Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Histopathology ; 76(3): 354-365, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31564060

ABSTRACT

AIMS: Hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome is caused by germline mutations in the Fumarate hydratase (FH) gene. In young women, the syndrome often presents with symptomatic uterine leiomyomas, leading to myomectomy or hysterectomy. In this study, we aimed to investigate the incidence and mutational profiles of FH-negative leiomyomas from young patients, thus allowing for early identification and triage of syndromic patients for surveillance. METHODS AND RESULTS: We evaluated 153 cases of uterine leiomyomas from women aged up to 30 years for loss of FH expression by tissue microarray (TMA)-based immunohistochemical staining. Mutational analysis of tumours with loss of FH was carried out by polymerase chain reaction (PCR) amplification of 10 exons within the FH gene and subsequent Sanger sequencing. The status of promoter methylation was assessed by bisulphite sequencing. Loss of FH protein expression was detected in seven (4.6%) of 153 tested uterine leiomyomas from young patients. All FH-negative leiomyomas displayed staghorn vasculature and fibrillary/neurophil-like cytoplasm. We found that six (86%) of seven FH-negative tumours detected by immunohistochemistry harboured FH mutations, 50% of which contained germline mutations. In particular, the germline mutational rate in FH gene was 2.0% (three of 153 cases). Bisulphite sequencing analysis failed to detect promoter methylation in any of the seven tumours. CONCLUSION: Our study showed a relatively high rate of FH germline mutation in FH-negative uterine leiomyomas from patients aged up to 30 years. While genetic mutations confer protein expression loss, epigenetic regulation of the FH gene appears to be unrelated to this phenotype.


Subject(s)
Fumarate Hydratase/genetics , Leiomyoma/genetics , Leiomyomatosis/genetics , Neoplastic Syndromes, Hereditary/genetics , Skin Neoplasms/genetics , Uterine Neoplasms/genetics , Adolescent , Adult , DNA Mutational Analysis , Female , Fumarate Hydratase/metabolism , Germ-Line Mutation , Humans , Immunohistochemistry , Leiomyoma/enzymology , Leiomyoma/pathology , Leiomyomatosis/enzymology , Leiomyomatosis/pathology , Mutation , Neoplastic Syndromes, Hereditary/enzymology , Neoplastic Syndromes, Hereditary/pathology , Prevalence , Retrospective Studies , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Tissue Array Analysis , Uterine Neoplasms/enzymology , Uterine Neoplasms/pathology , Young Adult
2.
Cancer Sci ; 109(9): 2757-2766, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29917289

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a hereditary cancer syndrome characterized by inactivation of the Krebs cycle enzyme fumarate hydratase (FH). HLRCC patients are at high risk of developing kidney cancer of type 2 papillary morphology that is refractory to current radiotherapy, immunotherapy and chemotherapy. Hence, an effective therapy for this deadly form of cancer is urgently needed. Here, we show that FH inactivation (FH-/- ) proves synthetic lethal with inducers of ferroptosis, an iron-dependent and nonapoptotic form of cell death. Specifically, we identified gene signatures for compound sensitivities based on drug responses for 9 different drug classes against the NCI-60 cell lines. These signatures predicted that ferroptosis inducers would be selectively toxic to FH-/- cell line UOK262. Preferential cell death against UOK262-FH-/- was confirmed with 4 different ferroptosis inducers. Mechanistically, the FH-/- sensitivity to ferroptosis is attributed to dysfunctional GPX4, the primary cellular defender against ferroptosis. We identified that C93 of GPX4 is readily post-translationally modified by fumarates that accumulate in conditions of FH-/- , and that C93 modification represses GPX4 activity. Induction of ferroptosis in FH-inactivated tumors represents an opportunity for synthetic lethality in cancer.


