Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Natl Cancer Inst ; 104(22): 1750-64, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23150719

ABSTRACT

BACKGROUND: Birt-Hogg-Dubé (BHD) syndrome is a hereditary hamartoma syndrome that predisposes patients to develop hair follicle tumors, lung cysts, and kidney cancer. Genetic studies of BHD patients have uncovered the causative gene, FLCN, but its function is incompletely understood. METHODS: Mice with conditional alleles of FLCN and/or peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A), a transcriptional coactivator that regulates mitochondrial biogenesis, were crossbred with mice harboring either muscle creatine kinase (CKM) -Cre or myogenin (MYOG) -Cre transgenes to knock out FLCN and/or PPARGC1A in muscle, or cadherin 16 (CDH16)- Cre transgenes to knock out FLCN and/or PPARGC1A in kidney. Real-time polymerase chain reaction, immunoblotting, electron microscopy, and metabolic profiling assay were performed to evaluate mitochondrial biogenesis and function in muscle. Immunoblotting, electron microscopy, and histological analysis were used to investigate expression and the pathological role of PPARGC1A in FLCN-deficient kidney. Real-time polymerase chain reaction, oxygen consumption measurement, and flow cytometry were carried out using a FLCN-null kidney cancer cell line. All statistical analyses were two-sided. RESULTS: Muscle-targeted FLCN knockout mice underwent a pronounced metabolic shift toward oxidative phosphorylation, including increased mitochondrial biogenesis (FLCN ( f/f ) vs FLCN ( f/f ) /CKM-Cre: % mitochondrial area mean = 7.8% vs 17.8%; difference = 10.0%; 95% confidence interval = 5.7% to 14.3%; P < .001), and the observed increase in mitochondrial biogenesis was PPARGC1A dependent. Reconstitution of FLCN-null kidney cancer cells with wild-type FLCN suppressed mitochondrial metabolism and PPARGC1A expression. Kidney-targeted PPARGC1A inactivation partially rescued the enlarged kidney phenotype and abrogated the hyperplastic cells observed in the FLCN-deficient kidney. CONCLUSION: FLCN deficiency and subsequent increased PPARGC1A expression result in increased mitochondrial function and oxidative metabolism as the source of cellular energy, which may give FLCN-null kidney cells a growth advantage and drive hyperplastic transformation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Muscles/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Birt-Hogg-Dube Syndrome/genetics , Blotting, Western , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Mice, Knockout , Microscopy, Electron , Mitochondria/metabolism , Mitochondrial Turnover , Muscles/pathology , Oxidation-Reduction , Oxygen Consumption , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction , Trans-Activators/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
2.
Blood ; 120(6): 1254-61, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22709692

ABSTRACT

Birt-Hogg-Dubé (BHD) syndrome is an autosomal dominant disorder characterized by cutaneous fibrofolliculomas, pulmonary cysts, and kidney malignancies. Affected individuals carry germ line mutations in folliculin (FLCN), a tumor suppressor gene that becomes biallelically inactivated in kidney tumors by second-hit mutations. Similar to other factors implicated in kidney cancer, FLCN has been shown to modulate activation of mammalian target of rapamycin (mTOR). However, its precise in vivo function is largely unknown because germ line deletion of Flcn results in early embryonic lethality in animal models. Here, we describe mice deficient in the newly characterized folliculin-interacting protein 1 (Fnip1). In contrast to Flcn, Fnip1(-/-) mice develop normally, are not susceptible to kidney neoplasia, but display a striking pro-B cell block that is entirely independent of mTOR activity. We show that this developmental arrest results from rapid caspase-induced pre-B cell death, and that a Bcl2 transgene reconstitutes mature B-cell populations, respectively. We also demonstrate that conditional deletion of Flcn recapitulates the pro-B cell arrest of Fnip1(-/-) mice. Our studies thus demonstrate that the FLCN-FNIP complex deregulated in BHD syndrome is absolutely required for B-cell differentiation, and that it functions through both mTOR-dependent and independent pathways.


Subject(s)
B-Lymphocytes/physiology , Birt-Hogg-Dube Syndrome/genetics , Carrier Proteins/genetics , Cell Differentiation/genetics , Gene Deletion , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cell Differentiation/immunology , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Species Specificity , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology
3.
Proc Natl Acad Sci U S A ; 106(44): 18722-7, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19850877

ABSTRACT

Germline mutations in the BHD/FLCN tumor suppressor gene predispose patients to develop renal tumors in the hamartoma syndrome, Birt-Hogg-Dubé (BHD). BHD encodes folliculin, a protein with unknown function that may interact with the energy- and nutrient-sensing AMPK-mTOR signaling pathways. To clarify BHD function in the mouse, we generated a BHD knockout mouse model. BHD homozygous null (BHD(d/d)) mice displayed early embryonic lethality at E5.5-E6.5, showing defects in the visceral endoderm. BHD heterozygous knockout (BHDd(/+)) mice appeared normal at birth but developed kidney cysts and solid tumors as they aged (median kidney-lesion-free survival = 23 months, median tumor-free survival = 25 months). As observed in human BHD kidney tumors, three different histologic types of kidney tumors developed in BHD(d/+) mice including oncocytic hybrid, oncocytoma, and clear cell with concomitant loss of heterozygosity (LOH), supporting a tumor suppressor function for BHD in the mouse. The PI3K-AKT pathway was activated in both human BHD renal tumors and kidney tumors in BHD(d/+) mice. Interestingly, total AKT protein was elevated in kidney tumors compared to normal kidney tissue, but without increased levels of AKT mRNA, suggesting that AKT may be regulated by folliculin through post translational or post-transcriptional modification. Finally, BHD inactivation led to both mTORC1 and mTORC2 activation in kidney tumors from BHD(d/+) mice and human BHD patients. These data support a role for PI3K-AKT pathway activation in kidney tumor formation caused by loss of BHD and suggest that inhibitors of both mTORC1 and mTORC2 may be effective as potential therapeutic agents for BHD-associated kidney cancer.


Subject(s)
Embryo Loss/pathology , Homozygote , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Embryo Loss/genetics , Embryo, Mammalian/pathology , Embryonic Development , Enzyme Activation , Humans , Kidney Neoplasms/complications , Kidney Neoplasms/enzymology , Loss of Heterozygosity/genetics , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Proteins , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...