Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
JCI Insight ; 6(17)2021 09 08.
Article in English | MEDLINE | ID: mdl-34237033

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) tumors are characterized by a desmoplastic reaction resulting in dense deposition of collagen that is known to promote cancer progression. A central mediator of protumorigenic collagen signaling is the receptor tyrosine kinase discoid domain receptor 1 (DDR1). DDR1 is a critical driver of a mesenchymal and invasive cancer cell PDAC phenotype. Previous studies have demonstrated that genetic or pharmacologic inhibition of DDR1 reduces PDAC tumorigenesis and metastasis. Here, we investigated whether DDR1 signaling has cancer cell nonautonomous effects that promote PDAC progression and metastasis. We demonstrate that collagen-induced DDR1 activation in cancer cells is a major stimulus for CXCL5 production, resulting in the recruitment of tumor-associated neutrophils (TANs), the formation of neutrophil extracellular traps (NETs), and subsequent cancer cell invasion and metastasis. Moreover, we have identified that collagen-induced CXCL5 production was mediated by a DDR1/PKCθ/SYK/NF-κB signaling cascade. Together, these results highlight the critical contribution of the collagen I-DDR1 interaction in the formation of an immune microenvironment that promotes PDAC metastasis.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Discoidin Domain Receptor 1/genetics , Extracellular Traps/genetics , Gene Expression Regulation, Neoplastic , Neoplasms, Experimental , Neutrophils/pathology , Pancreatic Neoplasms/genetics , Animals , Carcinogenesis , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA, Neoplasm/genetics , Discoidin Domain Receptor 1/biosynthesis , Extracellular Traps/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis , Neutrophils/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction , Tumor Microenvironment
2.
In Vivo ; 27(4): 473-84, 2013.
Article in English | MEDLINE | ID: mdl-23812217

ABSTRACT

BACKGROUND: The murine model of 4-nitroquinoline 1-oxide (4-NQO)-induced oral and esophageal cancer is frequently used to assess the effects of different cancer prevention/ therapy agents in vivo, but the molecular mechanisms in those 4-NQO-induced carcinogenesis are unknown. This study investigated aberrant expression of cell growth-critical genes in 4-NQO-induced oral and esophageal cancer tissues in mice compared to those present in the human disease for association with survival of patients. MATERIALS AND METHODS: C57LB6/129Sv mice were given 4-NQO in their drinking water to induce oral and esophageal cancer. Quantitative-reverse transcription polymerase chain reaction (qRT-PCR), western blot, and immunohistochemistry were used to detect gene expression in the cancer tissues from mice and in 4-NQO-treated human esophageal cancer cell lines and esophageal cancer tissues. Methylation-specific PCR and DNA sequencing were performed to assess methylation of the Rarb2 promoter in murine tissues. Kaplan-Meier analysis was performed to associate gene expression in esophageal cancer tissues with survival data for patients with esophageal cancer. RESULTS: 4-NQO dose-dependently induced pre-malignant and malignant lesions in the oral cavity and esophagus in mice that pathologically and morphologically mimicked human oral and esophageal cancer. Molecularly, 4-NQO inhibited Rarß2 but induced expression of phosphorylated extracellular-signal-regulated kinase-1 and -2 (p-ERK1/2) and Cox2 proteins and Rarß2 gene promoter methylation in murine tumors. In vitro treatment with 4-NQO altered expression of RARß2, p-ERK1/2, and COX2 in human esophageal cancer cells. In tissues from 90 patients with esophageal cancer, expression of p-ERK1/2 and COX2 was up-regulated, and p-ERK1/2 expression was associated with advanced clinical tumor stage and consumption of hot beverages, while COX2 expression was associated with tumor de-differentiation in esophageal cancer. Furthermore, expression of p-ERK1/2 was associated with a worse overall survival rate of patients (p=0.014), whereas the association of COX2 expression with worse overall survival rate did not reach statistical significance (p=0.19). Knockdown of COX2 expression using transient transfection of a COX2 antisense expression vector inhibited Ki67 expression, an indicator of cell proliferation, in human esophageal cancer cells. CONCLUSION: 4-NQO-induced cancer in oral cavity and esophagus of mice not only pathologically and morphologically mimicked human oral and esophageal cancer, but also shared some molecular alterations (e.g. aberrant expression of Rarb2, p-ERK1/2, and Cox2). This study further demonstrated that targeting of the altered RARß2-led gene pathway could effectively suppress the development of this deadly type of cancer.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Neoplasms/mortality , Mouth Neoplasms/genetics , Mouth Neoplasms/mortality , 4-Nitroquinoline-1-oxide/adverse effects , Animals , Carcinogens/pharmacology , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , DNA Methylation/drug effects , Disease Models, Animal , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mouth Neoplasms/pathology , Promoter Regions, Genetic , Receptors, Retinoic Acid/genetics
3.
Mol Cancer ; 11: 4, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22239913