Subject(s)
Fumarate Hydratase/physiology , Leiomyomatosis/enzymology , Neoplastic Syndromes, Hereditary/enzymology , Skin Neoplasms/enzymology , Uterine Neoplasms/enzymology , Cell Line, Tumor , Enzyme Activation , Glutathione Peroxidase/physiology , Humans , Leiomyomatosis/pathology , Neoplastic Syndromes, Hereditary/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase , Reactive Oxygen Species/metabolism , Skin Neoplasms/pathology , Uterine Neoplasms/pathology
4.
Nature ; 477(7363): 225-8, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849978

ABSTRACT

Fumarate hydratase (FH) is an enzyme of the tricarboxylic acid cycle (TCA cycle) that catalyses the hydration of fumarate into malate. Germline mutations of FH are responsible for hereditary leiomyomatosis and renal-cell cancer (HLRCC). It has previously been demonstrated that the absence of FH leads to the accumulation of fumarate, which activates hypoxia-inducible factors (HIFs) at normal oxygen tensions. However, so far no mechanism that explains the ability of cells to survive without a functional TCA cycle has been provided. Here we use newly characterized genetically modified kidney mouse cells in which Fh1 has been deleted, and apply a newly developed computer model of the metabolism of these cells to predict and experimentally validate a linear metabolic pathway beginning with glutamine uptake and ending with bilirubin excretion from Fh1-deficient cells. This pathway, which involves the biosynthesis and degradation of haem, enables Fh1-deficient cells to use the accumulated TCA cycle metabolites and permits partial mitochondrial NADH production. We predicted and confirmed that targeting this pathway would render Fh1-deficient cells non-viable, while sparing wild-type Fh1-containing cells. This work goes beyond identifying a metabolic pathway that is induced in Fh1-deficient cells to demonstrate that inhibition of haem oxygenation is synthetically lethal when combined with Fh1 deficiency, providing a new potential target for treating HLRCC patients.


Subject(s)
Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Genes, Lethal/genetics , Genes, Tumor Suppressor , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Mutation/genetics , Animals , Bilirubin/metabolism , Cell Line , Cells, Cultured , Citric Acid Cycle , Computer Simulation , Fumarate Hydratase/deficiency , Fumarates/metabolism , Glutamine/metabolism , Heme/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Leiomyomatosis/congenital , Leiomyomatosis/drug therapy , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Mice , Mitochondria/metabolism , NAD/metabolism , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms
5.
Am J Dermatopathol ; 38(12): 887-891, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27097334

ABSTRACT

AIMS: Hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome is an autosomal dominant disorder caused by heterozygotic germline mutations in fumarate hydratase (FH) with incomplete penetrance, and clinically challenging to diagnose. Immunohistochemical stainings may favor an earlier diagnosis. METHODS AND RESULTS: The authors have tested 31 smooth muscle neoplasms. Ten of the 13 lesions from patients with HLRCC syndrome showed negative FH staining. Most sporadic piloleiomyomas presented strongly positive FH staining although 5 cases were negative. Sensitivity of FH staining in our series is 83.3% but specificity is 75%. Anti-S-(2-succino)-cysteine (2SC) showed the opposite intensity staining pattern and showed great correlation with anti-FH (rho spearman = -0.797). Anti-2SC staining increased the diagnostic accuracy in 19% of the cases. LIMITATIONS: The main limitation of this study is the lack additional clinical data to further classify the cases as the case inclusion was histopathological. CONCLUSIONS: Negative FH staining could indicate a high risk of HLRCC but it could also suggest the presence of a syndrome in up to 25% of sporadic cases. Thus, when there is a doubtful case, anti-2SC may be added to exclude the syndrome if a negative staining is found.


Subject(s)
Biomarkers, Tumor/analysis , Cysteine/analogs & derivatives , Fumarate Hydratase/analysis , Immunohistochemistry , Leiomyomatosis/enzymology , Protein Processing, Post-Translational , Skin Neoplasms/enzymology , Uterine Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Cysteine/analysis , DNA Mutational Analysis , Down-Regulation , Early Detection of Cancer , Female , Fumarate Hydratase/genetics , Humans , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Male , Middle Aged , Mutation , Neoplastic Syndromes, Hereditary , Predictive Value of Tests , Reproducibility of Results , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
6.
Invest Clin ; 57(1): 59-65, 2016 Mar.
Article in Spanish | MEDLINE | ID: mdl-27382802

ABSTRACT

We report the case of a 56 year-old male with an atypical leiomyoma in the context of a cutaneous leiomyomatosis and a family history of uterine leiomyomatosis. The genetic study revealed a mutation in the gene for the enzyme fumarate hydratase, but he has not had any renal malignancy so far. Atypical leiomyoma is a rare tumor that usually presents as a single lesion and is exceptional in patients with cutaneous leiomyomatosis. The relation between fumarate hydratase enzyme mutations with multiple leiomyomas, uterine leiomyomatosis and an increased risk of developing kidney cancer is widely known. However, the role of these mutations in the development of atypical leiomyomas is still impossible to clarify given the few cases reported in the literature.


Subject(s)
Fumarate Hydratase/genetics , Leiomyomatosis/genetics , Mutation , Skin Neoplasms/genetics , Humans , Leiomyoma/enzymology , Leiomyoma/genetics , Leiomyoma/pathology , Leiomyomatosis/enzymology , Leiomyomatosis/pathology , Male , Middle Aged , Skin Neoplasms/enzymology , Skin Neoplasms/pathology
7.
Cancer Cell ; 11(4): 303-5, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17418405

ABSTRACT

Molecular genetic analysis of hereditary leiomyomatosis and renal cell cancer (HLRCC) unexpectedly revealed germline defects in the gene encoding the Krebs cycle enzyme fumarate hydratase (FH), stimulating great interest in the underlying mechanism of oncogenesis. It has been proposed that the associated accumulation of fumarate competitively inhibits the 2-oxoglutarate-dependent dioxygenases that regulate hypoxia-inducible factor (HIF), thus activating oncogenic hypoxia pathways. In this issue of Cancer Cell, Pollard and colleagues describe a genetic mouse model of FH deficiency that recapitulates aspects of the human disease, including HIF activation and renal cysts, enabling further insights into this unusual cancer syndrome.


Subject(s)
Fumarate Hydratase/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/etiology , Leiomyomatosis/etiology , Animals , Disease Models, Animal , Fumarate Hydratase/genetics , Germ-Line Mutation , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Mice , Neoplastic Syndromes, Hereditary/enzymology , Neoplastic Syndromes, Hereditary/etiology , Neoplastic Syndromes, Hereditary/genetics , Signal Transduction
8.
Dermatol Online J ; 21(10)2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26632803

ABSTRACT

Reed syndrome is an autosomal dominant disorder characterized by cutaneous leiomyomas, uterine leiomyomas, and renal cell carcinoma caused by mutations in the fumarate hydratase gene. Dermatologic evaluation is often the first or only opportunity to discover the diagnosis of Reed syndrome in affected patients, which may prove to be life-saving. We present a 40-year-old woman with history of large uterine leiomyomas who presented with a two-year history of a pruritic papular eruption on the left neck refractory to topical corticosteroids. After histopathologic examination and genetic work-up, the patient was found to have a novel mutation in the fumarate hydratase gene and was subsequently diagnosed with Reed syndrome.


Subject(s)
DNA, Neoplasm/genetics , Fumarate Hydratase/genetics , Leiomyomatosis/genetics , Mutation , Skin Neoplasms/genetics , Uterine Neoplasms/genetics , Adult , Biopsy , DNA Mutational Analysis , Female , Fumarate Hydratase/metabolism , Humans , Leiomyomatosis/enzymology , Leiomyomatosis/pathology , Neoplastic Syndromes, Hereditary , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Uterine Neoplasms/enzymology , Uterine Neoplasms/pathology
9.
PLoS Biol ; 8(3): e1000328, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20231875

ABSTRACT

In eukaryotes, fumarase (FH in human) is a well-known tricarboxylic-acid-cycle enzyme in the mitochondrial matrix. However, conserved from yeast to humans is a cytosolic isoenzyme of fumarase whose function in this compartment remains obscure. A few years ago, FH was surprisingly shown to underlie a tumor susceptibility syndrome, Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC). A biallelic inactivation of FH has been detected in almost all HLRCC tumors, and therefore FH was suggested to function as a tumor suppressor. Recently it was suggested that FH inhibition leads to elevated intracellular fumarate, which in turn acts as a competitive inhibitor of HPH (HIF prolyl hydroxylase), thereby causing stabilization of HIF (Hypoxia-inducible factor) by preventing proteasomal degradation. The transcription factor HIF increases the expression of angiogenesis regulated genes, such as VEGF, which can lead to high microvessel density and tumorigenesis. Yet this mechanism does not fully explain the large cytosolic population of fumarase molecules. We constructed a yeast strain in which fumarase is localized exclusively to mitochondria. This led to the discovery that the yeast cytosolic fumarase plays a key role in the protection of cells from DNA damage, particularly from DNA double-strand breaks. We show that the cytosolic fumarase is a member of the DNA damage response that is recruited from the cytosol to the nucleus upon DNA damage induction. This function of fumarase depends on its enzymatic activity, and its absence in cells can be complemented by high concentrations of fumaric acid. Our findings suggest that fumarase and fumaric acid are critical elements of the DNA damage response, which underlies the tumor suppressor role of fumarase in human cells and which is most probably HIF independent. This study shows an exciting crosstalk between primary metabolism and the DNA damage response, thereby providing a scenario for metabolic control of tumor propagation.


Subject(s)
Cell Nucleus/metabolism , Cytosol/metabolism , DNA Damage , Fumarate Hydratase/metabolism , Isoenzymes/metabolism , Mitochondria/enzymology , Fumarate Hydratase/genetics , Fumarates/metabolism , Gene Knockdown Techniques , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoenzymes/genetics , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
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
11.
Nat Rev Cancer ; 3(3): 193-202, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12612654

ABSTRACT

Mitochondrial defects have been associated with neurological disorders, as well as cancers. Two ubiquitously expressed mitochondrial enzymes--succinate dehydrogenase (SDH) and fumarate hydratase (FH, fumarase)--catalyse sequential steps in the Krebs tricarboxylic-acid cycle. Inherited heterozygous mutations in the genes encoding these enzymes cause predispositions to two types of inherited neoplasia syndromes that do not share any component tumours. Homozygous mutations in the same genes result in severe neurological impairment. Understanding this link between inherited cancer syndromes and neurological disease could provide further insights into the mechanisms by which mitochondrial deficiencies lead to tumour development.


Subject(s)
Citric Acid Cycle/genetics , Fumarate Hydratase/physiology , Mitochondria/enzymology , Mitochondrial Encephalomyopathies/enzymology , Multienzyme Complexes/physiology , Neoplastic Syndromes, Hereditary/enzymology , Oxidoreductases/physiology , Succinate Dehydrogenase/physiology , Apoptosis/genetics , Apoptosis/physiology , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , Electron Transport Complex II , Energy Metabolism , Forecasting , Free Radicals , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Heterozygote , Homozygote , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Mitochondrial Encephalomyopathies/genetics , Multienzyme Complexes/deficiency , Multienzyme Complexes/genetics , Mutation , Neoplastic Syndromes, Hereditary/genetics , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Oxidoreductases/deficiency , Oxidoreductases/genetics , Paraganglioma/enzymology , Paraganglioma/genetics , Pheochromocytoma/enzymology , Pheochromocytoma/genetics , Protein Subunits , Succinate Dehydrogenase/deficiency , Succinate Dehydrogenase/genetics
12.
Clin Genet ; 79(1): 49-59, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20618355

ABSTRACT

Heterozygous fumarate hydratase (FH) germline mutations cause hereditary leiomyomatosis and renal cell cancer (HLRCC), an autosomal dominant syndrome characterized by multiple cutaneous piloleiomyomas, uterine leiomyomas and papillary type 2 renal cancer. The main objective of our study was to evaluate clinical and genetic data from families suspected of HLRCC on a nationwide level. All families referred for FH mutation analysis in the Netherlands were assessed. We performed FH sequence analysis and multiplex ligation-dependent probe amplification. Families with similar FH mutations were examined for haplotype sharing. In 14 out of 33 families, we identified 11 different pathogenic FH germline mutations, including 4 novel mutations and 1 whole-gene deletion. Clinical data were available for 35 FH mutation carriers. Cutaneous leiomyomas were present in all FH mutation carriers older than 40 years of age. Eleven out of 21 female FH mutation carriers underwent surgical treatment for symptomatic uterine leiomyomas at an average of 35 years. Two FH mutation carriers had papillary type 2 renal cancer and Wilms' tumour, respectively. We evaluated the relevance of our findings for clinical practice and have proposed clinical diagnostic criteria, indications for FH mutation analysis and recommendations for management.


Subject(s)
Carcinoma, Renal Cell/genetics , Fumarate Hydratase , Germ-Line Mutation , Kidney Neoplasms/genetics , Leiomyomatosis , Skin Neoplasms/genetics , Uterine Neoplasms/genetics , Adolescent , Adult , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/enzymology , Child , Child, Preschool , DNA Mutational Analysis , Female , Fumarate Hydratase/genetics , Genetic Predisposition to Disease , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/enzymology , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Netherlands , Pedigree , Skin Neoplasms/diagnosis , Skin Neoplasms/enzymology , Syndrome , Uterine Neoplasms/diagnosis , Uterine Neoplasms/enzymology , Young Adult
13.
J Clin Pathol ; 74(10): 615-619, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34353877

ABSTRACT

Fumarate hydratase (FH), encoded by the FH gene, is an enzyme which catalyses the conversion of fumarate to L-malate as part of the tricarboxylic acid cycle. Biallelic germline mutations in FH result in fumaric aciduria, a metabolic disorder resulting in severe neurological and developmental abnormalities. Heterozygous germline mutations in FH result in hereditary leiomyomatosis and renal cell carcinoma, a cancer predisposition syndrome. FH deficiency has multiple oncogenic mechanisms including through promotion of aerobic glycolysis, induction of pseudohypoxia, post-translational protein modification and impairment of DNA damage repair by homologous recombination. FH-deficient neoplasms can present with characteristic morphological features that raise suspicion for FH alterations and also frequently demonstrate loss of FH immunoreactivity and intracellular accumulation of 2-succinocysteine, also detected by immunohistochemistry.


Subject(s)
Fumarate Hydratase/deficiency , Fumarate Hydratase/metabolism , Metabolism, Inborn Errors/enzymology , Muscle Hypotonia/enzymology , Neoplasms/enzymology , Psychomotor Disorders/enzymology , Animals , Fumarate Hydratase/genetics , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , Humans , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Metabolism, Inborn Errors/genetics , Muscle Hypotonia/genetics , Mutation , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Syndromes, Hereditary/enzymology , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Phenotype , Psychomotor Disorders/genetics , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Uterine Neoplasms/enzymology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
14.
Sci Signal ; 14(664)2021 01 05.
Article in English | MEDLINE | ID: mdl-33402335

ABSTRACT

Understanding the mechanisms of the Warburg shift to aerobic glycolysis is critical to defining the metabolic basis of cancer. Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is an aggressive cancer characterized by biallelic inactivation of the gene encoding the Krebs cycle enzyme fumarate hydratase, an early shift to aerobic glycolysis, and rapid metastasis. We observed impairment of the mitochondrial respiratory chain in tumors from patients with HLRCC. Biochemical and transcriptomic analyses revealed that respiratory chain dysfunction in the tumors was due to loss of expression of mitochondrial DNA (mtDNA)-encoded subunits of respiratory chain complexes, caused by a marked decrease in mtDNA content and increased mtDNA mutations. We demonstrated that accumulation of fumarate in HLRCC tumors inactivated the core factors responsible for replication and proofreading of mtDNA, leading to loss of respiratory chain components, thereby promoting the shift to aerobic glycolysis and disease progression in this prototypic model of glucose-dependent human cancer.


Subject(s)
Carcinoma, Renal Cell/genetics , Citric Acid Cycle , DNA Damage , DNA, Mitochondrial/metabolism , Fumarate Hydratase/genetics , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Neoplastic Syndromes, Hereditary/enzymology , Skin Neoplasms/enzymology , Uterine Neoplasms/enzymology , Adult , Aged , Carcinoma, Renal Cell/etiology , Carcinoma, Renal Cell/metabolism , DNA Repair , DNA Replication , Female , Fumarate Hydratase/deficiency , Gene Expression Profiling , Humans , Kidney Neoplasms/etiology , Kidney Neoplasms/metabolism , Leiomyomatosis/complications , Male , Middle Aged , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Neoplastic Syndromes, Hereditary/complications , Skin Neoplasms/complications , Uterine Neoplasms/complications , Young Adult
15.
Br J Dermatol ; 163(6): 1337-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20560959

ABSTRACT

Multiple cutaneous and uterine leiomyomatosis (MCUL)/hereditary leiomyomatosis and renal cell cancer (HLRCC) (OMIM 150800/OMIM 605839) is a rare hereditary disorder leading to the development of benign cutaneous and uterine smooth muscle tumours in young adults.(1,2) This disease is characterized by an increased risk of developing renal cell carcinomas.(3) It results from dominantly inherited autosomal mutations in the fumarate hydratase (FH) gene.(4) This gene encodes a Krebs cycle enzyme, present in both cytosolic and mitochondrial compartments, and probably acts as a tumour suppressor gene. We report a 22-year-old man affected by cutaneous leiomyomatosis associated with cutis verticis gyrata, disseminated collagenoma and Charcot-Marie-Tooth disease, who was harbouring the novel FH gene mutation c.821C > T, p.Ala274Val.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Fumarate Hydratase/genetics , Leiomyomatosis/genetics , Mutation , Skin Diseases/genetics , Skin Neoplasms/genetics , Carcinoma, Renal Cell/genetics , Collagen Diseases/genetics , Genetic Predisposition to Disease , Humans , Leiomyomatosis/enzymology , Leiomyomatosis/pathology , Male , Scalp Dermatoses/genetics , Sequence Analysis, DNA , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Young Adult
16.
Eur J Dermatol ; 19(6): 545-51, 2009.
Article in English | MEDLINE | ID: mdl-19939761

ABSTRACT

Multiple cutaneous and uterine leiomyomatosis syndrome (MCUL; OMIM 150800) is an autosomal dominantly inherited tumor predisposition disorder, characterized by leiomyomas of the skin and uterus. When associated with kidney cancer, this syndrome is known as hereditary leiomyomatosis and renal cell cancer (HLRCC; OMIM 605839). All disease variants result from heterozygous mutations in the fumarate hydratase (FH) gene. Cutaneous leiomyoma can easily be recognized and confirmed by histological examination. Recognition of these benign skin tumors can lead to the diagnosis of MCUL or HLRCC. Timely diagnosis is crucial for offering affected individuals and families potentially life-saving regular prophylactic screening examinations for renal tumors. Here we provide an overview of clinical and genetic features of this complex tumor syndrome and discuss patient management and current therapeutic strategies.


Subject(s)
Fumarate Hydratase/genetics , Leiomyomatosis/genetics , Mutation , Biomarkers/metabolism , Biopsy , Diagnosis, Differential , Female , Fumarate Hydratase/deficiency , Genetic Counseling , Genetic Predisposition to Disease , Humans , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Leiomyomatosis/pathology , Leiomyomatosis/therapy , Metabolism, Inborn Errors/genetics , Skin Neoplasms/genetics , Uterine Neoplasms/genetics
17.
Am J Surg Pathol ; 43(5): 639-655, 2019 05.
Article in English | MEDLINE | ID: mdl-30741757

ABSTRACT

Pathology-based screening of uterine smooth muscle tumors (uSMT) for morphology suggestive of fumarate hydratase deficiency (FH-d morphology) has been proposed as a method to identify women at increased risk for hereditary leiomyomatosis renal cell carcinoma (HLRCC) syndrome. For 5 years our clinical diagnostic practice has evaluated all women with any type of uSMT for FH-d morphology (defined, at low magnification, as staghorn shaped blood vessels and alveolar pattern edema and, at high magnification, as tumor macronucleoli surrounded by a halo and cytoplasmic eosinophilic globules) and, when present, used the pathology report to advise genetic counseling to further evaluate for HLRCC syndrome. We now report the results of this prospective screening strategy, with emphasis on the incidence and clinicopathologic features of FH-d morphology in uSMT, the rate of patient uptake of referral to genetic counseling, and the results of genetic testing for FH germline mutation. Among 2060 women with a uSMT, FH-d morphology was reported in 1.4% (30 women). Ten women elected to undergo FH genetic testing and 6 of 10 (60%) had a FH germline mutation: 5 were pathogenic mutations and 1 was a mutation variant of unknown significance. Therefore, the screening program led to a confirmed genetic diagnosis of HLRCC syndrome in 0.24% of all women with any type of uSMT. The women with a pathogenic mutation were ages 24 to 40 years. Although the majority of leiomyoma with bizarre nuclei exhibited FH-d morphology, the uSMT were conventional leiomyomas with FH-d morphology in 2 of 5 women found to have a pathogenic FH germline mutation. Relying on an abnormal FH immunostain result to trigger genetic counseling referral would have resulted in 2 of 5 (40%) cases with pathogenic FH germline mutation but normal FH immunoexpression going undetected, both of which were missense type mutations. There was no difference in the incidence of pathogenic FH germline mutation between FH-d morphology uSMT with an abnormal versus a normal FH immunostain result. Overall, this study demonstrates that prospective morphology-based screening, integrated with referral for genetic counseling, can result in the diagnosis of HLRCC syndrome in otherwise unselected women with uSMT. We conclude that this strategy should be incorporated in the routine pathologic examination of all uterine smooth muscle tumors.


Subject(s)
Biomarkers, Tumor/genetics , DNA Mutational Analysis , Fumarate Hydratase/genetics , Genetic Counseling , Germ-Line Mutation , Leiomyomatosis/genetics , Neoplastic Syndromes, Hereditary/genetics , Skin Neoplasms/genetics , Uterine Neoplasms/genetics , Adult , Aged , Biopsy , Female , Genetic Predisposition to Disease , Heredity , Humans , Leiomyomatosis/enzymology , Leiomyomatosis/pathology , Leiomyomatosis/therapy , Middle Aged , Neoplastic Syndromes, Hereditary/enzymology , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/therapy , Patient Acceptance of Health Care , Pedigree , Phenotype , Predictive Value of Tests , Prognosis , Prospective Studies , Referral and Consultation , San Francisco , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Time Factors , Uterine Neoplasms/enzymology , Uterine Neoplasms/pathology , Uterine Neoplasms/therapy , Young Adult
18.
BMC Med Genet ; 9: 20, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18366737

ABSTRACT

BACKGROUND: Fumarate hydratase (HGNC approved gene symbol - FH), also known as fumarase, is an enzyme of the tricarboxylic acid (TCA) cycle, involved in fundamental cellular energy production. First described by Zinn et al in 1986, deficiency of FH results in early onset, severe encephalopathy. In 2002, the Multiple Leiomyoma Consortium identified heterozygous germline mutations of FH in patients with multiple cutaneous and uterine leiomyomas, (MCUL: OMIM 150800). In some families renal cell cancer also forms a component of the complex and as such has been described as hereditary leiomyomatosis and renal cell cancer (HLRCC: OMIM 605839). The identification of FH as a tumor suppressor was an unexpected finding and following the identification of subunits of succinate dehydrogenase in 2000 and 2001, was only the second description of the involvement of an enzyme of intermediary metabolism in tumorigenesis. DESCRIPTION: The FH mutation database is a part of the TCA cycle gene mutation database (formerly the succinate dehydrogenase gene mutation database) and is based on the Leiden Open (source) Variation Database (LOVD) system. The variants included in the database were derived from the published literature and annotated to conform to current mutation nomenclature. The FH database applies HGVS nomenclature guidelines, and will assist researchers in applying these guidelines when directly submitting new sequence variants online. Since the first molecular characterization of an FH mutation by Bourgeron et al in 1994, a series of reports of both FH deficiency patients and patients with MCUL/HLRRC have described 107 variants, of which 93 are thought to be pathogenic. The most common type of mutation is missense (57%), followed by frameshifts & nonsense (27%), and diverse deletions, insertions and duplications. Here we introduce an online database detailing all reported FH sequence variants. CONCLUSION: The FH mutation database strives to systematically unify all current genetic knowledge of FH variants. We believe that this knowledge will assist clinical geneticists and treating physicians when advising patients and their families, will provide a rapid and convenient resource for research scientists, and may eventually assist in gaining novel insights into FH and its related clinical syndromes.


Subject(s)
Databases, Nucleic Acid , Fumarate Hydratase/genetics , Germ-Line Mutation , Leiomyomatosis/genetics , Metabolism, Inborn Errors/genetics , Skin Neoplasms/genetics , Uterine Neoplasms/genetics , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , Female , Fumarate Hydratase/deficiency , Genes, Tumor Suppressor , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Skin Neoplasms/enzymology , Uterine Neoplasms/enzymology
19.
Cancer Cytopathol ; 126(12): 992-1002, 2018 12.
Article in English | MEDLINE | ID: mdl-30339328

ABSTRACT

BACKGROUND: Fumarate hydratase (FH)-deficient renal cell carcinoma (RCC) is rare and highly aggressive and is believed to arise mostly in the setting of hereditary leiomyomatosis-RCC syndrome with a germline mutation of FH. Because of the aggressiveness of these tumors and a frequent lack of ascertainable family history, these tumors may first present as metastases and be sampled by cytology. The cytologic findings of FH-deficient RCC have not previously been reported. METHODS: Cytologic and limited biopsy samples from patients with FH-deficient RCC were reviewed retrospectively. RESULTS: In total, 24 cytologic and limited biopsy samples from 19 patients (6 women and 13 men; age range, 22-69 years) who had FH-deficient RCC and metastasis at presentation were evaluated. These included 21 cytology samples ranging from malignant effusions (n = 7) to metastases (n = 11), to samples of primary kidney tumors (n = 3). The samples exhibited cells, often in clusters and abortive papillae, with voluminous, finely vacuolated cytoplasm and large, pleomorphic nuclei with prominent, viral inclusion-like nucleoli. A distinctive finding of peripheral cytoplasmic clearing frequently was apparent, and intranuclear cytoplasmic pseudoinclusions were less frequent. Of 7 cell block and biopsy samples, several of which represented sampling from the same patient, all demonstrated tissue fragments that had discernable morphologic patterns associated with FH-deficient RCC, including tubulocystic and intracystic papillary growth. CONCLUSIONS: Features characteristic and suggestive of FH-deficient RCC may be identified in cytologic and small biopsy samples. Although the current samples were identified retrospectively in well characterized cases of FH-deficient RCC, the authors argue that, with appropriate clinical correlation, these features are sufficiently distinctive to trigger recognition and confirmatory workup.


Subject(s)
Carcinoma, Renal Cell/genetics , Fumarate Hydratase/genetics , Germ-Line Mutation , Kidney Neoplasms/genetics , Kidney/metabolism , Adult , Aged , Biopsy , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Female , Fumarate Hydratase/deficiency , Genetic Predisposition to Disease/genetics , Humans , Kidney/enzymology , Kidney/pathology , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Male , Middle Aged , Retrospective Studies , Young Adult
20.
Am J Surg Pathol ; 31(10): 1578-85, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17895761

ABSTRACT

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is an autosomal dominant familial syndrome characterized by the development of cutaneous and uterine leiomyomas as well as renal tumors. The mutation of this condition has been identified in the fumarate hydratase (FH, 1q42.3-q43) gene. The histology of the renal cancers has not been well described or illustrated because of the newness of the syndrome. We reviewed 40 renal tumors resected from 38 patients belonging to HLRCC families with proven fumarate hydratase germline mutation. Patients ranged in age from 17 to 75 years of age. Tumors were unilateral in all but 2 cases. The size of the tumors varied between 2.3 and 20 cm and there was no laterality preference. Several different architectural patterns were recognized: papillary (25 cases), tubulo-papillary (8 cases), tubular (2 cases), and solid (1 case). Mixed patterns were also present in 4 cases. The most important histologic feature of these neoplasms, which we believe to be the hallmark of the HLRCC tumors, is the presence of a characteristic large nucleus with a very prominent inclusion like orangiophilic or eosinophilic nucleolus, surrounded by a clear halo. Immunohistochemical studies did not provide a specific marker for these tumors, however, loss of heterozygosity at 1q32 and 1q42-44 was frequently found. These tumors are associated with poor prognosis and frequent spread to regional lymph nodes. At the moment, morphology is the best tool to recognize these tumors. Proper diagnosis of this syndrome by the pathologist may assist in early detection of these tumors.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Leiomyomatosis/pathology , Adolescent , Adult , Aged , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , Cell Nucleus/pathology , DNA, Neoplasm/analysis , Female , Fumarate Hydratase/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Loss of Heterozygosity , Male , Middle Aged , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL