Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Dig Dis Sci ; 58(9): 2528-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23839340

ABSTRACT

BACKGROUND: Enhanced cell expression of MAdCAM-1 is critical in tissue recruitment of lymphocytes in response to stimuli expressing the α4ß7 integrin. MAdCAM-1 is well characterized in gut mucosa with emerging evidence of hepatic expression. AIMS: (i) Compare quantitative/semi-quantitatively MAdCAM-1 expression in relation to early and advanced liver diseases (ii) Define the fine structure of vascular plexuses/lymphatics in the portal tract on which MAdCAM-1 is expressed. METHODS: Using alkaline phosphatase anti-alkaline phosphatase methodology on paraffin embedded tissue sections (n=28) from cirrhotic individuals who underwent orthotopic liver transplant, we evaluated MAdCAM-1 expression and compared with pre-cirrhotic, fulminant hepatitis B, and non-cirrhotic portal hypertension tissue sections. The positive controls included normal colon tissue with negative controls without primary antibody and isotype-matched purified IgG. We developed a real time PCR to quantify levels of MAdCAM-1 mRNA in our samples. RESULTS: MAdCAM-1 was expressed in 27/28 of the cirrhotic sections, localized primarily to septal areas within (i) endothelium of the peribiliary vascular plexus (PBP) (ii) lymphoid aggregates, with absence from normal, non-cirrhotic portal hypertension and pre-cirrhotic livers. There was significant upregulation of MAdCAM-1 mRNA in cirrhosis (p<0.011), consistent with immunohistochemical analysis. CONCLUSIONS: MAdCAM-1 is up-regulated in cirrhosis with expression on PBP and lymphoid aggregates. MAdCAM-1 is likely to contribute to the localization and recruitment of α4ß7 lymphocytes during the pathogenesis of cirrhosis. MAdCAM-1 could be a useful marker of advanced liver disease. Further studies with respect to the expression of MAdCAM-1 in the presence of reversible and non-reversible stages of liver disease may be of merit.


Subject(s)
Immunoglobulins/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Mucoproteins/metabolism , Adult , Cell Adhesion Molecules , Colon/metabolism , Endothelium, Lymphatic/metabolism , Female , Humans , Immunohistochemistry , Liver/blood supply , Lymphoid Tissue/metabolism , Male , Middle Aged , RNA, Messenger/metabolism , Young Adult
2.
Nature ; 428(6984): 754-8, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15085134

ABSTRACT

Vascular development is a complex but orderly process that is tightly regulated. A number of secreted factors produced by surrounding cells regulate endothelial cell (EC) differentiation, proliferation, migration and coalescence into cord-like structures. Vascular cords then undergo tubulogenesis to form vessels with a central lumen. But little is known about how tubulogenesis is regulated in vivo. Here we report the identification and characterization of a new EC-derived secreted factor, EGF-like domain 7 (Egfl7). Egfl7 is expressed at high levels in the vasculature associated with tissue proliferation, and is downregulated in most of the mature vessels in normal adult tissues. Loss of Egfl7 function in zebrafish embryos specifically blocks vascular tubulogenesis. We uncover a dynamic process during which gradual separation and proper spatial arrangement of the angioblasts allow subsequent assembly of vascular tubes. This process fails to take place in Egfl7 knockdown embryos, leading to the failure of vascular tube formation. Our study defines a regulator that controls a specific and important step in vasculogenesis.


Subject(s)
Blood Vessels/embryology , Embryo, Mammalian/blood supply , Endothelial Cells/metabolism , Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Blood Vessels/cytology , Calcium-Binding Proteins , Cell Adhesion , Cell Count , EGF Family of Proteins , Embryo, Mammalian/abnormalities , Embryo, Mammalian/cytology , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/cytology , Endothelial Cells/cytology , In Situ Hybridization , Mice , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish/abnormalities , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
Oncogene ; 24(29): 4688-700, 2005 Jul 07.
Article in English | MEDLINE | ID: mdl-15782116