ABSTRACT

BACKGROUND: Although cigarette smoking is the principal cause of lung carcinogenesis, chronic obstructive pulmonary disease (COPD), an inflammatory disease of the lung, has been identified as an independent risk factor for lung cancer. Bacterial colonization, particularly with non-typeable Haemophilus influenzae (NTHi), has been implicated as a cause of airway inflammation in COPD besides cigarette smoke. Accordingly, we hypothesized that lung cancer promotion may occur in a chronic inflammatory environment in the absence of concurrent carcinogen exposure. RESULTS: Herein, we investigated the effects of bacterial-induced COPD-like inflammation and tobacco carcinogen-enhanced tumorigenesis/inflammation in the retinoic acid inducible G protein coupled receptor knock out mouse model (Gprc5a-/- mouse) characterized by late-onset, low multiplicity tumor formation. Three-month-old Gprc5a-/- mice received 4 intraperitoneal injections of the tobacco-specific carcinogen, NNK, followed by weekly exposure to aerosolized NTHi lysate for 6 months. The numbers of inflammatory cells in the lungs and levels of several inflammatory mediators were increased in Gprc5a-/- mice treated with NTHi alone, and even more so in mice pretreated with NNK followed by NTHi. The incidence of spontaneous lung lesions in the Gprc5a-/- mice was low, but NTHi exposure led to enhanced development of hyperplastic lesions. Gprc5a-/- mice exposed to NNK alone developed multiple lung tumors, while NTHi exposure increased the number of hyperplastic foci 6-fold and the tumor multiplicity 2-fold. This was associated with increased microvessel density and HIF-1α expression. CONCLUSION: We conclude that chronic extrinsic lung inflammation induced by bacteria alone or in combination with NNK enhances lung tumorigenesis in Gprc5a-/- mice.


Subject(s)
Cell Transformation, Neoplastic , Lung Neoplasms/etiology , Pulmonary Disease, Chronic Obstructive/complications , Receptors, G-Protein-Coupled/genetics , Animals , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Haemophilus Infections/complications , Haemophilus Infections/microbiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/microbiology , Nitrosamines , Pulmonary Disease, Chronic Obstructive/microbiology
4.
Cancer Res ; 70(21): 8917-26, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20959490

ABSTRACT

Signal transducers and activators of transcription 3 (Stat3) is activated by cytokines and growth factors in lung cancers and regulates expression of genes implicated in cell growth, survival, and transformation. Previously, we found that mice with a deletion of the G protein-coupled receptor, family C, group 5, member a (Gprc5a) gene develop lung tumors, indicating that Gprc5a is a tumor suppressor. Herein, we show that epithelial cells from Gprc5a knockout mouse lung (Gprc5a(-/-) cells) survive better in vitro in medium deprived of exogenous growth factors and form more colonies in semisolid medium than their counterparts from wild-type mice (Gprc5a(+/+) cells). Stat3 tyrosine 705 phosphorylation and expression of several Stat3-regulated antiapoptotic genes were higher in Gprc5a(-/-) than in Gprc5a(+/+) cells. Both cell types secreted leukemia inhibitory factor (Lif); however, whereas Stat3 activation was persistent in Gprc5a(-/-) cells, it was transient in Gprc5a(+/+) cells. Lung adenocarcinoma cells isolated from Gprc5a(-/-) mice also exhibited autocrine Lif-mediated Stat3 activation. The level of Socs3, the endogenous Stat3 inhibitory protein, was higher in Gprc5a(+/+) than in Gprc5a(-/-) cells, and expression of the tumor suppressor stabilized Socs3. Inhibition of Stat3 signaling in Gprc5a(-/-) normal and cancer cells by the Janus-activated kinase 2 inhibitor AG490 or by a dominant negative Stat3(Y705F) increased starvation-induced apoptosis and inhibited colony formation. These results show that persistent Stat3 activation is important for the survival and transformation of Gprc5a(-/-) lung cells and suggest that the tumor suppressive effects of Gprc5a are mediated, at least in part, by inhibition of Stat3 signaling through Socs3 stabilization.


