Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Sci Rep ; 11(1): 9371, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931688

ABSTRACT

Pathogenic mutations in fumarate hydratase (FH) drive hereditary leiomyomatosis and renal cell cancer (HLRCC) and increase the risk of developing uterine leiomyomas (ULMs). An integrated proteogenomic analysis of ULMs from HLRCC (n = 16; FH-mutation confirmed) and non-syndromic (NS) patients (n = 12) identified a significantly higher protein:transcript correlation in HLRCC (R = 0.35) vs. NS ULMs (R = 0.242, MWU p = 0.0015). Co-altered proteins and transcripts (228) included antioxidant response element (ARE) target genes, such as thioredoxin reductase 1 (TXNRD1), and correlated with activation of NRF2-mediated oxidative stress response signaling in HLRCC ULMs. We confirm 185 transcripts previously described as altered between HLRCC and NS ULMs, 51 co-altered at the protein level and several elevated in HLRCC ULMs are involved in regulating cellular metabolism and glycolysis signaling. Furthermore, 367 S-(2-succino)cysteine peptides were identified in HLRCC ULMs, of which sixty were significantly elevated in HLRCC vs. NS ULMs (LogFC = 1.86, MWU p < 0.0001). These results confirm and define novel proteogenomic alterations in uterine leiomyoma tissues collected from HLRCC patients and underscore conserved molecular alterations correlating with inactivation of the FH tumor suppressor gene.


Subject(s)
Biomarkers, Tumor/analysis , Fumarate Hydratase/genetics , Genetic Predisposition to Disease , Leiomyomatosis/pathology , Mutation , Neoplastic Syndromes, Hereditary/pathology , Proteogenomics/methods , Proteome/metabolism , Skin Neoplasms/pathology , Uterine Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Case-Control Studies , Female , Humans , Leiomyomatosis/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Proteome/analysis , Skin Neoplasms/metabolism , Uterine Neoplasms/metabolism
2.
Genes Chromosomes Cancer ; 60(3): 210-216, 2021 03.
Article in English | MEDLINE | ID: mdl-33099845

ABSTRACT

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is an autosomal dominant familial syndrome that results from germline mutation in the fumarate hydratase (FH) gene and is associated with an increased risk for smooth muscle tumors of the uterus and skin and renal cell carcinoma. HLRCC associated RCC develop in up to 25% of patients, often presenting in the fourth decade and are high stage, aggressive tumors with poor clinical outcome. Most women with HLRCC develop large and bulky uterine smooth muscle tumors (USMT) in the second to third decade, thus presenting the ideal opportunity for early detection of HLCC to enable timely implementation of surveillance for their RCC risk. However, the concept of screening women with USMT for HLRCC is challenging given that HLRCC is rare but USMT are common. In addition, FH deficiency in USMT can also result from sporadic FH gene aberrations, unrelated to HLRCC, further complicating any potential screening process. Recent studies show that tumor morphology can be used to identify FH deficiency in USMT and thereby direct patients to formal genetic counseling. The low magnification clues of staghorn shaped blood vessels and alveolar pattern should prompt for high magnification examination for eosinophilic cytoplasmic inclusions and oval nuclei containing prominent eosinophilic macronucleoli surrounded by a halo. Additional clues include Schwannoma-like growth and chain-like distribution of the tumor cells. Although immunostains exist for FH and 2SC, their role is limited in the presence of well-developed FH deficient morphology. The prevalence of germline pathogenic mutation in FH among women with USMT with FH deficient morphology is as high as 50% in some studies, with somatic FH mutation accounting for the remainder. Therefore, morphologic evaluation of USMT for features of FH deficiency can serve as a screening tool for HLRCC syndrome by triaging patients to formal hereditary risk assessment.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Leiomyomatosis/genetics , Neoplastic Syndromes, Hereditary/genetics , Skin Neoplasms/genetics , Smooth Muscle Tumor/genetics , Uterine Neoplasms/genetics , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Leiomyomatosis/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neoplasms, Connective and Soft Tissue/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Skin Neoplasms/metabolism , Smooth Muscle Tumor/diagnosis , Smooth Muscle Tumor/metabolism , Smooth Muscle Tumor/pathology , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
3.
J Biol Chem ; 295(39): 13410-13418, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32820045