ABSTRACT

Septins are an evolutionarily conserved family of GTPases with diverse functions including roles in cytokinesis that have been implicated in neoplasia. To address the potential role of SEPT9 in tumorigenesis, we assessed the expression of SEPT9 in 7287 fresh frozen human tissue samples and 292 human cell lines by microarray analysis. In addition, we used a sensitive RT-PCR strategy to define the expression of SEPT9 isoforms in archival formalin-fixed and paraffin-embedded normal human tissues. The mRNA data were further confirmed by immunohistological analyses of SEPT9 protein expression in normal human tissues using antisera that detect SEPT9 isoforms. Using these complementary approaches, we demonstrate that SEPT9 mRNA and protein are expressed ubiquitously, with the isoforms showing tissue-specific expression. The microarray analysis indicates that there is consistent overexpression of SEPT9 in diverse human tumours including breast, CNS, endometrium, kidney, liver, lung, lymphoid, oesophagus, ovary, pancreas, skin, soft tissue and thyroid. Since tumours are commonly associated with enhanced cell proliferation, we examined the possible correlation of Ki67 and SEPT9 expression in normal tissues and tumours. Our data indicate that the overexpression of SEPT9 in neoplasia is not simply a proliferation-associated phenomenon, despite its role in cytokinesis.


Subject(s)
GTP Phosphohydrolases/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Oligonucleotide Array Sequence Analysis , Cell Movement , Cell Proliferation , Humans , Immunohistochemistry , Neoplasms/genetics , Neoplasms/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Septins
4.
J Clin Oncol ; 23(25): 5900-9, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16043828

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) mutations have been associated with tumor response to treatment with single-agent EGFR inhibitors in patients with relapsed non-small-cell lung cancer (NSCLC). The implications of EGFR mutations in patients treated with EGFR inhibitors plus first-line chemotherapy are unknown. KRAS is frequently activated in NSCLC. The relationship of KRAS mutations to outcome after EGFR inhibitor treatment has not been described. PATIENTS AND METHODS: Previously untreated patients with advanced NSCLC in the phase III TRIBUTE study who were randomly assigned to carboplatin and paclitaxel with erlotinib or placebo were assessed for survival, response, and time to progression (TTP). EGFR exons 18 through 21 and KRAS exon 2 were sequenced in tumors from 274 patients. Outcomes were correlated with EGFR and KRAS mutations in retrospective subset analyses. RESULTS: EGFR mutations were detected in 13% of tumors and were associated with longer survival, irrespective of treatment (P < .001). Among erlotinib-treated patients, EGFR mutations were associated with improved response rate (P < .05) and there was a trend toward an erlotinib benefit on TTP (P = .092), but not improved survival (P = .96). KRAS mutations (21% of tumors) were associated with significantly decreased TTP and survival in erlotinib plus chemotherapy-treated patients. CONCLUSION: EGFR mutations may be a positive prognostic factor for survival in advanced NSCLC patients treated with chemotherapy with or without erlotinib, and may predict greater likelihood of response. Patients with KRAS-mutant NSCLC showed poorer clinical outcomes when treated with erlotinib and chemotherapy. Further studies are needed to confirm the findings of this retrospective subset analysis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Genes, ras , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Quinazolines/therapeutic use , Adult , Aged , Aged, 80 and over , Carboplatin/administration & dosage , Combined Modality Therapy , DNA Mutational Analysis , Erlotinib Hydrochloride , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Placebos , Predictive Value of Tests , Prognosis , Survival Analysis , Treatment Outcome
5.
Methods Mol Biol ; 326: 255-64, 2006.
Article in English | MEDLINE | ID: mdl-16780208

ABSTRACT

Tissue microarrays enable the rapid histological localization of gene expression in hundreds of archival samples by in situ hybridization. However, the scoring of tissue microarray data may be influenced by intra- and inter-observer variations, and categorizing continuous variables risks discarding potentially meaningful information. Quantitation imposes a greater degree of objectivity, is more reproducible than subjective discriminations, and facilitates the communication and clarity of definitions. Phosphorimaging has been successfully used to quantitate the hybridization signal intensity from arrayed tissues. The process is rapid and has a wide dynamic range, surpassing the densitometric analysis of autoradiograms. This paper presents a detailed method for quantitative isotopic in situ hybridization on formalin-fixed paraffin-embedded tissue microarrays. In addition, the method includes a protocol for the development of synthetic agarose cores to control for the specificity and sensitivity of hybridization.