Subject(s)
Adenocarcinoma/pathology , Cell Transformation, Neoplastic/pathology , Leukemia Inhibitory Factor/pharmacology , Lung Neoplasms/pathology , Lung/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , STAT3 Transcription Factor/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Kidney/cytology , Kidney/metabolism , Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , Signal Transduction
5.
PLoS One ; 5(7): e11847, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20686609

ABSTRACT

BACKGROUND: Improved understanding of lung cancer development and progression, including insights from studies of animal models, are needed to combat this fatal disease. Previously, we found that mice with a knockout (KO) of G-protein coupled receptor 5A (Gprc5a) develop lung tumors after a long latent period (12 to 24 months). METHODOLOGY/PRINCIPAL FINDINGS: To determine whether a tobacco carcinogen will enhance tumorigenesis in this model, we administered 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) i.p. to 2-months old Gprc5a-KO mice and sacrificed groups (n=5) of mice at 6, 9, 12, and 18 months later. Compared to control Gprc5a-KO mice, NNK-treated mice developed lung tumors at least 6 months earlier, exhibited 2- to 4-fold increased tumor incidence and multiplicity, and showed a dramatic increase in lesion size. A gene expression signature, NNK-ADC, of differentially expressed genes derived by transcriptome analysis of epithelial cell lines from normal lungs of Gprc5a-KO mice and from NNK-induced adenocarcinoma was highly similar to differential expression patterns observed between normal and tumorigenic human lung cells. The NNK-ADC expression signature also separated both mouse and human adenocarcinomas from adjacent normal lung tissues based on publicly available microarray datasets. A key feature of the signature, up-regulation of Ube2c, Mcm2, and Fen1, was validated in mouse normal lung and adenocarcinoma tissues and cells by immunohistochemistry and western blotting, respectively. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that lung tumorigenesis in the Gprc5a-KO mouse model is augmented by NNK and that gene expression changes induced by tobacco carcinogen(s) may be conserved between mouse and human lung epithelial cells. Further experimentation to prove the reliability of the Gprc5a knockout mouse model for the study of tobacco-induced lung carcinogenesis is warranted.


Subject(s)
Adenocarcinoma/genetics , Carcinogens/toxicity , Genomics/methods , Lung Neoplasms/genetics , Nitrosamines/toxicity , Receptors, G-Protein-Coupled/physiology , Animals , Blotting, Western , Cyclin D1/genetics , Cyclin D1/metabolism , Flap Endonucleases/genetics , Flap Endonucleases/metabolism , Humans , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, G-Protein-Coupled/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
6.
Neoplasia ; 12(6): 499-505, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20563252

ABSTRACT

Increasing the understanding of the impact of changes in oncogenes and tumor suppressor genes is essential for improving the management of lung cancer. Recently, we identified a new mouse lung-specific tumor suppressor-the G protein-coupled receptor 5A (Gprc5a). Microarray analysis of the transcriptomes of lung epithelial cells cultured from normal tracheas of Gprc5a knockout and wild-type mice defined a loss-of-Gprc5a gene signature, which revealed many aberrations in cancer-associated pathways. To assess the relevance of this mouse tumor suppressor to human lung cancer, the loss-of-Gprc5a signature was cross species compared with and integrated with publicly available gene expression data of human normal lung tissue and non-small cell lung cancers. The loss-of-Gprc5a signature was prevalent in human lung adenocarcinomas compared with squamous cell carcinomas or normal lung. Furthermore, it identified subsets of lung adenocarcinomas with poor outcome. These results demonstrate that gene expression patterns of Gprc5a loss in nontumorigenic mouse lung epithelial cells are evolutionarily conserved and important in human lung adenocarcinomas.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/genetics , Lung Neoplasms/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Female , Gene Expression Profiling , Genes, Tumor Suppressor , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
7.
Cancer Prev Res (Phila) ; 3(4): 424-37, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20354164

ABSTRACT

Mouse models can be useful for increasing the understanding of lung tumorigenesis and assessing the potential of chemopreventive agents. We explored the role of inflammation in lung tumor development in mice with knockout of the tumor suppressor Gprc5a. Examination of normal lung tissue and tumors from 51 Gprc5a(+/+) (adenoma incidence, 9.8%; adenocarcinoma, 0%) and 38 Gprc5a(-/-) mice (adenoma, 63%; adenocarcinoma, 21%) revealed macrophage infiltration into lungs of 45% of the Gprc5a(-/-) mice and 8% of Gprc5a(+/+) mice and the direct association of macrophages with 42% of adenomas and 88% of adenocarcinomas in the knockout mice. Gprc5a(-/-) mouse lungs contained higher constitutive levels of proinflammatory cytokines and chemokines and were more sensitive than lungs of Gprc5a(+/+) mice to stimulation of NF-kappaB activation by lipopolysaccharide in vivo. Studies with epithelial cells cultured from tracheas of Gprc5a(-/-) and Gprc5a(+/+) mice revealed that Gprc5a loss is associated with increased cell proliferation, resistance to cell death in suspension, and increased basal, tumor necrosis factor alpha-induced, and lipopolysaccharide-induced NF-kappaB activation, which were reversed partially in Gprc5a(-/-) adenocarcinoma cells by reexpression of Gprc5a. Compared with Gprc5a(+/+) cells, the Gprc5a(-/-) cells produced higher levels of chemokines and cytokines and their conditioned medium induced more extensive macrophage migration. Silencing Gprc5a and the p65 subunit of NF-kappaB in Gprc5a(+/+) and Gprc5a(-/-) cells, respectively, reversed these effects. Thus, Gprc5a loss enhances NF-kappaB activation in lung epithelial cells, leading to increased autocrine and paracrine interactions, cell autonomy, and enhanced inflammation, which may synergize in the creation of a tumor-promoting microenvironment.


Subject(s)
Adenocarcinoma/metabolism , Cell Transformation, Neoplastic/metabolism , Lung Neoplasms/metabolism , NF-kappa B/metabolism , Pneumonia/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Animals , Cell Transformation, Neoplastic/genetics , Chemotaxis, Leukocyte/immunology , Electrophoretic Mobility Shift Assay , Enzyme Activation/genetics , Immunoblotting , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/genetics , Pneumonia/immunology , RNA, Messenger/analysis , RNA, Small Interfering , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction
8.
Cancer Prev Res (Phila) ; 2(3): 274-80, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19258542

ABSTRACT

Retinoic acid receptor-beta2 (RAR-beta2) is a putative tumor suppressor gene in various cancers. To determine the underlying molecular mechanisms, we transfected RAR-beta2 cDNA into esophageal cancer TE-1 and TE-8 cells and found that RAR-beta2 suppressed tumor cell growth in vitro and tumor formation in nude mice in TE-8 cells, whereas the stable transfection of RAR-beta2 did not restore retinoid sensitivity or inhibit tumor formation in nude mouse in TE-1 cells. Molecularly, we revealed that RAR-beta2 antitumor activity was associated with expression and suppression of cyclooxygenase-2 (COX-2) in these tumor cell lines. Moreover, antisense RAR-beta2 cDNA induced COX-2 expression in TE-3 cells. Furthermore, when COX-2 expression is first blocked by using antisense COX-2 expression vector, the effect of RAR-beta2 is diminished in these tumor cells. In addition, we analyzed expression of RAR-beta2 and COX-2 mRNA in tissue specimens and found that RAR-beta2 expression is associated with low levels of COX-2 expression in esophageal cancer tissues. Induction of RAR-beta2 expression in oral leukoplakia tissues after the patients treated with 13-cis RA correlated with a reduction in COX-2 expression and clinical response. Our findings indicate that some of RAR-beta2 antitumor activities are mediated by suppression of COX-2 expression in some of these esophageal cancer cells. After correlating antitumor effect of RAR-beta2 with COX-2 expression in the published studies, we also found the association. Thus, further studies will determine whether manipulation of COX-2 expression in different cancers can antagonize RAR-beta2 activity.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/prevention & control , Receptors, Retinoic Acid/metabolism , Animals , Anticarcinogenic Agents/therapeutic use , Cell Survival , DNA, Complementary/metabolism , Genetic Vectors , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Transfection
9.
J Natl Cancer Inst ; 99(22): 1668-82, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18000218

