Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Urology ; 179: 58-70, 2023 09.
Article in English | MEDLINE | ID: mdl-37331486

ABSTRACT

OBJECTIVE: To characterize the clinical manifestations and genetic basis of a familial cancer syndrome in patients with lipomas and Birt-Hogg-Dubé-like clinical manifestations including fibrofolliculomas and trichodiscomas and kidney cancer. METHODS: Genomic analysis of blood and renal tumor DNA was performed. Inheritance pattern, phenotypic manifestations, and clinical and surgical management were documented. Cutaneous, subcutaneous, and renal tumor pathologic features were characterized. RESULTS: Affected individuals were found to be at risk for a highly penetrant and lethal form of bilateral, multifocal papillary renal cell carcinoma. Whole genome sequencing identified a germline pathogenic variant in PRDM10 (c.2029 T>C, p.Cys677Arg), which cosegregated with disease. PRDM10 loss of heterozygosity was identified in kidney tumors. PRDM10 was predicted to abrogate expression of FLCN, a transcriptional target of PRDM10, which was confirmed by tumor expression of GPNMB, a TFE3/TFEB target and downstream biomarker of FLCN loss. In addition, a sporadic papillary RCC from the TCGA cohort was identified with a somatic PRDM10 mutation. CONCLUSION: We identified a germline PRDM10 pathogenic variant in association with a highly penetrant, aggressive form of familial papillary RCC, lipomas, and fibrofolliculomas/trichodiscomas. PRDM10 loss of heterozygosity and elevated GPNMB expression in renal tumors indicate that PRDM10 alteration leads to reduced FLCN expression, driving TFE3-induced tumor formation. These findings suggest that individuals with Birt-Hogg-Dubé-like manifestations and subcutaneous lipomas, but without a germline pathogenic FLCN variant, should be screened for germline PRDM10 variants. Importantly, kidney tumors identified in patients with a pathogenic PRDM10 variant should be managed with surgical resection instead of active surveillance.


Subject(s)
Birt-Hogg-Dube Syndrome , Carcinoma, Renal Cell , Kidney Neoplasms , Lipoma , Skin Neoplasms , Humans , Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/genetics , Birt-Hogg-Dube Syndrome/complications , Birt-Hogg-Dube Syndrome/genetics , Birt-Hogg-Dube Syndrome/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Lipoma/complications , Lipoma/genetics , Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , DNA-Binding Proteins , Membrane Glycoproteins
2.
Hum Mutat ; 42(5): 520-529, 2021 05.
Article in English | MEDLINE | ID: mdl-33675279

ABSTRACT

Von Hippel-Lindau (VHL) is a hereditary multisystem disorder caused by germline alterations in the VHL gene. VHL patients are at risk for benign as well as malignant lesions in multiple organs including kidney, adrenal, pancreas, the central nervous system, retina, endolymphatic sac of the ear, epididymis, and broad ligament. An estimated 30%-35% of all families with VHL inherit a germline deletion of one, two, or all three exons. In this study, we have extensively characterized germline deletions identified in patients from 71 VHL families managed at the National Cancer Institute, including 59 partial (PD) and 12 complete VHL deletions (CD). Deletions that ranged in size from 1.09 to 355 kb. Fifty-eight deletions (55 PD and 3 CD) have been mapped to the exact breakpoints. Ninety-five percent (55 of 58) of mapped deletions involve Alu repeats at both breakpoints. Several novel classes of deletions were identified in this cohort, including two cases that have complex rearrangements involving both deletion and inversion, two cases with inserted extra Alu-like sequences, six cases that involve breakpoints in Alu repeats situated in opposite orientations, and a "hotspot" PD of Exon 3 observed in 12 families that involves the same pair of Alu repeats.


Subject(s)
von Hippel-Lindau Disease , Female , Gene Deletion , Germ Cells , Germ-Line Mutation , Humans , Male , Von Hippel-Lindau Tumor Suppressor Protein/genetics , von Hippel-Lindau Disease/genetics
3.
Hum Mol Genet ; 26(2): 354-366, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28007907