Subject(s)
In Situ Hybridization/methods , Oligonucleotide Array Sequence Analysis/methods , Animals , Fixatives , Formaldehyde , Histological Techniques , Humans , Phosphorus Radioisotopes , Sensitivity and Specificity , Tissue Embedding , Tissue Fixation
6.
Clin Cancer Res ; 11(19 Pt 1): 6780-6, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16203764

ABSTRACT

Anillin is an actin-binding protein that can bind septins and is a component of the cytokinetic ring. We assessed the anillin expression in 7,579 human tissue samples and cell lines by DNA microarray analysis. Anillin is expressed ubiquitously but with variable levels of expression, being highest in the central nervous system. The median level of anillin mRNA expression was higher in tumors than normal tissues (median fold increase 2.58; 95% confidence intervals, 2.19-5.68, P < 0.0001) except in the central nervous system where anillin mRNA levels were lower in tumors. We developed a sensitive reverse transcription-PCR strategy to show that anillin mRNA is expressed in cell lines and in cDNA panels derived from fetal and adult tissues, thus validating the microarray data. We compared anillin with Ki67 mRNA expression and found a significant linear relationship between anillin and Ki67 mRNA expression (Spearmann r approximately 0.6, P < 0.0001). Anillin mRNA expression was analyzed during tumor progression in breast, ovarian, kidney, colorectal, hepatic, lung, endometrial, and pancreatic tumors and in all tissues there was progressive increase in anillin mRNA expression from normal to benign to malignant to metastatic disease. Finally, we used anti-anillin sera and found nuclear anillin immunoreactivity to be widespread in normal tissues, often not correlating with proliferative compartments. These data provide insight into the existence of nonproliferation-associated activities of anillin and roles in interphase nuclei. Thus, anillin is overexpressed in diverse common human tumors, but not simply as a consequence of being a proliferation marker. Anillin may have potential as a novel biomarker.


Subject(s)
Contractile Proteins/chemistry , Contractile Proteins/physiology , Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Actins/metabolism , Biomarkers, Tumor , Blotting, Northern , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Central Nervous System/embryology , Cloning, Molecular , DNA, Complementary/metabolism , Exons , HeLa Cells , Humans , Immunohistochemistry , Ki-67 Antigen/biosynthesis , Microfilament Proteins/metabolism , Mitochondrial Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein Binding , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Transcription, Genetic
7.
Cancer Res ; 62(9): 2546-53, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11980648

ABSTRACT

We conducted an expression analysis of prostate stem cell antigen (PSCA)in normal urogenital tissues, benign prostatic hyperplasia (n = 21), prostatic intraepithelial neoplasia (n = 33), and primary (n = 137) and metastatic (n = 42) prostate adenocarcinoma, using isotopic in situ hybridization on tissue microarrays. In normal prostate, we observe PSCA expression in the terminally differentiated, secretory epithelium; strong expression was also seen in normal urothelium. Forty-eight percent of primary and 64% of metastatic prostatic adenocarcinomas expressed PSCA RNA. Our studies did not confirm a positive correlation between level of PSCA RNA expression and high Gleason grade. We characterized monoclonal anti-PSCA antibodies that recognize PSCA expressed on the surface of live cells, are efficiently internalized after antigen recognition, and kill tumor cells in vitro in an antigen-specific fashion upon conjugation with maytansinoid. Unconjugated anti-PSCA antibodies demonstrated efficacy against PSCA-positive tumors by delaying progressive tumor growth in vivo. Maytansinoid-conjugated antibodies caused complete regression of established tumors in a large proportion of animals. Our results strongly suggest that maytansinoid-conjugated anti-PSCA monoclonal antibodies should be evaluated as a therapeutic modality for patients with advanced prostate cancer.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/therapy , Antibodies, Monoclonal/pharmacology , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Adenocarcinoma/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antigens, Neoplasm , Female , GPI-Linked Proteins , Humans , Immunization, Passive/methods , Immunotoxins/pharmacokinetics , Immunotoxins/pharmacology , In Situ Hybridization , Male , Maytansine/pharmacokinetics , Maytansine/pharmacology , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/metabolism
8.
Endocrinology ; 144(6): 2606-16, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12746324

ABSTRACT

We recently described human endocrine gland-derived vascular endothelial growth factor (EG-VEGF) as an endothelial cell mitogen with a novel selective activity and an expression pattern essentially limited to steroidogenic glands. Herein we present the identification and characterization of the mouse ortholog. The mouse cDNA and predicted amino acid sequences are, respectively, 86% and 88% identical with the human. Surprisingly, the mouse EG-VEGF transcript is predominantly expressed in liver and kidney. A comparison of human and mouse EG-VEGF promoter sequences revealed a potential binding site for NR5A1, which is known to be a pivotal element for steroidogenic-specific transcription, in the human but not mouse promoter. In situ hybridization studies localized expression of mouse EG-VEGF mRNA to hepatocytes and renal tubule cells. Interestingly, capillary endothelial cells in these sites share several common structural features with those found in steroidogenic glands. Within liver and kidney, EG-VEGF receptor expression was largely restricted to endothelial cells. Mouse EG-VEGF promoted proliferation and survival of endothelial cells. We propose that mouse EG-VEGF, like human EG-VEGF, plays a role in regulating the phenotype and growth properties of endothelial cells within distinct capillary beds.


Subject(s)
Angiogenesis Inducing Agents/genetics , Endothelium, Vascular/physiology , Gastrointestinal Hormones/genetics , Neovascularization, Physiologic/physiology , Neuropeptides , Vascular Endothelial Growth Factor A , Animals , Cell Survival/physiology , Chromosome Mapping , DNA, Complementary , Endocrine Glands/physiology , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Gene Expression Regulation, Developmental , In Situ Hybridization , Kidney/blood supply , Liver/blood supply , Mice , Mitogens/genetics , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , Receptors, Vascular Endothelial Growth Factor/analysis , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Species Specificity , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived
11.
Clin Cancer Res ; 17(2): 372-81, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21224365

ABSTRACT

PURPOSE: The addition of bevacizumab to cytotoxic chemotherapy has demonstrated a progression-free survival (PFS) benefit in the first-line and second-line treatment of advanced or metastatic breast cancer (MBC). However, the addition of bevacizumab to capecitabine in heavily pretreated MBC patients did not show a PFS benefit (AVF2119g phase III trial). The aim of this study was to evaluate the expression of novel putative biomarkers as predictors of benefit from bevacizumab in retrospective subset analyses of the AVF2119g trial. EXPERIMENTAL DESIGN: In the AVF2119g trial, 462 patients with MBC were randomly assigned to receive capecitabine or capecitabine plus bevacizumab. Primary tumor tissue and outcome data were available for 223 patients. Biomarker expression was assessed by in situ hybridization (VEGF-A, VEGF-B, thrombospondin-2 and Flt4) or immunohistochemistry (VEGF-C, PDGF-C, neuropilin-1, delta-like ligand (Dll) 4, Bv8, p53 and thymidine phosphorylase) on formalin-fixed, paraffin-embedded tissue. PFS was associated with these variables in retrospective subset analyses. RESULTS: Patients with low scores for Dll4, VEGF-C, and neuropilin-1 showed trends toward improvement in PFS associated with the addition of bevacizumab to capecitabine (P values = 0.01, 0.05, and 0.07, respectively). These observations were not statistically significant following correction for multiple hypothesis testing. CONCLUSION: These retrospective subset analyses suggest that expression of Dll4, VEGF-C, and neuropilin-1 may predict benefit from bevacizumab. Such observations are not conclusive but warrant additional testing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Breast Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized , Bevacizumab , Breast Neoplasms/pathology , Capecitabine , Clinical Trials, Phase III as Topic , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease-Free Survival , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Middle Aged , Neoplasm Metastasis , Neuropilin-1 , Vascular Endothelial Growth Factor C
12.
Nat Methods ; 4(9): 713-5, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17704782