ABSTRACT

BACKGROUND: Lung cancers develop via multiple genetic and epigenetic changes, including inactivation of tumor suppressor genes. We previously cloned human G protein-coupled receptor family C type 5A (GPRC5A), whose expression is suppressed in some human lung carcinoma cells, and its mouse homolog Gprc5a. METHODS: We generated Gprc5a knockout mice by homologous recombination and studied their phenotype by macroscopic observation and microscopic histologic analysis of embryos and lungs of 1- to 2-year-old mice. GPRC5A mRNA expression was analyzed by reverse transcription-polymerase chain reaction in surgical specimens of 18 human lung tumors and adjacent normal tissues and by analyzing previously published data from 186 lung tumor tissues of a variety of histologic types and 17 normal lung samples. Human embryonic kidney, human non-small-cell lung cancer, and mouse lung adenocarcinoma cells were transfected with a GPRC5A expression vector or a control vector, and colony formation in semisolid medium was assayed. Statistical tests were two-sided. RESULTS: Homozygous knockout mice developed many more lung tumors at 1-2 years of age (incidence: 76% adenomas and 17% adenocarcinomas) than heterozygous (11% adenomas) or wild-type (10% adenomas) mice. Human GPRC5A mRNA levels were lower in most (11 of 18 [61%]) human lung tumors than in adjacent normal tissues. The mean GPRC5A mRNA level in adenocarcinoma (n = 139), squamous cell carcinoma (n = 21), small-cell lung cancer (n = 6), and carcinoid (n = 20) tissues was 46.2% (P = .014), 7.5% (P<.001), 5.3% (P<.001), and 1.8% (P<.001), respectively, that in normal lung tissues (n = 17) GPRC5A transfection suppressed colony formation in semisolid medium of immortalized human embryonic kidney, human non-small-cell lung cancer, and mouse lung adenocarcinoma cells by 91%, 91%, and 68%, respectively, compared with vector controls (all P<.001). CONCLUSIONS: Gprc5a functions as a tumor suppressor in mouse lung, and human GPRC5A may share this property. The Gprc5a-deficient mouse is a novel model to study lung carcinogenesis and chemoprevention.


Subject(s)
Adenocarcinoma/chemistry , Genes, Tumor Suppressor , Lung Neoplasms/chemistry , Lung/chemistry , Neoplastic Stem Cells/chemistry , Receptors, G-Protein-Coupled/genetics , Respiratory Mucosa/pathology , Adenocarcinoma/pathology , Animals , Blotting, Northern , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Small Cell/chemistry , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Embryonic Stem Cells , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Genes, ras , Genetic Predisposition to Disease , Homozygote , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Lung Neoplasms/pathology , Mice , Mice, Knockout , Mutation , Neoplasm Proteins/genetics , Phenotype , Protein Array Analysis , RNA, Messenger/analysis , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Stem Cell Assay
10.
Am J Pathol ; 162(4): 1241-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12651616

ABSTRACT

Using LoxP/Cre technology, we generated a knockout mouse homozygous for a null mutation in exon 2 of Cav1. In male Cav1-/- animals, we observed a dramatic increase in the incidence of urinary calcium stone formation. In 5-month-old male mice, the incidence of early urinary calculi was 67% in Cav1-/- mice compared to 19% in Cav1+/+ animals. Frank stone formation was observed in 13% of Cav1-/- males but was not seen in Cav1+/+ mice. Urine calcium concentration was significantly higher in Cav1-/- male mice compared to Cav1+/+ mice. In Cav1-/- mice, distal convoluted tubule cells were completely devoid of Cav1 and the localization of plasma membrane calcium ATPase was disrupted. Functional studies confirmed that active calcium absorption was significantly reduced in Cav1-/- compared to Cav1+/+ male mice. These results demonstrate that disruption of the Cav1 gene promotes the progressive steps required for urinary calcium stone formation and establish a new mouse model for urinary stone disease.


Subject(s)
Calcium Metabolism Disorders/pathology , Calcium/urine , Caveolins/genetics , Kidney/pathology , Urinary Calculi/pathology , Animals , Calcium/metabolism , Calcium Metabolism Disorders/genetics , Caveolin 1 , Caveolins/deficiency , Caveolins/physiology , Creatinine/urine , DNA Primers , Disease Models, Animal , Exons , Mice , Mice, Knockout , Polymerase Chain Reaction , Restriction Mapping , Urinary Calculi/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...