblotting , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/blood , Interleukin-8/analysis , Interleukin-8/blood , Lymphokines/analysis , Lymphokines/blood , Melanoma/metabolism , Mice , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/drug effects , Transcription Factors/metabolism , Transplantation, Heterologous , Triazoles/administration & dosage , Tumor Cells, Cultured , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Int J Cancer ; 99(1): 149-53, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11948506

ABSTRACT

Interferon-inducible protein 10 (IP-10) is an immunomodulatory chemokine recently recognized to have potent antiangiogenic activity in vivo. Due to difficulties in the stability, manufacture and chronic administration of recombinant forms of endogenous antiangiogenic proteins, antiangiogenic gene therapy has emerged as a promising new form of cancer treatment. We retrovirally transduced A375 human melanoma cells with the human IP-10 gene and injected cells subcutaneously into nude mice. IP-10-transduced cells also were mixed with null-transduced cells in varying proportions before injection. In vivo growth of IP-10-transduced melanoma cells was markedly diminished compared to parental or null-transduced cells (p = 0.0002, Kruskal-Wallis test). This growth inhibition was associated with a marked reduction in microvessel density. The degree of growth inhibition of tumors following injection of a mixed population of null- and IP-10-transduced cells was directly associated with the fraction of IP-10-transduced cells present. We conclude that retroviral transduction of human melanoma cells with the IP-10 gene leads to sufficient protein secretion to inhibit angiogenesis and tumor growth. These findings suggest that IP-10 gene therapy might be an effective therapy in patients with cancer.


Subject(s)
Chemokines, CXC/genetics , Interferon-gamma/genetics , Melanoma, Experimental/therapy , Retroviridae/genetics , Animals , Blotting, Western , Cell Division , Cells, Cultured , Chemokine CXCL10 , Female , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Immunoenzyme Techniques , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Nude , Mitotic Index , Necrosis , Neoplasm Transplantation , Polymerase Chain Reaction , Transduction, Genetic , Transplantation, Heterologous , Tumor Cells, Cultured , Umbilical Veins
17.
Mol Imaging ; 1(3): 301-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12920855

ABSTRACT

Current methods of studying angiogenesis are limited in their ability to serially evaluate in vivo function throughout a target tissue. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and pharmacokinetic modeling provide a useful method for evaluating tissue vasculature based on contrast accumulation and washout. While it is often assumed that areas of high contrast enhancement and washout comprise areas of increased angiogenesis and tumor activity, the actual molecular pathways that are active in such areas are poorly understood. Using DCE-MRI in a murine subcutaneous tumor model, we were able to perform pharmacokinetic functional analysis of a tumor, coregistration of MRI images with histological cross-sections, immunohistochemistry, laser capture microdissection, and genetic profiling of tumor heterogeneity based on pharmacokinetic parameters. Using imaging as a template for biologic investigation, we have not found evidence of increased expression of proangiogenic modulators at the transcriptional level in either distinct pharmacokinetic region. Furthermore, these regions show no difference on histology and CD31 immunohistochemistry. However, the expression of ribosomal proteins was greatly increased in high enhancement and washout regions, implying increased protein translation and consequent increased cellular activity. Together, these findings point to the potential importance of posttranscriptional regulation in angiogenesis and the need for the development of angiogenesis-specific contrast agents to evaluate in vivo angiogenesis at a molecular level.


Subject(s)
Gene Expression Profiling/methods , Magnetic Resonance Imaging/methods , Neoplasms, Experimental/genetics , Oligonucleotide Array Sequence Analysis/methods , Animals , Base Sequence , Contrast Media , DNA Primers/genetics , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neovascularization, Pathologic
SELECTION OF CITATIONS
SEARCH DETAIL
...