ABSTRACT

The discovery of somatic mutations in cancer tissue is extremely laborious, time-consuming and costly. In an evaluation comparing mismatch repair detection (MRD) against Sanger sequencing for somatic-mutation detection, we found that MRD had a specificity of 96% and a sensitivity of 92%. Our results showed that MRD is a robust and cost-effective alternative to Sanger sequencing for identifying somatic mutations in human tumors.


Subject(s)
Base Pair Mismatch/genetics , DNA, Neoplasm/genetics , Escherichia coli/genetics , Mutation , Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cloning, Molecular , Humans , Lung Neoplasms/genetics , Polymerase Chain Reaction , Sensitivity and Specificity
13.
Int J Cancer ; 118(5): 1325-9, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16161048

ABSTRACT

Ovarian carcinoma represents the most lethal gynaecological malignancy. A variety of morphological subtypes are recognised (e.g. serous, mucinous, endometrioid), which may be benign, borderline or malignant. While their relationship is controversial, knowledge of the molecular mechanisms of ovarian tumorigenesis may help resolve this issue and perhaps identify early markers of disease. Perturbed patterns of expression of the SEPT9 gene on chromosome 17q25.3 have been implicated in a variety of tumour types including both breast and ovarian neoplasia. In preliminary studies, we showed that SEPT9 mRNA was upregulated in a bank of ovarian tumours, which included benign, borderline and malignant tumours, and reported increased levels of one splice variant, SEPT9_v4*. We now describe a comprehensive analysis of SEPT9 expression specifically in serous and mucinous ovarian tumours (benign, borderline and malignant), using cDNA microarray, semi- and quantitative RTPCR of microdissected archival tumour material. Our data show consistent and specific overexpression of both SEPT9_v1 and SEPT9_v4* transcripts in the epithelial component of ovarian tumours. These transcripts show highest levels of expression in serous and mucinous borderline tumours. SEPT9_v1 is also upregulated in both serous and mucinous carcinomas. Interestingly, highest levels of expression are observed in serous borderline and low-grade tumours rather than high-grade in keeping with a model of progression of benign, borderline and low-grade serous tumours.


Subject(s)
Cell Transformation, Neoplastic/genetics , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Transcription, Genetic , DNA, Complementary/genetics , Female , Humans , Oligonucleotide Array Sequence Analysis , Septins
14.
J Clin Oncol ; 24(2): 217-27, 2006 Jan 10.
Article in English | MEDLINE | ID: mdl-16365183

ABSTRACT

PURPOSE: Bevacizumab is a monoclonal antibody to vascular endothelial growth factor-A (VEGF). In the pivotal trial in metastatic colorectal cancer (mCRC), addition of bevacizumab to first-line irinotecan, fluorouracil, and leucovorin (IFL) significantly prolonged median survival. The aim of these retrospective subset analyses was to evaluate VEGF, thrombospondin-2 (THBS-2), and microvessel density (MVD) as prognostic factors and/or predictors of benefit from bevacizumab. PATIENTS AND METHODS: In the pivotal trial, 813 patients with untreated mCRC were randomly assigned to receive IFL plus bevacizumab or placebo. Of 312 tissue samples collected (285 primaries, 27 metastases), outcome data were available for 278 (153 bevacizumab, 125 placebo). Epithelial and stromal VEGF expression were assessed by in situ hybridization (ISH) and immunohistochemistry on tissue microarrays and whole sections. Stromal THBS-2 expression was examined by ISH on tissue microarrays. MVD was quantified by Chalkley count. Overall survival was associated with these variables in retrospective subset analyses. RESULTS: In all subgroups, estimated hazard ratios (HRs) for risk of death were < 1 for bevacizumab-treated patients regardless of the level of VEGF or THBS-2 expression or MVD. Patients with a high THBS-2 score showed a nonsignificant improvement in survival following bevacizumab treatment (HR = 0.11; 95% CI, 0.02 to 0.51) compared to patients with a low score (HR = 0.65; 95% CI, 0.41 to 1.02); interaction analysis P = .22. VEGF or THBS-2 expression and MVD were not significant prognostic factors. CONCLUSION: These exploratory analyses suggest that in patients with mCRC addition of bevacizumab to IFL improves survival regardless of the level of VEGF or THBS-2 expression, or MVD.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/drug therapy , Thrombospondins/analysis , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab , Colorectal Neoplasms/mortality , Female , Humans , Immunohistochemistry , Male , Microcirculation/drug effects , Middle Aged , Neoplasm Metastasis , Prognosis , RNA, Messenger/analysis , Thrombospondins/genetics , Treatment Outcome , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/genetics
15.
Biochem Biophys Res Commun ; 333(2): 328-35, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-15961063