ABSTRACT

Germline H255Y and K508R missense mutations in the folliculin (FLCN) gene have been identified in patients with bilateral multifocal (BMF) kidney tumours and clinical manifestations of Birt-Hogg-Dubé (BHD) syndrome, or with BMF kidney tumours as the only manifestation; however, their impact on FLCN function remains to be determined. In order to determine if FLCN H255Y and K508R missense mutations promote aberrant kidney cell proliferation leading to pathogenicity, we generated mouse models expressing these mutants using BAC recombineering technology and investigated their ability to rescue the multi-cystic phenotype of Flcn-deficient mouse kidneys. Flcn H255Y mutant transgene expression in kidney-targeted Flcn knockout mice did not rescue the multi-cystic kidney phenotype. However, expression of the Flcn K508R mutant transgene partially, but not completely, abrogated the phenotype. Notably, expression of the Flcn K508R mutant transgene in heterozygous Flcn knockout mice resulted in development of multi-cystic kidneys and cardiac hypertrophy in some mice. These results demonstrate that both FLCN H255Y and K508R missense mutations promote aberrant kidney cell proliferation, but to different degrees. Based on the phenotypes of our preclinical models, the FLCN H255Y mutant protein has lost it tumour suppressive function leading to the clinical manifestations of BHD, whereas the FLCN K508R mutant protein may have a dominant negative effect on the function of wild-type FLCN in regulating kidney cell proliferation and, therefore, act as an oncoprotein. These findings may provide mechanistic insight into the role of FLCN in regulating kidney cell proliferation and facilitate the development of novel therapeutics for FLCN-deficient kidney cancer.


Subject(s)
Birt-Hogg-Dube Syndrome/genetics , Kidney Diseases, Cystic/genetics , Kidney Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Birt-Hogg-Dube Syndrome/pathology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Humans , Kidney/pathology , Kidney Diseases, Cystic/pathology , Kidney Neoplasms/pathology , Mice , Mice, Knockout , Mutation, Missense
4.
Hum Mutat ; 34(9): 1231-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23636849

ABSTRACT

Metastatic castration-resistant prostate cancer (mCRPC) is a lethal disease, and molecular markers that differentiate indolent from aggressive subtypes are needed. We sequenced the exomes of five metastatic tumors and healthy kidney tissue from an index case with mCRPC to identify lesions associated with disease progression and metastasis. An Ashkenazi Jewish (AJ) germline founder mutation, del185AG in BRCA1, was observed and AJ ancestry was confirmed. Sixty-two somatic variants altered proteins in tumors, including cancer-associated genes, TMPRSS2-ERG, PBRM1, and TET2. The majority (n = 53) of somatic variants were present in all metastases and only a subset (n = 31) was observed in the primary tumor. Integrating tumor next-generation sequencing and DNA copy number showed somatic loss of BRCA1 and TMPRSS2-ERG. We sequenced 19 genes with deleterious mutations in the index case in additional mCRPC samples and detected a frameshift, two somatic missense alterations, tumor loss of heterozygosity, and combinations of germline missense SNPs in TET2. In summary, genetic analysis of metastases from an index case permitted us to infer a chronology for the clonal spread of disease based on sequential accrual of somatic lesions. The role of TET2 in mCRPC deserves additional analysis and may define a subset of metastatic disease.


Subject(s)
DNA-Binding Proteins/genetics , Genes, BRCA1 , Neoplasm Metastasis/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Aged , Amino Acid Sequence , Dioxygenases , Frameshift Mutation , Germ-Line Mutation , Humans , Loss of Heterozygosity , Male , Middle Aged , Molecular Sequence Data , Mutation, Missense , Neoplasm Metastasis/pathology , Phylogeny , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
5.
J Urol ; 188(6): 2063-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23083876

ABSTRACT

PURPOSE: Recently, a new renal cell cancer syndrome has been linked to germline mutation of multiple subunits (SDHB/C/D) of the Krebs cycle enzyme, succinate dehydrogenase. We report our experience with the diagnosis, evaluation and treatment of this novel form of hereditary kidney cancer. MATERIALS AND METHODS: Patients with suspected hereditary kidney cancer were enrolled on a National Cancer Institute institutional review board approved protocol to study inherited forms of kidney cancer. Individuals from families with germline SDHB, SDHC and SDHD mutations, and kidney cancer underwent comprehensive clinical and genetic evaluation. RESULTS: A total of 14 patients from 12 SDHB mutation families were evaluated. Patients presented with renal cell cancer at an early age (33 years, range 15 to 62), metastatic kidney cancer developed in 4 and some families had no manifestation other than kidney tumors. An additional family with 6 individuals found to have clear cell renal cell cancer that presented at a young average age (47 years, range 40 to 53) was identified with a germline SDHC mutation (R133X) Metastatic disease developed in 2 of these family members. A patient with a history of carotid body paragangliomas and an aggressive form of kidney cancer was evaluated from a family with a germline SDHD mutation. CONCLUSIONS: SDH mutation associated renal cell carcinoma can be an aggressive type of kidney cancer, especially in younger individuals. Although detection and management of early tumors is most often associated with a good outcome, based on our initial experience with these patients and our long-term experience with hereditary leiomyomatosis and renal cell carcinoma, we recommend careful surveillance of patients at risk for SDH mutation associated renal cell carcinoma and wide surgical excision of renal tumors.


Subject(s)
Carcinoma, Renal Cell/genetics , Germ-Line Mutation , Kidney Neoplasms/genetics , Succinate Dehydrogenase/genetics , Adolescent , Adult , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/surgery , Disease Progression , Female , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/surgery , Genetic Testing , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/surgery , Male , Middle Aged , Pedigree , Young Adult
6.
Clin Cancer Res ; 14(15): 4726-34, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676741

ABSTRACT

PURPOSE: To provide a comprehensive, thorough analysis of somatic mutation and promoter hypermethylation of the von Hippel-Lindau (VHL) gene in the cancer genome, unique to clear cell renal cancer (ccRCC). Identify relationships between the prevalence of VHL gene alterations and alteration subtypes with patient and tumor characteristics. EXPERIMENTAL DESIGN: As part of a large kidney cancer case-control study conducted in Central Europe, we analyzed VHL mutations and promoter methylation in 205 well-characterized, histologically confirmed patient tumor biopsies using a combination of sensitive, high-throughput methods (endonuclease scanning and Sanger sequencing) and analysis of 11 CpG sites in the VHL promoter. RESULTS: We identified mutations in 82.4% of cases, the highest VHL gene mutation prevalence reported to date. Analysis of 11 VHL promoter CpG sites revealed that 8.3% of tumors were hypermethylated and all were mutation negative. In total, 91% of ccRCCs exhibited alteration of the gene through genetic or epigenetic mechanisms. Analysis of patient and tumor characteristics revealed that certain mutation subtypes were significantly associated with Fuhrman nuclear grade, metastasis, node positivity, and self-reported family history of RCC. CONCLUSION: Detection of VHL gene alterations using these accurate, sensitive, and practical methods provides evidence that the vast majority of histologically confirmed ccRCC tumors possess genetic or epigenetic alteration of the VHL gene and support the hypothesis that VHL alteration is an early event in ccRCC carcinogenesis. These findings also indicate that VHL molecular subtypes can provide a sensitive marker of tumor heterogeneity among histologically similar ccRCC cases for etiologic, prognostic, and translational studies.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Adenocarcinoma, Clear Cell/metabolism , Carcinoma, Renal Cell/metabolism , Case-Control Studies , CpG Islands , DNA Methylation , Female , Gene Expression Profiling , Humans , Male , Mutation , Neoplasms/metabolism , Promoter Regions, Genetic , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
7.
J Natl Cancer Inst ; 100(2): 140-54, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18182616

ABSTRACT

BACKGROUND: Patients with Birt-Hogg-Dubé (BHD) syndrome harbor germline mutations in the BHD tumor suppressor gene that are associated with an increased risk for kidney cancer. BHD encodes folliculin, a protein that may interact with the energy- and nutrient-sensing 5'-AMP-activated protein kinase-mammalian target of rapamycin (AMPK-mTOR) signaling pathways. METHODS: We used recombineering methods to generate mice with a conditional BHD allele and introduced the cadherin 16 (KSP)-Cre transgene to target BHD inactivation to the kidney. Kidney cell proliferation was measured by BrdU incorporation and phospho-histone H3 staining. Kidney weight data were analyzed with Wilcoxon's rank-sum, Student's t, and Welch's t tests. Hematoxylin and eosin staining and immunoblot analysis and immunohistochemistry of cell cycle and signaling proteins were performed on mouse kidney cells and tissues. BHD knockout mice and kidney cells isolated from BHD knockout and control mice were treated with the mTOR inhibitor rapamycin. Mouse survival was evaluated by Kaplan-Meier analyses. All statistical tests were two-sided. RESULTS: BHD knockout mice developed enlarged polycystic kidneys and died from renal failure by 3 weeks of age. Targeted BHD knockout led to the activation of Raf-extracellular signal-regulated protein kinase (Erk)1/2 and Akt-mTOR pathways in the kidneys and increased expression of cell cycle proteins and cell proliferation. Rapamycin-treated BHD knockout mice had smaller kidneys than buffer-treated BHD knockout mice had (n = 4-6 mice per group, relative kidney/body weight ratios, mean = 4.64% vs 12.2%, difference = 7.6%, 95% confidence interval = 5.2% to 10.0%; P < .001) and longer median survival time (n = 4-5 mice per group, 41.5 vs 23 days; P = .0065 ). CONCLUSIONS: Homozygous loss of BHD may initiate renal tumorigenesis in the mouse. The conditional BHD knockout mouse may be a useful research model for dissecting multistep kidney carcinogenesis, and rapamycin may be considered as a potential treatment for Birt-Hogg-Dubé syndrome.


Subject(s)
Cell Proliferation , Gene Silencing , Kidney Neoplasms/genetics , Kidney/metabolism , Kidney/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Polycystic Kidney Diseases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Blotting, Southern , Cell Proliferation/drug effects , Disease Models, Animal , Estrone/genetics , Fluorescent Antibody Technique , Genotype , Germ-Line Mutation , Immunoblotting , Immunohistochemistry , Kaplan-Meier Estimate , Kidney/drug effects , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Polycystic Kidney Diseases/complications , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Protein Kinases/drug effects , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sirolimus/pharmacology , Syndrome , TOR Serine-Threonine Kinases
8.
J Urol ; 177(6): 2074-9; discussion 2079-80, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17509289

ABSTRACT

PURPOSE: Hereditary leiomyomatosis and renal cell cancer is a recently described hereditary cancer syndrome in which affected individuals are at risk for cutaneous and uterine leiomyomas, and kidney cancer. Our initial experience revealed the aggressive behavior of these renal tumors, often with early metastasis, despite small primary tumor size. We report the clinical characteristics and urological treatment of patients with hereditary leiomyomatosis and renal cell cancer associated renal tumors. MATERIALS AND METHODS: A total of 19 patients with hereditary leiomyomatosis and renal cell cancer associated renal tumors were evaluated. The 11 women and 8 men had a median age at diagnosis of 39 years (range 22 to 67), and a median clinical and radiological followup of 34 months (range 6 to 141). Hereditary leiomyomatosis and renal cell cancer manifestations in patients with renal tumors included cutaneous leiomyomas in 11 of 17 evaluable patients (65%) and uterine leiomyomas in 7 of 7 evaluable females (100%). RESULTS: Median pathological tumor size was 7.8 cm (range 1.5 to 20). Histological subtypes were consistent with hereditary leiomyomatosis and renal cell cancer renal carcinoma. Four of 7 patients with 2.0 to 6.7 cm T1 tumors had spread to regional lymph nodes or metastases at nephrectomy. Overall 9 of 19 patients (47%) presented with nodal or distant metastases. CONCLUSIONS: Renal tumors in patients with hereditary leiomyomatosis and renal cell cancer syndrome are significantly more aggressive than those in patients with other hereditary renal tumor syndromes. In contrast to other familial renal cancer syndromes, the observation of 3 cm or less renal tumors associated with hereditary leiomyomatosis and renal cell cancer is not recommended. Careful followup of affected and at risk individuals in families is necessary.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Leiomyomatosis/pathology , Neoplastic Syndromes, Hereditary/pathology , Skin Neoplasms/pathology , Uterine Neoplasms/pathology , Adult , Aged , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/therapy , Cohort Studies , Female , Humans , Kidney Neoplasms/mortality , Kidney Neoplasms/therapy , Leiomyomatosis/mortality , Leiomyomatosis/therapy , Male , Middle Aged , Neoplastic Syndromes, Hereditary/mortality , Neoplastic Syndromes, Hereditary/therapy , Pedigree , Retrospective Studies , Skin Neoplasms/mortality , Skin Neoplasms/therapy , Survival Rate , Uterine Neoplasms/mortality , Uterine Neoplasms/therapy
9.
Am J Respir Crit Care Med ; 175(10): 1044-53, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17322109

ABSTRACT

RATIONALE: Birt-Hogg-Dubé syndrome (BHDS) is an autosomal, dominantly inherited genodermatosis that predisposes to fibrofolliculomas, kidney neoplasms, lung cysts, and spontaneous pneumothorax. OBJECTIVES: We evaluated 198 patients from 89 families with BHDS to characterize the risk factors for pneumothorax and genotype-pulmonary associations. METHODS: Helical computed tomography scans of the chest were used to screen for pulmonary abnormalities. BHD mutation data were used for genotype-pulmonary associations. We examined the relationship of pneumothorax with categorical parameters (sex, smoking history, and lung cysts) and continuous parameters (number of cysts, lung cyst volume, and largest cyst diameter and volume). Logistic regression analyses were used to identify the risk factors associated with pneumothorax. MEASUREMENTS AND MAIN RESULTS: Twenty-four percent (48/198) of patients with BHDS had a history of pneumothorax. The presence of lung cysts was significantly associated with pneumothorax (p = 0.006). Total lung cyst volume, largest cyst diameter and volume, and every parameter related to the number of lung cysts were significantly associated (p < 0.0001) with pneumothorax. A logistic regression analysis showed that only the total number of cysts in the right parenchymal lower lobe and the total number of cysts located on the pleural surface in the right middle lobe were needed to classify a patient as to whether or not he or she was likely to have a pneumothorax. Exon location of the BHD mutation was associated with the numbers of cysts (p = 0.0002). CONCLUSIONS: This study indicates that patients with BHDS have a significant association between lung cysts and spontaneous pneumothorax.