ABSTRACT

An important context in which metabolism influences tumorigenesis is the genetic cancer syndrome hereditary leiomyomatosis and renal cell carcinoma (HLRCC), a disease in which mutation of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) causes hyperaccumulation of fumarate. This electrophilic oncometabolite can alter gene activity at the level of transcription, via reversible inhibition of epigenetic dioxygenases, as well as posttranslationally, via covalent modification of cysteine residues. To better understand the potential for metabolites to influence posttranslational modifications important to tumorigenesis and cancer cell growth, here we report a chemoproteomic analysis of a kidney-derived HLRCC cell line. Using a general reactivity probe, we generated a data set of proteomic cysteine residues sensitive to the reduction in fumarate levels caused by genetic reintroduction of active FH into HLRCC cell lines. This revealed a broad up-regulation of cysteine reactivity upon FH rescue, which evidence suggests is caused by an approximately equal proportion of transcriptional and posttranslational modification-mediated regulation. Gene ontology analysis highlighted several new targets and pathways potentially modulated by FH mutation. Comparison of the new data set with prior studies highlights considerable heterogeneity in the adaptive response of cysteine-containing proteins in different models of HLRCC. This is consistent with emerging studies indicating the existence of cell- and tissue-specific cysteine-omes, further emphasizing the need for characterization of diverse models. Our analysis provides a resource for understanding the proteomic adaptation to fumarate accumulation and a foundation for future efforts to exploit this knowledge for cancer therapy.


Subject(s)
Cysteine/metabolism , Fumarate Hydratase/metabolism , Fumarates/metabolism , Kidney Neoplasms/metabolism , Leiomyomatosis/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Skin Neoplasms/metabolism , Uterine Neoplasms/metabolism , Cell Line, Tumor , Cysteine/genetics , Fumarate Hydratase/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
4.
Clin Nucl Med ; 45(10): 827-829, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32701814

ABSTRACT

Cutaneous leiomyomas are rare, sporadic, or inherited benign tumors arising from smooth muscle cells of the skin associated with various disorders. We present a case of multiple cutaneous leiomyomas showing increased FDG uptake with SUVmax of 19.9. This case indicates cutaneous leiomyoma should be considered as a rare differential diagnosis in patients with hypermetabolic cutaneous lesions. Careful correlation with clinical history is needed to avoid misdiagnosis.


Subject(s)
Fluorodeoxyglucose F18/metabolism , Leiomyomatosis/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Skin Neoplasms/metabolism , Uterine Neoplasms/metabolism , Biological Transport , Diagnosis, Differential , Female , Humans , Leiomyomatosis/diagnostic imaging , Male , Neoplastic Syndromes, Hereditary/diagnostic imaging , Skin Neoplasms/diagnostic imaging , Uterine Neoplasms/diagnostic imaging
5.
Mod Pathol ; 33(9): 1844-1860, 2020 09.
Article in English | MEDLINE | ID: mdl-32341498

ABSTRACT

Intravenous leiomyomatosis (IVL) is an unusual uterine smooth muscle proliferation that can be associated with aggressive clinical behavior despite a histologically benign appearance. It has some overlapping molecular characteristics with both uterine leiomyoma and leiomyosarcoma based on limited genetic data. In this study, we assessed the clinical and morphological characteristics of 28 IVL and their correlation with molecular features and protein expression, using array comparative genomic hybridization (aCGH) and Cyclin D1, p16, phosphorylated-Rb, SMARCB1, SOX10, CAIX, SDHB and FH immunohistochemistry. The most common morphologies were cellular (n = 15), usual (n = 11), and vascular (n = 5; including 3 cellular IVL showing both vascular and cellular features). Among the immunohistochemical findings, the most striking was that all IVL showed differential expression of either p16 or Cyclin D1 in comparison to surrounding nonneoplastic tissue. Cytoplasmic phosphorylated-Rb was present in all but one IVL with hyalinization. SMARCB1, FH, and SDHB were retained; S0X10 and CAIX were not expressed. The most common genetic alterations involved 1p (39%), 22q (36%), 2q (29%), 1q (25%), 13q (21%), and 14q (21%). Hierarchical clustering analysis of recurrent aberrations revealed three molecular groups: Groups 1 (29%) and 2 (18%) with associated del(22q), and Group 3 (18%) with del(10q). The remaining IVL had nonspecific or no alterations by aCGH. Genomic index scores were calculated for all cases and showed no significant difference between the 14 IVL associated with aggressive clinical behavior (extrauterine extension or recurrence) and those without (median scores 5.15 vs 3.5). Among the 5 IVL associated with recurrence, 4 had a vascular morphology and 3 had alterations of 8q. Recurrent chromosome alterations detected herein overlap with those observed in the spectrum of uterine smooth muscle tumors and involve genes implicated in mesenchymal tumors at different sites with distinct morphological features.


Subject(s)
Leiomyomatosis/genetics , Uterine Neoplasms/genetics , Uterus/pathology , Adult , Aged , Aged, 80 and over , Comparative Genomic Hybridization , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Humans , Leiomyomatosis/metabolism , Leiomyomatosis/pathology , Middle Aged , Phosphorylation , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Uterus/metabolism
6.
Biomolecules ; 10(1)2020 01 16.
Article in English | MEDLINE | ID: mdl-31963199

ABSTRACT

Elevated expression of heme oxygenase-1 (HO-1, encoded by HMOX1) is observed in various types of tumors. Hence, it is suggested that HO-1 may serve as a potential target in anticancer therapies. A novel approach to inhibit HO-1 is related to the synthetic lethality of this enzyme and fumarate hydratase (FH). In the current study, we aimed to validate the effect of genetic and pharmacological inhibition of HO-1 in cells isolated from patients suffering from hereditary leiomyomatosis and renal cell carcinoma (HLRCC)-an inherited cancer syndrome, caused by FH deficiency. Initially, we confirmed that UOK 262, UOK 268, and NCCFH1 cell lines are characterized by non-active FH enzyme, high expression of Nrf2 transcription factor-regulated genes, including HMOX1 and attenuated oxidative phosphorylation. Later, we demonstrated that shRNA-mediated genetic inhibition of HMOX1 resulted in diminished viability and proliferation of cancer cells. Chemical inhibition of HO activity using commercially available inhibitors, zinc and tin metalloporphyrins as well as recently described new imidazole-based compounds, especially SLV-11199, led to decreased cancer cell viability and clonogenic potential. In conclusion, the current study points out the possible relevance of HO-1 inhibition as a potential anti-cancer treatment in HLRCC. However, further studies revealing the molecular mechanisms are still needed.


Subject(s)
Fumarate Hydratase/genetics , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Leiomyomatosis/genetics , Leiomyomatosis/therapy , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/therapy , Skin Neoplasms/genetics , Skin Neoplasms/therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/therapy , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Fumarate Hydratase/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Heme Oxygenase-1/metabolism , Humans , Leiomyomatosis/drug therapy , Leiomyomatosis/metabolism , Metalloporphyrins/pharmacology , Neoplastic Syndromes, Hereditary/drug therapy , Neoplastic Syndromes, Hereditary/metabolism , RNA, Small Interfering/pharmacology , RNAi Therapeutics , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Uterine Neoplasms/drug therapy , Uterine Neoplasms/metabolism
7.
Semin Cancer Biol ; 61: 158-166, 2020 04.
Article in English | MEDLINE | ID: mdl-31689495

ABSTRACT

Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC) is an autosomal dominant hereditary cancer syndrome with incomplete penetrance. It is caused by a germline amorphic allele of the FH gene, which encodes the TCA cycle enzyme, fumarate hydratase (FH). HLRCC patients are genetically predisposed to develop skin leiomyomas, uterine fibroids, and the aggressive kidney cancer of type 2 papillary morphology. Loss-of-heterozygocity at the FH locus that cause a complete loss of FH enzymatic function is always detected in these tumor tissues. Molecular pathway elucidation, genomic studies, and systematic genetics screens reported over the last two decades have identified several FH-inactivation driven pathways alterations, as well as rationally conceived treatment strategies that specifically target FH-/- tumor cells. These treatment strategies include ferroptosis induction, oxidative stress promotion, and metabolic alteration. As the fundamental biology of HLRCC continues to be uncovered, these treatment strategies continue to be refined and may one day lead to a strategy to prevent disease onset among HLRCC patients. With a more complete picture of HLRCC biology, the safe translation of experimental treatment strategies into clinical practice is achievable in the foreseeable future.


Subject(s)
Leiomyomatosis/etiology , Neoplastic Syndromes, Hereditary/etiology , Skin Neoplasms/etiology , Uterine Neoplasms/etiology , Biomarkers, Tumor , Disease Management , Disease Susceptibility , Fumarate Hydratase/genetics , Genes, Tumor Suppressor , Genetic Predisposition to Disease , Genetic Testing , Genomics/methods , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leiomyomatosis/diagnosis , Leiomyomatosis/metabolism , Leiomyomatosis/therapy , Mutation , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/therapy , Proteome , Signal Transduction , Skin Neoplasms/diagnosis , Skin Neoplasms/metabolism , Skin Neoplasms/therapy , Translational Research, Biomedical , Uterine Neoplasms/diagnosis , Uterine Neoplasms/metabolism , Uterine Neoplasms/therapy
8.
Clin Cancer Res ; 26(2): 391-396, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31636096

ABSTRACT

PURPOSE: Inherited pathogenic variants in genes encoding the metabolic enzymes succinate dehydrogenase (SDH) and fumarate hydratase predispose to tumor development through accumulation of oncometabolites (succinate and fumarate, respectively; ref. 1). Noninvasive in vivo detection of tumor succinate by proton magnetic resonance spectroscopy (1H-MRS) has been reported in SDH-deficient tumors, but the potential utility of this approach in the management of patients with hereditary leiomyomatosis and renal cell cancer syndrome or Reed syndrome is unknown. EXPERIMENTAL DESIGN: Magnetic resonance spectroscopy (1H-MRS) was performed on three cases and correlated with germline genetic results and tumor IHC when available. RESULTS: Here, we have demonstrated a proof of principle that 1H-MRS can provide a noninvasive diagnosis of hereditary leiomyomatosis and renal cell cancer syndrome or Reed syndrome through detection of fumarate accumulation in vivo. CONCLUSIONS: This study demonstrates that in vivo detection of fumarate could be employed as a functional biomarker.


Subject(s)
Fumarates/metabolism , Germ-Line Mutation , Kidney Neoplasms/diagnosis , Leiomyomatosis/diagnosis , Neoplastic Syndromes, Hereditary/diagnosis , Proton Magnetic Resonance Spectroscopy/methods , Skin Neoplasms/diagnosis , Uterine Neoplasms/diagnosis , Adult , Female , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Humans , Kidney Neoplasms/metabolism , Leiomyomatosis/metabolism , Male , Middle Aged , Neoplastic Syndromes, Hereditary/metabolism , Skin Neoplasms/metabolism , Succinate Dehydrogenase/genetics , Uterine Neoplasms/metabolism
9.
Sci Rep ; 9(1): 18409, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804603

ABSTRACT

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is characterized by germline mutations of the FH gene that encodes for the TCA cycle enzyme, fumarate hydratase. HLRCC patients are at risk for the development of an aggressive form of type 2 papillary renal cell carcinoma. By studying the mechanism of action of marizomib, a proteasome inhibitor able to cross the blood-brain barrier, we found that it modulates the metabolism of HLRCC cells. Marizomib decreased glycolysis in vitro and in vivo by downregulating p62 and c-Myc. C-Myc downregulation decreased the expression of lactate dehydrogenase A, the enzyme catalyzing the conversion of pyruvate to lactate. In addition, proteasomal inhibition lowered the expression of the glutaminases GLS and GLS2, which support glutamine metabolism and the maintenance of the redox balance. Thus, in HLRCC cells, proteasome inhibition disrupts glucose and glutamine metabolism, restricting nutrients and lowering the cells' anti-oxidant response capacity. Although the cytotoxicity induced by proteasome inhibitors is complex, the understanding of their metabolic effects in HLRCC may lead to the development of effective therapeutic strategies or to the development of markers of efficacy.


Subject(s)
Fumarate Hydratase/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/drug therapy , Lactones/pharmacology , Leiomyomatosis/drug therapy , Neoplastic Syndromes, Hereditary/drug therapy , Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Pyrroles/pharmacology , Sequestosome-1 Protein/genetics , Skin Neoplasms/drug therapy , Uterine Neoplasms/drug therapy , Animals , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , Fumarate Hydratase/deficiency , Germ-Line Mutation , Glutaminase/genetics , Glutaminase/metabolism , Glycolysis/drug effects , Glycolysis/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lactate Dehydrogenase 5/genetics , Lactate Dehydrogenase 5/metabolism , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Leiomyomatosis/pathology , Mice , Mice, Nude , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/pathology , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Sequestosome-1 Protein/antagonists & inhibitors , Sequestosome-1 Protein/metabolism , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Methods Enzymol ; 622: 431-448, 2019.
Article in English | MEDLINE | ID: mdl-31155064

ABSTRACT

Dysregulated cellular metabolism is an emerging hallmark of cancer. Improved methods to profile aberrant metabolic activity thus have substantial applications as tools for diagnosis and understanding the biology of malignant tumors. Here we describe the utilization of a bioorthogonal ligation to fluorescently detect the TCA cycle oncometabolite fumarate. This method enables the facile measurement of fumarate hydratase activity in cell and tissue samples, and can be used to detect disruptions in metabolism that underlie the genetic cancer syndrome hereditary leiomyomatosis and renal cell cancer (HLRCC). The current method has substantial utility for sensitive fumarate hydratase activity profiling, and also provides a foundation for future applications in diagnostic detection and imaging of cancer metabolism.


Subject(s)
Citric Acid Cycle , Fumarate Hydratase/metabolism , Fumarates/metabolism , Leiomyomatosis/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Skin Neoplasms/metabolism , Uterine Neoplasms/metabolism , Click Chemistry/methods , Cycloaddition Reaction , Enzyme Assays/methods , Female , Fluorescent Dyes/analysis , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Fluorometry/methods , Fumarates/analysis , Humans
11.
Am J Surg Pathol ; 43(9): 1170-1179, 2019 09.
Article in English | MEDLINE | ID: mdl-31162287

ABSTRACT

Hereditary leiomyomatosis and renal cell carcinoma syndrome (HLRCC), caused by a germline mutation in the fumarate hydratase (FH) gene, predisposes patients to uterine and cutaneous smooth muscle tumors and an aggressive type of renal cell carcinoma. Almost all women with HLRCC develop symptomatic uterine leiomyomas resulting in surgery at young ages, presenting an ideal opportunity for early detection of these patients and the implementation of surveillance measures for renal cell carcinoma. FH-deficient uterine leiomyomas can show characteristic morphologic features (FH-d morphology) that have been previously described. Immunohistochemistry (IHC) for FH can also be helpful in detecting FH deficiency in leiomyomas, which manifests as complete loss of staining for FH. However, the distribution and topography of FH-d morphology and FH loss by IHC in the context of multiple leiomyomas in patients with HLRCC has not been evaluated. The aim of this study is to describe in detail the clinical and pathologic characteristics of uterine leiomyomas from women with HLRCC. Six patients with proven FH germline mutations were included. All available slides were reviewed and FH IHC staining was performed on multiple blocks when possible. Clinical data were extracted from online medical records. All 6 patients presented with symptomatic uterine fibroids and underwent myomectomy (age 24 to 36 y), followed by hysterectomy in 2 patients (age 31 and 40 y). Specimens showed conventional leiomyomas, cellular leiomyomas and leiomyomas with bizarre nuclei. FH-d morphology was present in leiomyomas from all patients and was typically observed as a diffuse finding in the majority of slides across different leiomyoma types. FH-d morphology was absent in some leiomyoma sections from one patient and the morphologic features were focal and subtle in leiomyomas from 2 patients. Both hysterectomy specimens were also notable for showing scattered irregular tongues and nodules of smooth muscle proliferation (leiomyomatosis-like) in the background myometrium. Immunohistochemical staining of multiple slides per patient for FH showed either retained staining in all sections (2/6 cases), loss of staining in all sections (1 case) or variable staining across different leiomyomas (3 cases). In conclusion, patients with HLRCC undergo surgery at young ages for highly symptomatic uterine leiomyomas. FH-d morphology is usually a diffuse and well developed finding across different leiomyomas but may be absent or focal and subtle. FH IHC can show variable results and presence of retained FH staining should not be used to exclude the possibility of HLRCC. Referral for genetic counselling and testing should be considered in a young patient with uterine leiomyomas showing FH-d morphology even if immunohistochemical staining for FH is retained.


Subject(s)
Fumarate Hydratase/biosynthesis , Leiomyomatosis/metabolism , Leiomyomatosis/pathology , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Uterus/metabolism , Uterus/pathology , Adult , Female , Fumarate Hydratase/analysis , Fumarate Hydratase/deficiency , Humans , Hysterectomy , Immunohistochemistry , Metabolism, Inborn Errors , Muscle Hypotonia , Psychomotor Disorders , Uterine Myomectomy
12.
Pak J Pharm Sci ; 32(2): 743-749, 2019 Mar.
Article in English | MEDLINE | ID: mdl-31081789

ABSTRACT

The several types of heterogeneous kidney cancers are interrelated by their primary sites of pathology. Despite its origin in the kidney, renal cell carcinoma (RCC) is associated with its varying genetic basis. Von Hippel-Lindau (VHL) syndrome is the earliest and, thus the most highly, characterized of genetic forms of kidney cancer, which is associated with alterations in the Von Hippel-Lindau (VHL) gene. As a result of his studies and investigations, Otto Warburg reached the conclusion that cancer's fundamental cause is altered mechanism. But this theory was disdained because of the discovery of tumor suppressor genes and oncogenes. Lately, the breakthrough finding about the tumor suppressing role of gene coding for enzymes involved in Krebs cycle has revived the interest in Warburg's hypothesis. This effect has led to the uncovering of the links between metabolic alterations, mitochondrial dysfunction and cancer. One such metastatic cancer characterized by the germ-line inactivating mutation of the gene coding for fumarate hydratase (FH), a Krebs cycle's enzyme, is hereditary leiomyomatosis and renal cell carcinoma (HLRCC). In this review paper, we have discussed the background of this carcinoma, the metabolic dysfunction causing it and its therapeutic solutions.


Subject(s)
Fumarate Hydratase/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Fumarate Hydratase/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Leiomyomatosis/pathology , Mutation , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/pathology , Sesquiterpenes, Guaiane/pharmacology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
13.
Nat Chem Biol ; 15(4): 391-400, 2019 04.
Article in English | MEDLINE | ID: mdl-30718813

ABSTRACT

Hereditary cancer disorders often provide an important window into novel mechanisms supporting tumor growth. Understanding these mechanisms thus represents a vital goal. Toward this goal, here we report a chemoproteomic map of fumarate, a covalent oncometabolite whose accumulation marks the genetic cancer syndrome hereditary leiomyomatosis and renal cell carcinoma (HLRCC). We applied a fumarate-competitive chemoproteomic probe in concert with LC-MS/MS to discover new cysteines sensitive to fumarate hydratase (FH) mutation in HLRCC cell models. Analysis of this dataset revealed an unexpected influence of local environment and pH on fumarate reactivity, and enabled the characterization of a novel FH-regulated cysteine residue that lies at a key protein-protein interface in the SWI-SNF tumor-suppressor complex. Our studies provide a powerful resource for understanding the covalent imprint of fumarate on the proteome and lay the foundation for future efforts to exploit this distinct aspect of oncometabolism for cancer diagnosis and therapy.


Subject(s)
Fumarates/metabolism , Leiomyomatosis/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Skin Neoplasms/metabolism , Uterine Neoplasms/metabolism , Cell Line, Tumor , Chromatography, Liquid/methods , Cysteine , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Leiomyomatosis/genetics , Models, Biological , Neoplastic Syndromes, Hereditary/genetics , Proteomics , Signal Transduction , Skin Neoplasms/genetics , Tandem Mass Spectrometry/methods , Uterine Neoplasms/genetics
14.
Sci Rep ; 9(1): 1442, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30723247

ABSTRACT

The purpose of this study was to explore the potential relationship between intravenous leiomyomatosis (IVL) and uterine myoma (UM) at the molecular level. RNA-sequencing was performed on IVL tumours, UM tumours, and adjacent normal uterine muscle. We compared the gene expression levels between IVL and normal uterine muscle, UM and normal uterine muscle, to identify differentially expressed genes (DEGs). Then we used Gene Ontology Enrichment Analysis to determine the functions of the DEGs and performed specimen cluster analysis. We obtained 98 DEGs between IVL and adjacent normal uterine muscle, and 61 DEGs between UM and adjacent normal uterine muscle. Functional enrichment of both IVL and UM DEGs showed that they are associated with hormone stimulus, extracellular matrix, and cell adhesion. Unsupervised clustering analysis showed that IVL and UM could not be separated completely. Among these dysregulated genes, we found that HOXA13 showed a distinct dysregulated status between IVL and UM. HOXA13 may therefore serves as a biomarker to distinguish IVL and UM. Our results showed that IVL and UM may have similar dysregulated gene networks. They may be closely related, and HOXA13 may serves as a biomarker to distinguish between IVL and UM.


Subject(s)
Leiomyomatosis/genetics , Myoma/genetics , Transcriptome , Uterine Neoplasms/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leiomyomatosis/metabolism , Middle Aged , Myoma/metabolism , Uterine Neoplasms/metabolism , Uterus/metabolism , Uterus/pathology
15.
J Clin Invest ; 129(2): 442-451, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30614813

ABSTRACT

The field of hereditary kidney cancer has begun to mature following the identification of several germline syndromes that define genetic and molecular features of this cancer. Molecular defects within these hereditary syndromes demonstrate consistent deficits in angiogenesis and metabolic signaling, largely driven by altered hypoxia signaling. The classical mutation, loss of function of the von Hippel-Lindau (VHL) tumor suppressor, provides a human pathogenesis model for critical aspects of pseudohypoxia. These features are mimicked in a less common hereditary renal tumor syndrome, known as hereditary leiomyomatosis and renal cell carcinoma. Here, we review renal tumor angiogenesis and metabolism from a HIF-centric perspective, considering alterations in the hypoxic landscape, and molecular deviations resulting from high levels of HIF family members. Mutations underlying HIF deregulation drive multifactorial aberrations in angiogenic signals and metabolism. The mechanisms by which these defects drive tumor growth are still emerging. However, the distinctive patterns of angiogenesis and glycolysis-/glutamine-dependent bioenergetics provide insight into the cellular environment of these cancers. The result is a scenario permissive for aggressive tumorigenesis especially within the proximal renal tubule. These features of tumorigenesis have been highly actionable in kidney cancer treatments, and will likely continue as central tenets of kidney cancer therapeutics.


Subject(s)
Carcinoma, Renal Cell , Genetic Diseases, Inborn , Kidney Neoplasms , Leiomyomatosis , Neoplastic Syndromes, Hereditary , Neovascularization, Pathologic , Von Hippel-Lindau Tumor Suppressor Protein , Cell Hypoxia/genetics , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Genetic Diseases, Inborn/therapy , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Leiomyomatosis/pathology , Leiomyomatosis/therapy , Models, Biological , Mutation , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
16.
Cancer Res ; 78(23): 6539-6548, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30297534

ABSTRACT

: Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is an inherited cancer syndrome associated with a highly aggressive form of type 2 papillary renal cell carcinoma (PRCC). Germline inactivating alterations in fumarate hydratase (FH) cause HLRCC and result in elevated levels of reactive oxygen species (ROS). Recent work indicates that FH-/- PRCC cells have increased activation of ABL1, which promotes tumor growth, but how ABL1 is activated remains unclear. Given that oxidation can regulate protein-tyrosine phosphatase (PTP) catalytic activity, inactivation of an ABL-directed PTP by ROS might account for ABL1 activation in this malignancy. Our group previously developed "q-oxPTPome," a method that globally monitors the oxidation of classical PTPs. In this study, we present a refined q-oxPTPome, increasing its sensitivity by >10×. Applying q-oxPTPome to FH-deficient cell models showed that multiple PTPs were either highly oxidized (including PTPN12) or overexpressed. Highly oxidized PTPs were those with relatively high sensitivity to exogenous H2O2. Most PTP oxidation in FH-deficient cells was reversible, although nearly 40% of PTPN13 was irreversibly oxidized to the sulfonic acid state. Using substrate-trapping mutants, we mapped PTPs to their putative substrates and found that only PTPN12 could target ABL1. Furthermore, knockdown experiments identified PTPN12 as the major ABL1 phosphatase, and overexpression of PTPN12 inhibited ABL1 phosphorylation and HLRCC cell growth. These results show that ROS-induced oxidation of PTPN12 accounts for ABL1 phosphorylation in HLRCC-associated PRCC, revealing a novel mechanism for inactivating a tumor suppressor gene product and establishing a direct link between pathologic PTP oxidation and neoplastic disease. SIGNIFICANCE: This work identifies a novel mechanism of activation of the oncogenic kinase ABL1 via ROS-induced, oxidation-mediated inactivation of cognate protein tyrosine phosphatases.


Subject(s)
Leiomyomatosis/etiology , Leiomyomatosis/metabolism , Neoplastic Syndromes, Hereditary/etiology , Neoplastic Syndromes, Hereditary/metabolism , Oxidation-Reduction , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Uterine Neoplasms/etiology , Uterine Neoplasms/metabolism , Biomarkers , Cell Line, Tumor , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Germ-Line Mutation , Humans , Leiomyomatosis/diagnosis , Metabolome , Metabolomics/methods , Models, Biological , Neoplastic Syndromes, Hereditary/diagnosis , Phosphorylation , Protein Binding , Reactive Oxygen Species , Skin Neoplasms/diagnosis , Uterine Neoplasms/diagnosis
17.
Mod Pathol ; 31(6): 974-983, 2018 06.
Article in English | MEDLINE | ID: mdl-29410489

ABSTRACT

Hereditary leiomyomatosis and renal cell carcinoma syndrome is characterized by an increased risk of agressive renal cell carcinoma, often of type 2 papillary histology, and is caused by FH germline mutations. A prominent eosinophilic macronucleolus with a perinucleolar clear halo is distinctive of hereditary leiomyomatosis and renal cell carcinoma syndrome-associated renal cell carcinoma according to the 2012 ISUP and 2016 WHO kidney tumor classification. From an immunohistochemistry perspective, tumors are often FH-negative and S-(2-succino)-cysteine (2SC) positive. We performed a pathology review of 24 renal tumors in 23 FH mutation carriers, and compared them to 12 type 2 papillary renal cell carcinomas from FH wild-type patients. Prominent eosinophilic nucleoli with perinucleolar halos were present in almost all FH-deficient renal cell carcinomas (23/24). Unexpectedly, they were also present in 58% of type 2 papillary renal cell carcinomas from wild-type patients. Renal cell carcinoma in mutation carriers displayed a complex architecture with multiple patterns, typically papillary, tubulopapillary, and tubulocystic, but also sarcomatoid and rhabdoid. Such pattern diversity was not seen in non-carriers. FH/2SC immunohistochemistry was informative as all hereditary leiomyomatosis and renal cell carcinoma-associated renal cell carcinomas were either FH- or 2SC+. For FH and 2SC immunohistochemistries taken separately, sensitivity of negative anti-FH immunohistochemistry was 87.5% and specificity was 100%. For positive anti-2SC immunohistochemistry, sensitivity, and specificity were 91.7% and 91.7%, respectively. All FH wild-type renal cell carcinoma were FH-positive, and all but one were 2SC-negative. In conclusion, multiplicity of architectural patterns, rhabdoid/sarcomatoid components and combined FH/2SC staining, but not prominent eosinophilic nucleoli with perinucleolar halos, differentiate hereditary leiomyomatosis and renal cell carcinoma-associated renal cell carcinoma from type 2 papillary renal cell carcinoma with efficient FH gene. Our findings are crucial in identifying who should be referred to Cancer Genetics clinics for genetic counseling and testing.


Subject(s)
Carcinoma, Renal Cell/diagnosis , Fumarate Hydratase/metabolism , Kidney Neoplasms/diagnosis , Leiomyomatosis/diagnosis , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Diagnosis, Differential , Fumarate Hydratase/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Leiomyomatosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...