ABSTRACT

Vascular endothelial growth factor (VEGF) is an endothelial cell-specific mitogen in vitro and an angiogenic inducer in vivo. The tyrosine kinases Flt-1 (VEGFR-1) and Flk-1/KDR (VEGFR-2) are high affinity VEGF receptors. VEGF plays an essential role in developmental angiogenesis and is important also for reproductive and bone angiogenesis. Substantial evidence also implicates VEGF as a mediator of pathological angiogenesis. Anti-VEGF monoclonal antibodies and other VEGF inhibitors block the growth of several tumor cell lines in nude mice. Clinical trials with VEGF inhibitors in a variety of malignancies are ongoing. Recently, a humanized anti-VEGF monoclonal antibody (bevacizumab; Avastin) has been approved by the FDA as a first-line treatment for metastatic colorectal cancer in combination with chemotherapy. Furthermore, VEGF is implicated in intraocular neovascularization associated with diabetic retinopathy and age-related macular degeneration.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal/administration & dosage , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor A/metabolism , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Bevacizumab , Clinical Trials as Topic , Humans , Neoplasms/immunology , Neoplasms/metabolism , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/immunology
16.
J Pathol ; 206(3): 269-78, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15915442

ABSTRACT

The septins are an evolutionarily conserved family of GTP-binding proteins involved in diverse processes including vesicle trafficking, apoptosis, remodelling of the cytoskeleton, infection, neurodegeneration, and neoplasia. The present paper reports a comprehensive study of septin gene expression by DNA microarray methods in 10 360 samples of normal, diseased, and tumour tissues. A novel septin, SEPT13, has been identified and is shown to be related to SEPT7. It is shown that SEPT13 and the other known human septins are expressed in all tissue types but some show high expression in lymphoid (SEPT1, 6, 9, and 12) or brain tissues (SEPT2, 3, 4, 5, 7, 8, and 11). For a given septin, some isoforms are highly expressed in the brain and others are not. For example, SEPT8_v2 and v1, 1* and 3 are highly expressed in the brain and cluster with SEPT2, 3, 4, 5, 7, and 11. However, a probe set specific for SEPT8_v1 with low brain expression clusters away from this set. Similarly, SEPT4 has lymphoid and non-lymphoid forms; SEPT2 has lymphoid and central nervous system (CNS) forms; and SEPT6 and SEPT9 are elevated in lymphoid tissues but both have forms that cluster away from the lymphoid forms. Perturbation of septin expression was widespread in disease and tumours of the various tissues examined, particularly for conditions of the CNS, where alterations in all 13 septin genes were identified. This analysis provides a comprehensive catalogue of the septin family in health and disease. It is a key step in understanding the role of septins in physiological and pathological states and provides insight into the complexity of septin biology.


