Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 112(13): E1624-31, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25775561

ABSTRACT

Folliculin (FLCN)-interacting proteins 1 and 2 (FNIP1, FNIP2) are homologous binding partners of FLCN, a tumor suppressor for kidney cancer. Recent studies have revealed potential functions for Flcn in kidney; however, kidney-specific functions for Fnip1 and Fnip2 are unknown. Here we demonstrate that Fnip1 and Fnip2 play critical roles in kidney tumor suppression in cooperation with Flcn. We observed no detectable phenotype in Fnip2 knockout mice, whereas Fnip1 deficiency produced phenotypes similar to those seen in Flcn-deficient mice in multiple organs, but not in kidneys. We found that absolute Fnip2 mRNA copy number was low relative to Fnip1 in organs that showed phenotypes under Fnip1 deficiency but was comparable to Fnip1 mRNA copy number in mouse kidney. Strikingly, kidney-targeted Fnip1/Fnip2 double inactivation produced enlarged polycystic kidneys, as was previously reported in Flcn-deficient kidneys. Kidney-specific Flcn inactivation did not further augment kidney size or cystic histology of Fnip1/Fnip2 double-deficient kidneys, suggesting pathways dysregulated in Flcn-deficient kidneys and Fnip1/Fnip2 double-deficient kidneys are convergent. Heterozygous Fnip1/homozygous Fnip2 double-knockout mice developed kidney cancer at 24 mo of age, analogous to the heterozygous Flcn knockout mouse model, further supporting the concept that Fnip1 and Fnip2 are essential for the tumor-suppressive function of Flcn and that kidney tumorigenesis in human Birt-Hogg-Dubé syndrome may be triggered by loss of interactions among Flcn, Fnip1, and Fnip2. Our findings uncover important roles for Fnip1 and Fnip2 in kidney tumor suppression and may provide molecular targets for the development of novel therapeutics for kidney cancer.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Alleles , Animals , Apoptosis Regulatory Proteins/genetics , Birt-Hogg-Dube Syndrome/genetics , Carrier Proteins/genetics , Disease Models, Animal , Female , Kidney/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Polycystic Kidney Diseases/metabolism , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
2.
Hum Mol Genet ; 23(21): 5706-19, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24908670

ABSTRACT

Cardiac hypertrophy, an adaptive process that responds to increased wall stress, is characterized by the enlargement of cardiomyocytes and structural remodeling. It is stimulated by various growth signals, of which the mTORC1 pathway is a well-recognized source. Here, we show that loss of Flcn, a novel AMPK-mTOR interacting molecule, causes severe cardiac hypertrophy with deregulated energy homeostasis leading to dilated cardiomyopathy in mice. We found that mTORC1 activity was upregulated in Flcn-deficient hearts, and that rapamycin treatment significantly reduced heart mass and ameliorated cardiac dysfunction. Phospho-AMP-activated protein kinase (AMPK)-alpha (T172) was reduced in Flcn-deficient hearts and nonresponsive to various stimulations including metformin and AICAR (5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide). ATP levels were elevated and mitochondrial function was increased in Flcn-deficient hearts, suggesting that excess energy resulting from up-regulated mitochondrial metabolism under Flcn deficiency might attenuate AMPK activation. Expression of Ppargc1a, a central molecule for mitochondrial metabolism, was increased in Flcn-deficient hearts and indeed, inactivation of Ppargc1a in Flcn-deficient hearts significantly reduced heart mass and prolonged survival. Ppargc1a inactivation restored phospho-AMPK-alpha levels and suppressed mTORC1 activity in Flcn-deficient hearts, suggesting that up-regulated Ppargc1a confers increased mitochondrial metabolism and excess energy, leading to inactivation of AMPK and activation of mTORC1. Rapamycin treatment did not affect the heart size of Flcn/Ppargc1a doubly inactivated hearts, further supporting the idea that Ppargc1a is the critical element leading to deregulation of the AMPK-mTOR-axis and resulting in cardiac hypertrophy under Flcn deficiency. These data support an important role for Flcn in cardiac homeostasis in the murine model.


Subject(s)
Cardiomegaly/genetics , Cardiomegaly/metabolism , Estrone/genetics , Gene Silencing , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Cardiomegaly/complications , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cell Line , Disease Models, Animal , Enzyme Activation , Heart Failure/etiology , Heart Failure/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Mitochondrial Turnover , Organ Size/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Signal Transduction , Sirolimus/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Ventricular Function/drug effects
3.
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
4.
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
5.
Mol Cancer ; 9: 160, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20573232

ABSTRACT

BACKGROUND: Germline mutations in the FLCN gene are responsible for the development of fibrofolliculomas, lung cysts and renal neoplasia in Birt-Hogg-Dube' (BHD) syndrome. The encoded protein folliculin (FLCN) is conserved across species but contains no classic motifs or domains and its function remains unknown. Somatic mutations or loss of heterozygosity in the remaining wild type copy of the FLCN gene have been found in renal tumors from BHD patients suggesting that FLCN is a classic tumor suppressor gene. RESULTS: To examine the tumor suppressor function of FLCN, wild-type or mutant FLCN (H255R) was stably expressed in a FLCN-null renal tumor cell line, UOK257, derived from a BHD patient. When these cells were injected into nude mice, tumor development was inversely dependent upon the level of wild-type FLCN expression. We identified genes that were differentially expressed in the cell lines with or without wild-type FLCN, many of which are involved in TGF-beta signaling, including TGF-beta2 (TGFB2), inhibin beta A chain (INHBA), thrombospondin 1 (THBS1), gremlin (GREM1), and SMAD3. In support of the in vitro data, TGFB2, INHBA, THBS1 and SMAD3 expression levels were significantly lower in BHD-associated renal tumors compared with normal kidney tissue. Although receptor mediated SMAD phosphorylation was not affected, basal and maximal TGF-beta-induced levels of TGFB2, INHBA and SMAD7 were dramatically reduced in FLCN-null cells compared with FLCN-restored cells. Secreted TGF-beta2 and activin A (homo-dimer of INHBA) protein levels were also lower in FLCN-null cells compared with FLCN-restored cells. Consistent with a growth suppressive function, activin A (but not TGF-beta2) completely suppressed anchorage-independent growth of FLCN-null UOK257 cells. CONCLUSIONS: Our data demonstrate a role for FLCN in the regulation of key molecules in TGF-beta signaling and confirm deregulation of their expression in BHD-associated renal tumors. Thus, deregulation of genes involved in TGF-beta signaling by FLCN inactivation is likely to be an important step for tumorigenesis in BHD syndrome.


Subject(s)
Gene Expression Regulation/physiology , Genes, Tumor Suppressor , Kidney Neoplasms/pathology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/physiology , Animals , Cell Line, Tumor , Genetic Vectors , Humans , Kidney Neoplasms/genetics , Lentivirus/genetics , Mice , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
6.
PLoS One ; 5(12): e15793, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21209915

ABSTRACT

BACKGROUND: Germline mutations in a tumor suppressor gene FLCN lead to development of fibrofolliculomas, lung cysts and renal cell carcinoma (RCC) in Birt-Hogg-Dubé syndrome. TFE3 is a member of the MiTF/TFE transcription factor family and Xp11.2 translocations found in sporadic RCC involving TFE3 result in gene fusions and overexpression of chimeric fusion proteins that retain the C-terminal DNA binding domain of TFE3. We found that GPNMB expression, which is regulated by MiTF, was greatly elevated in renal cancer cells harboring either TFE3 translocations or FLCN inactivation. Since TFE3 is implicated in RCC, we hypothesized that elevated GPNMB expression was due to increased TFE3 activity resulting from the inactivation of FLCN. METHODOLOGY/PRINCIPAL FINDINGS: TFE3 knockdown reduced GPNMB expression in renal cancer cells harboring either TFE3 translocations or FLCN inactivation. Moreover, FLCN knockdown induced GPNMB expression in FLCN-restored renal cancer cells. Conversely, wildtype FLCN suppressed GPNMB expression in FLCN-null cells. FLCN inactivation was correlated with increased TFE3 transcriptional activity accompanied by its nuclear localization as revealed by elevated GPNMB mRNA and protein expression, and predominantly nuclear immunostaining of TFE3 in renal cancer cells, mouse embryo fibroblast cells, mouse kidneys and mouse and human renal tumors. Nuclear localization of TFE3 was associated with TFE3 post-translational modifications including decreased phosphorylation. CONCLUSIONS/SIGNIFICANCE: Increased TFE3 activity is a downstream event induced by FLCN inactivation and is likely to be important for renal tumor development. This study provides an important novel mechanism for induction of TFE3 activity in addition to TFE3 overexpression resulting from Xp11.2 translocations, suggesting that TFE3 may be more broadly involved in tumorigenesis.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Nucleus/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Mutation , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Animals , Humans , Kidney/metabolism , Mice , Phosphorylation , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...