Subject(s)
Bronchogenic Cyst/epidemiology , Pneumothorax/epidemiology , Proteins/genetics , Proto-Oncogene Proteins/genetics , Skin Diseases/genetics , Tumor Suppressor Proteins/genetics , Bronchogenic Cyst/diagnosis , Bronchogenic Cyst/genetics , DNA Mutational Analysis , Exons , Female , Genetic Linkage , Genetic Testing , Genotype , Humans , Male , Mutation , Phenotype , Pneumothorax/diagnosis , Pneumothorax/genetics , Risk Factors , Skin Diseases/epidemiology , Syndrome
10.
J Urol ; 177(2): 461-5; discussion 465, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17222609

ABSTRACT

PURPOSE: Familial renal carcinoma is defined as families with 2 or more individuals with renal cell carcinoma without evidence of known hereditary renal carcinoma syndromes. To better characterize this familial cancer we reviewed renal carcinoma families evaluated at the National Cancer Institute between 1990 and 2004 to identify distinctive features of these families. We also determined the risk of renal carcinoma in first-degree relatives of affected family members. MATERIALS AND METHODS: We evaluated 141 at risk asymptomatic relatives of affected individuals from 50 families with 2 or more members with renal carcinoma. Histology slides of renal tumors from affected family members were reviewed. At risk members from renal carcinoma families were screened for occult renal neoplasms by renal ultrasound and computerized tomography. DNA from select families was tested for germline mutations of known renal carcinoma genes when clinically indicated and constitutional cytogenetic analysis was performed to search for germline chromosome alterations. RESULTS: Familial renal carcinoma families could be subdivided into subtypes based on tumor multiplicity and renal tumor histology. Of 141 at risk members of renal carcinoma families screened for occult renal tumors 2 were found to have occult renal tumors, which were identified as renal oncocytoma and a solid tumor that was not resected, respectively. No histologically confirmed occult renal carcinomas were detected in at risk family members. Several families previously classified as having familial renal carcinoma were found on further evaluation to have hereditary renal cancer syndromes. CONCLUSIONS: Familial renal carcinoma is a heterogeneous clinical and pathological entity. Familial renal carcinoma was subdivided into groups based on tumor multiplicity and tumor pathology. The empirical risk of histologically documented renal carcinoma in first-degree relatives who were members of familial renal carcinoma families was less than 1:141. One renal oncocytoma and 1 small solid renal tumor were detected.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/classification , Carcinoma, Renal Cell/diagnosis , Female , Humans , Kidney Neoplasms/classification , Kidney Neoplasms/diagnosis , Male , Pedigree , Risk Factors
11.
Proc Natl Acad Sci U S A ; 103(42): 15552-7, 2006 Oct 17.
Article in English | MEDLINE | ID: mdl-17028174

ABSTRACT

Birt-Hogg-Dubé syndrome, a hamartoma disorder characterized by benign tumors of the hair follicle, lung cysts, and renal neoplasia, is caused by germ-line mutations in the BHD(FLCN) gene, which encodes a tumor-suppressor protein, folliculin (FLCN), with unknown function. The tumor-suppressor proteins encoded by genes responsible for several other hamartoma syndromes, LKB1, TSC1/2, and PTEN, have been shown to be involved in the mammalian target of rapamycin (mTOR) signaling pathway. Here, we report the identification of the FLCN-interacting protein, FNIP1, and demonstrate its interaction with 5' AMP-activated protein kinase (AMPK), a key molecule for energy sensing that negatively regulates mTOR activity. FNIP1 was phosphorylated by AMPK, and its phosphorylation was reduced by AMPK inhibitors, which resulted in reduced FNIP1 expression. AMPK inhibitors also reduced FLCN phosphorylation. Moreover, FLCN phosphorylation was diminished by rapamycin and amino acid starvation and facilitated by FNIP1 overexpression, suggesting that FLCN may be regulated by mTOR and AMPK signaling. Our data suggest that FLCN, mutated in Birt-Hogg-Dubé syndrome, and its interacting partner FNIP1 may be involved in energy and/or nutrient sensing through the AMPK and mTOR signaling pathways.


Subject(s)
Carrier Proteins/metabolism , Multienzyme Complexes/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins , Proto-Oncogene Proteins , Signal Transduction/physiology , Tumor Suppressor Proteins , AMP-Activated Protein Kinases , Animals , Carrier Proteins/genetics , Cell Line , Cloning, Molecular , Enzyme Activation , Humans , Molecular Sequence Data , Multiprotein Complexes , Protein Binding , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Syndrome , TOR Serine-Threonine Kinases , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Lab Invest ; 86(7): 664-75, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16652107

ABSTRACT

Inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene leads to the development of central nervous system hemangioblastomas, pheochromocytomas and renal cell carcinomas. The biological role of the VHL gene during development is poorly understood because of early lethality of VHL-null embryos. To overcome early embryo lethality observed in the conventional knockout mouse, we introduced a tamoxifen-inducible Cre (CreER(TM)) transgene for the stage specific inactivation of the VHL gene. Acute tamoxifen-induced inactivation of the VHL gene at E10.5 resulted in embryonic lethality between E14.5 and E15.0 with extensive hemorrhage and necrosis, while littermate controls showed normal development. Examination of the VHL-inactivated embryos between E10.5 and E14.5 revealed dilated blood vessels, hemorrhage and necrotizing liver damage. Concomitant with severe hemorrhage and abnormal vasculature at E15.0, blood circulation in the yolk sac was impaired in the VHL-inactivated embryos, which may be the cause of embryo death. Placental development looked normal before embryo death (E14.5); however, at E16.5 following embryo death, we observed reduced growth of the placental labyrinthine layer. Inactivation of the VHL gene resulted in hypoxia-inducible factor (HIF)-1alpha stabilization and induction of its target genes, VEGF and CAIX, in mouse embryonic fibroblasts (MEFs). In addition, we observed lactate overproduction and acidification of culture media by the inactivation of the VHL gene. Thus, by using a novel conditional VHL knockout mouse model, we could show that the VHL gene plays an important role in the developing vasculature and liver during embryogenesis through regulation of HIF-1alpha and its target genes. This mouse model will be useful for the screening of anti-HIF or anti-VEGF drugs in vivo. Additionally, this acute VHL inactivation system may provide a useful tool for the in vivo study of genes that cause early embryonic lethality.


Subject(s)
Blood Vessels/abnormalities , Embryonic Development , Genes, Tumor Suppressor , Liver Diseases/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Cell Culture Techniques , Cells, Cultured , Culture Media/chemistry , Female , Fibroblasts/cytology , Fibroblasts/enzymology , Hydrogen-Ion Concentration , Integrases/genetics , Lactic Acid/analysis , Lactic Acid/biosynthesis , Mice , Mice, Transgenic , Pregnancy , Transgenes , Viral Proteins/genetics
13.
Cancer Cell ; 8(2): 143-53, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16098467

ABSTRACT

Individuals with hemizygous germline fumarate hydratase (FH) mutations are predisposed to renal cancer. These tumors predominantly exhibit functional inactivation of the remaining wild-type allele, implicating FH inactivation as a tumor-promoting event. Hypoxia-inducible factors are expressed in many cancers and are increased in clear cell renal carcinomas. Under normoxia, the HIFs are labile due to VHL-dependent proteasomal degradation, but stabilization occurs under hypoxia due to inactivation of HIF prolyl hydroxylase (HPH), which prevents HIF hydroxylation and VHL recognition. We demonstrate that FH inhibition, together with elevated intracellular fumarate, coincides with HIF upregulation. Further, we show that fumarate acts as a competitive inhibitor of HPH. These data delineate a novel fumarate-dependent pathway for regulating HPH activity and HIF protein levels.


Subject(s)
Carcinoma, Renal Cell/metabolism , DNA-Binding Proteins/metabolism , Fumarate Hydratase/genetics , Fumarates/metabolism , Kidney Neoplasms/metabolism , Leiomyomatosis/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Adult , Alleles , Basic Helix-Loop-Helix Transcription Factors , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Female , Fumarate Hydratase/antagonists & inhibitors , Fumarate Hydratase/metabolism , Fumarates/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Ketoglutaric Acids/pharmacology , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Male , Middle Aged , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Syndrome , Transcription Factors/analysis , Transcription Factors/genetics , Up-Regulation
14.
J Natl Cancer Inst ; 97(12): 931-5, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15956655

ABSTRACT

The Birt-Hogg-Dubé (BHD) syndrome is an inherited genodermatosis characterized by a predisposition to hamartomatous skin lesions, pulmonary cysts, and renal carcinoma. Seventy-seven renal tumors from 12 patients with germline BHD mutations were examined by DNA sequencing to identify somatic mutations in the second copy of BHD. Sequence alterations were detected in the majority of renal tumors (41 of 77, 53%), with loss of heterozygosity at the BHD locus in a minority of additional tumors (14 of 77, 17%). The somatic mutations were distributed across the entire gene, and the majority resulted in frameshifts that are predicted to truncate the BHD protein. These results support a role for BHD as a tumor suppressor gene that predisposes to the development of renal tumors when both copies are inactivated.


Subject(s)
Frameshift Mutation , Gene Frequency , Kidney Neoplasms/genetics , Proteins/genetics , Humans , Loss of Heterozygosity , Proto-Oncogene Proteins , Sequence Analysis, DNA , Tumor Suppressor Proteins
15.
J Urol ; 173(5): 1482-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15821464

ABSTRACT

PURPOSE: Herein we describe the evaluation and management of renal tumors in Birt-Hogg-Dubé (BHD), an autosomal dominant disorder predisposing to cutaneous fibrofolliculomas, pulmonary cysts, spontaneous pneumothorax and renal tumors. MATERIALS AND METHODS: A total of 124 affected individuals underwent comprehensive clinical evaluation, including body computerized tomography, to determine cutaneous, pulmonary and renal manifestations of BHD. Of these individuals 14 had their renal tumors managed at our institution. RESULTS: Of the 124 BHD affected individuals 34 (27%) had renal tumors of various histologies, most commonly hybrid oncocytic tumor and chromophobe renal carcinoma. Average age at renal tumor detection was 50.4 years and multiple tumors were found in a majority of patients. Some patients with renal tumors were identified that did not have the characteristic cutaneous hallmarks of BHD. In 4 of the 14 patients treated at our institution small (less than 3 cm) renal tumors were observed, while 10 others underwent a total of 12 renal procedures, including 4 radical and 8 partial nephrectomies. At a median of 38 months of followup 5 of these 10 patients remained free of disease, 3 had small renal tumors and 2 died of metastatic renal cancer. CONCLUSIONS: Patients with BHD are at risk for multiple renal tumors that are often malignant and can metastasize. Individuals at risk or affected by BHD should be radiographically screened for renal tumors at periodic intervals and they are best treated with nephron sparing surgical approaches. Genetic testing for this syndrome is now available.


Subject(s)
Kidney Neoplasms/diagnosis , Kidney Neoplasms/surgery , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/surgery , Adult , Aged , Female , Humans , Male , Middle Aged , Pedigree
16.
Am J Hum Genet ; 76(6): 1023-33, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15852235

ABSTRACT

Birt-Hogg-Dubé syndrome (BHD), a genodermatosis characterized by multiple hamartomas of the hair follicle (fibrofolliculoma), predisposes individuals to an increased risk of developing renal neoplasms and spontaneous pneumothorax. Previously, we localized the BHD locus (also known as FLCN) to chromosome 17p11.2 by linkage analysis and subsequently identified germline mutations in a novel gene in probands from eight of the nine families with BHD in our screening panel. Affected members of five of the families inherited an insertion/deletion of a cytosine in a C8 tract in exon 11. This mutation was also identified by exon 11 screening in probands from 22 of 52 additional families with BHD and therefore represents a hypermutable "hotspot" for mutation in BHD. Here, we screened the remaining 30 families from this large BHD cohort by direct sequence analysis and identified germline BHD mutations in 84% (51/61) of all families with BHD recruited to our study. Mutations were located along the entire length of the coding region, including 16 insertion/deletion, 3 nonsense, and 3 splice-site mutations. The majority of BHD mutations were predicted to truncate the BHD protein, folliculin. Among patients with a mutation in the exon 11 hotspot, significantly fewer renal tumors were observed in patients with the C-deletion than those with the C-insertion mutation. Coding-sequence mutations were not found, however, in probands from two large families with BHD whose affected members shared their family's BHD-affected haplotype. Of the 53 families with BHD whose members inherited either a germline mutation or the affected haplotype, 24 (45%) had at least one member with renal neoplasms. Three families classified with familial renal oncocytoma were identified with BHD mutations, which represents the first disease gene associated with this rare form of renal neoplasm. This study expands the BHD-mutation spectrum and evaluates genotype-phenotype correlations among families with BHD.


Subject(s)
Germ-Line Mutation , Phenotype , Proteins/genetics , Adenoma, Oxyphilic/genetics , Adenoma, Oxyphilic/pathology , Cohort Studies , Exons , Female , Gene Frequency , Genetic Testing , Haplotypes , Heterozygote , Humans , Introns , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Nuclear Family , Patient Selection , Pedigree , Proto-Oncogene Proteins , Retrospective Studies , Sequence Analysis, DNA , Syndrome , Tumor Suppressor Proteins
18.
J Urol ; 172(4 Pt 1): 1256-61, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15371818

ABSTRACT

PURPOSE: Hereditary papillary renal carcinoma (HPRC) is characterized by a predisposition to multiple, bilateral papillary type 1 renal tumors caused by inherited activating missense mutations in the tyrosine kinase domain of the MET proto-oncogene. In the current study we evaluated the clinical phenotype and germline MET mutation of 3 new HPRC families. We describe the early onset clinical features of HPRC. MATERIALS AND METHODS: We identified new HPRC families of Italian (family 177), Spanish (family 223) and Cuban (family 268) descent. We evaluated their clinical features, performed MET mutation analysis by denaturing high performance liquid chromatography and DNA sequencing, and estimated age dependent penetrance and survival using Kaplan-Meier analysis. We characterized renal tumors by histology and fluorescence in situ hybridization. RESULTS: Identical germline MET c.3522G --> A mutations (V1110I) were identified in families 177 and 268 but no evidence of a founder effect was found. Affected members of family 223 carried a germline c.3906G --> C.3522G --> A MET mutation (V1238I). Age dependent penetrance but not survival was significantly earlier for the c.3522G -->A mutation than for the c.3906G --> A mutation in these HPRC families. Trisomy of chromosome 7 and papillary renal carcinoma type 1 histology were detected in papillary renal tumors. CONCLUSIONS: HPRC can occur in an early onset form. The median age for renal tumor development in these 3 HPRC families was 46 to 63 years. HPRC associated papillary renal tumors may be aggressive and metastasize, leading to mortality. Median survival age was 60 to 70 years. Families with identical germline mutations in MET do not always share a common ancestor. HPRC is characterized by germline mutations in MET and papillary type 1 renal tumor histology.


Subject(s)
Adenocarcinoma, Papillary/genetics , Germ-Line Mutation , Kidney Neoplasms/genetics , Mutation, Missense , Neoplasms, Multiple Primary/genetics , Protein-Tyrosine Kinases/genetics , Proteins/genetics , Proto-Oncogene Proteins , Receptors, Growth Factor , Adenocarcinoma, Papillary/mortality , Adenocarcinoma, Papillary/pathology , Adult , Age Factors , Aged , Chromosomes, Human, Pair 7 , Exons , Female , Genetic Carrier Screening , Humans , Kidney/pathology , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasms, Multiple Primary/mortality , Neoplasms, Multiple Primary/pathology , Pedigree , Penetrance , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met , Survival Analysis , Trisomy , src Homology Domains/genetics
19.
Clin Cancer Res ; 10(18 Pt 2): 6282S-9S, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15448018

ABSTRACT

Studies during the past two decades have shown that kidney cancer is not a single disease; it is made up of a number of different types of cancer that occur in this organ. Clear cell renal carcinoma is characterized by mutation of the VHL gene. The VHL gene product forms a heterotrimeric complex with elongin C, elongin B, and Cul-2 to target hypoxia-inducible factors 1 and 2alpha for ubiquitin-mediated degradation. VHL-/- clear cell renal carcinoma overexpresses epidermal growth factor receptor and transforming growth factor alpha. Both hypoxia-inducible factor 1alpha and the epidermal growth factor receptor are potential therapeutic targets in clear cell renal carcinoma. Studies of the hereditary form of renal cell carcinoma (RCC) associated with hereditary papillary renal carcinoma (HPRC) determined that the c-Met proto-oncogene on chromosome 7 is the gene for HPRC and for a number of sporadic papillary RCCs. The HPRC c-Met mutations are activating mutations in the tyrosine kinase domain of the gene. The gene for a new form of hereditary RCC (Birt Hogg Dubé syndrome) associated with cutaneous tumors, lung cysts, and colon polyps or cancer has recently been identified. Studies are currently under way to determine what type of gene BHD is and how damage to this gene leads to kidney cancer. Individuals affected with hereditary leiomyomatosis renal cell carcinoma are at risk for the development of cutaneous leiomyomas, uterine leiomyomas (fibroids), and type 2 papillary RCC. The HLRC gene has been found to be the Krebs cycle enzyme, fumarate hydratase. Studies are under way to understand the downstream pathway of this cancer gene.


Subject(s)
Genetic Therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/pathology , Male , Proteins/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins , Von Hippel-Lindau Tumor Suppressor Protein , von Hippel-Lindau Disease/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...