Subject(s)
GTP-Binding Proteins/genetics , Gene Expression Profiling/methods , Amino Acid Sequence , Cell Cycle Proteins/genetics , Central Nervous System Diseases/genetics , Cytoskeletal Proteins/genetics , GTP Phosphohydrolases/genetics , Humans , Lymphoid Tissue/chemistry , Membrane Proteins/genetics , Oligonucleotide Array Sequence Analysis/methods , Phosphoric Monoester Hydrolases/genetics , Phylogeny , Septins
17.
J Natl Cancer Inst ; 97(13): 981-9, 2005 Jul 06.
Article in English | MEDLINE | ID: mdl-15998951

ABSTRACT

BACKGROUND: A recent phase III trial showed that the addition of bevacizumab, a monoclonal antibody to vascular endothelial growth factor-A, to first-line irinotecan, 5-fluorouracil, and leucovorin (IFL) prolonged median survival in patients with metastatic colorectal cancer. We carried out a retrospective analysis of patients in the trial to evaluate whether mutation status of k-ras, b-raf, or p53 or P53 expression could predict which patients were more likely to respond to bevacizumab. METHODS: Microdissected tumors from 295 patients (274 primary tumors, 21 metastases) were subject to DNA sequence analysis to identify mutations in k-ras, b-raf, and p53. Nuclear P53 expression was determined by immunohistochemistry. Hazard ratios and 95% confidence intervals (CI) for overall survival were estimated using Cox regression analysis. RESULTS: In all biomarker subgroups, estimated hazard ratios for risk of death were less than 1 for bevacizumab-treated patients as compared with those for placebo-treated patients. Mutations in k-ras and/or b-raf were observed in 88 of 213 patients (41%). Hazard ratios for death among patients with tumors with wild-type k-ras/b-raf status, as compared with those of patients with mutations in one or both genes, were 0.51 (95% CI = 0.28 to 0.95) among those treated with IFL plus bevacizumab and 0.66 (95% CI = 0.37 to 1.18) among those treated with IFL plus placebo. Mutations in p53 were found in 139 of 205 patients (68%), and P53 was overexpressed in 191 of 266 patients (72%); neither p53 mutation nor P53 overexpression was statistically significantly associated with survival. CONCLUSIONS: We did not find a statistically significant relationship between mutations of k-ras, b-raf, or p53 and the increase in median survival associated with the addition of bevacizumab to IFL in metastatic colorectal cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab , Biomarkers, Tumor/genetics , Clinical Trials, Phase III as Topic , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Laser Therapy , Male , Microdissection/instrumentation , Middle Aged , Predictive Value of Tests , Prognosis , Retrospective Studies , Treatment Outcome
18.
Proc Natl Acad Sci U S A ; 100(5): 2685-90, 2003 Mar 04.
Article in English | MEDLINE | ID: mdl-12604792

ABSTRACT

We recently identified an angiogenic mitogen, endocrine-gland-derived vascular endothelial growth factor (EG-VEGF), with selective activity for endothelial cells of endocrine tissues. Here we describe the characterization of a highly related molecule, Bv8, also known as prokineticin-2. Human Bv8 shares 60% identity and 75% similarity with EG-VEGF. The human and mouse Bv8 genes share a common structure. Like EG-VEGF, Bv8 is able to induce proliferation, survival and migration of adrenal cortical capillary endothelial cells. Bv8 gene expression is induced by hypoxic stress. Bv8 expression occurs predominantly in the testis and is largely restricted to primary spermatocytes. Adenoviral delivery of Bv8 or EG-VEGF to the mouse testis resulted in a potent angiogenic response. We have localized the expression of the Bv8EG-VEGF receptors within the testis to vascular endothelial cells. The testis exhibits relatively high turnover of endothelial cells. Therefore, Bv8 and EG-VEGF, along with other factors such as VEGF-A, may maintain the integrity and also regulate proliferation of the blood vessels in the testis.


Subject(s)
Gastrointestinal Hormones/metabolism , Gastrointestinal Hormones/physiology , Neovascularization, Pathologic , Neuropeptides , Receptors, Peptide/biosynthesis , Adenoviridae/genetics , Amino Acid Sequence , Animals , Base Sequence , Cattle , Cell Division , Cell Movement , Cells, Cultured , Chemotaxis , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Endothelial Growth Factors/metabolism , Endothelium/metabolism , Endothelium, Vascular/cytology , Humans , Hypoxia , Immunoblotting , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Male , Mice , Models, Genetic , Molecular Sequence Data , Protein Isoforms , RNA/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Spermatozoa/cytology , Testis/blood supply , Testis/metabolism , Testis/pathology , Tissue Distribution , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Science ; 299(5608): 890-3, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12574630

ABSTRACT

The vascular endothelium was once thought to function primarily in nutrient and oxygen delivery, but recent evidence suggests that it may play a broader role in tissue homeostasis. To explore the role of sinusoidal endothelial cells (LSECs) in the adult liver, we studied the effects of vascular endothelial growth factor (VEGF) receptor activation on mouse hepatocyte growth. Delivery of VEGF-A increased liver mass in mice but did not stimulate growth of hepatocytes in vitro, unless LSECs were also present in the culture. Hepatocyte growth factor (HGF) was identified as one of the LSEC-derived paracrine mediators promoting hepatocyte growth. Selective activation of VEGF receptor-1 (VEGFR-1) stimulated hepatocyte but not endothelial proliferation in vivo and reduced liver damage in mice exposed to a hepatotoxin. Thus, VEGFR-1 agonists may have therapeutic potential for preservation of organ function in certain liver disorders.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Hepatocytes/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Liver/cytology , Liver/physiology , Lymphokines/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , CHO Cells , Carbon Tetrachloride/toxicity , Cell Division , Cells, Cultured , Chemical and Drug Induced Liver Injury , Coculture Techniques , Cricetinae , DNA Replication , Endothelial Growth Factors/genetics , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/physiology , Gene Expression Regulation , Growth Substances/genetics , Growth Substances/metabolism , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Hepatocytes/cytology , Hepatocytes/pathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Liver/blood supply , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Liver Diseases/prevention & control , Liver Regeneration , Lymphokines/genetics , Lymphokines/pharmacology , Mice , Mice, Nude , Mitosis , Mutation , Necrosis , Neovascularization, Physiologic , Paracrine Communication , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factors
20.
Am J Pathol ; 160(6): 2295-307, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12057932

ABSTRACT

Most mouse models of hepatocellular carcinoma have expressed growth factors and oncogenes under the control of a liver-specific promoter. In contrast, we describe here the formation of liver tumors in transgenic mice overexpressing human fibroblast growth factor 19 (FGF19) in skeletal muscle. FGF19 transgenic mice had elevated hepatic alpha-fetoprotein mRNA as early as 2 months of age, and hepatocellular carcinomas were evident by 10 months of age. Increased proliferation of pericentral hepatocytes was demonstrated by 5-bromo-2'-deoxyuridine incorporation in the FGF19 transgenic mice before tumor formation and in nontransgenic mice injected with recombinant FGF19 protein. Areas of small cell dysplasia were initially evident pericentrally, and dysplastic/neoplastic foci throughout the hepatic lobule were glutamine synthetase-positive, suggestive of a pericentral origin. Consistent with chronic activation of the Wingless/Wnt pathway, 44% of the hepatocellular tumors from FGF19 transgenic mice had nuclear staining for beta-catenin. Sequencing of the tumor DNA encoding beta-catenin revealed point mutations that resulted in amino acid substitutions. These findings suggest a previously unknown role for FGF19 in hepatocellular carcinomas.


Subject(s)
Disease Models, Animal , Fibroblast Growth Factors/biosynthesis , Liver Neoplasms, Experimental/pathology , Muscle, Skeletal/metabolism , Trans-Activators , Zebrafish Proteins , Amino Acid Sequence , Amino Acid Substitution , Animals , Bromodeoxyuridine/metabolism , Cell Division/drug effects , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , DNA, Neoplasm/chemistry , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factors/genetics , Humans , Immunohistochemistry , Liver/metabolism , Liver Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Proto-Oncogene Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 4 , Receptors, Fibroblast Growth Factor/biosynthesis , Recombinant Proteins/metabolism , Wnt Proteins , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL