Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Elife ; 72018 09 18.
Article in English | MEDLINE | ID: mdl-30226466

ABSTRACT

Uterine leiomyomas (ULs) are benign tumors that are a major burden to women's health. A genome-wide association study on 15,453 UL cases and 392,628 controls was performed, followed by replication of the genomic risk in six cohorts. Effects of the risk alleles were evaluated in view of molecular and clinical characteristics. 22 loci displayed a genome-wide significant association. The likely predisposition genes could be grouped to two biological processes. Genes involved in genome stability were represented by TERT, TERC, OBFC1 - highlighting the role of telomere maintenance - TP53 and ATM. Genes involved in genitourinary development, WNT4, WT1, SALL1, MED12, ESR1, GREB1, FOXO1, DMRT1 and uterine stem cell marker antigen CD44, formed another strong subgroup. The combined risk contributed by the 22 loci was associated with MED12 mutation-positive tumors. The findings link genes for uterine development and genetic stability to leiomyomagenesis, and in part explain the more frequent occurrence of UL in women of African origin.


Subject(s)
Genetic Loci , Genetic Predisposition to Disease , Genomic Instability , Leiomyoma/genetics , Uterine Neoplasms/genetics , Female , Genome-Wide Association Study , Humans , Morphogenesis , Risk Assessment , Uterus/growth & development
2.
Br J Cancer ; 117(12): 1855-1864, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29073636

ABSTRACT

BACKGROUND: Uterine leiomyomas can be classified into molecularly distinct subtypes according to their genetic triggers: MED12 mutations, HMGA2 upregulation, or inactivation of FH. The aim of this study was to identify metabolites and metabolic pathways that are dysregulated in different subtypes of leiomyomas. METHODS: We performed global metabolomic profiling of 25 uterine leiomyomas and 17 corresponding myometrium specimens using liquid chromatography-tandem mass spectroscopy. RESULTS: A total of 641 metabolites were detected. All leiomyomas displayed reduced homocarnosine and haeme metabolite levels. We identified a clearly distinct metabolomic profile for leiomyomas of the FH subtype, characterised by metabolic alterations in the tricarboxylic acid cycle and pentose phosphate pathways, and increased levels of multiple lipids and amino acids. Several metabolites were uniquely elevated in leiomyomas of the FH subtype, including N6-succinyladenosine and argininosuccinate, serving as potential biomarkers for FH deficiency. In contrast, leiomyomas of the MED12 subtype displayed reduced levels of vitamin A, multiple membrane lipids and amino acids, and dysregulation of vitamin C metabolism, a finding which was also compatible with gene expression data. CONCLUSIONS: The study reveals the metabolomic heterogeneity of leiomyomas and provides the requisite framework for strategies designed to target metabolic alterations promoting the growth of these prevalent tumours.


Subject(s)
Leiomyoma/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Amino Acids/metabolism , Argininosuccinic Acid/metabolism , Ascorbic Acid/metabolism , Citric Acid Cycle , Female , Fumarate Hydratase/genetics , HMGA2 Protein/genetics , Humans , Leiomyoma/genetics , Lipid Metabolism , Mediator Complex/genetics , Metabolic Networks and Pathways , Metabolome , Pentose Phosphate Pathway , Vitamin A/metabolism
3.
Cardiovasc Pathol ; 30: 1-5, 2017.
Article in English | MEDLINE | ID: mdl-28623706

ABSTRACT

A male patient with obstructive jaundice was found to have an incidental nodule within the inferior vena cava (IVC), below the level of the renal vein, on abdominal imaging. At the time of the Whipple's procedure for pancreatic adenocarcinoma, the IVC mass measuring 3.4×2.7×2.2 cm was also removed. Histologically, the lesion was well circumscribed, composed focally of spindle-shaped cells with cigar-shaped nuclei reminiscent of smooth muscle and a dominant pervasive, pleomorphic, bizarre giant cell component. Two mitoses per 10 high-power fields were identified in the most mitotically active area of the entire tumor, with the vast majority of the tumor being mitotically inert. Additionally, no evidence of coagulative necrosis was noted. The bizarre giant cells had multi- and polylobated configurations, and several were replete with nuclear pseudoinclusions. Both the spindle cell and pleomorphic components displayed strong immunoreactivity for all smooth muscle markers. This lesion conformed morphologically to a smooth muscle tumor with bizarre nuclei or so-called symplastic/bizarre leiomyoma, as encountered in the uterus. However, current thinking based on location in the IVC and the presence of any mitotic activity with cellular atypia makes this lesion a leiomyosarcoma. Perhaps more pragmatic terminology would be smooth muscle tumor with bizarre nuclei and low malignant potential since the limited number of cases described thus far appear to have a more indolent course.


Subject(s)
Leiomyoma/pathology , Leiomyosarcoma/pathology , Smooth Muscle Tumor/pathology , Vena Cava, Inferior , Adenocarcinoma/pathology , Aged , Cell Nucleus/pathology , Diagnosis, Differential , Giant Cells/pathology , Humans , Leiomyoma/diagnosis , Leiomyoma/genetics , Leiomyosarcoma/diagnosis , Leiomyosarcoma/genetics , Male , Mediator Complex/genetics , Mitotic Index , Mutation , Neoplasms, Multiple Primary/pathology , Pancreatic Neoplasms/pathology , Smooth Muscle Tumor/diagnosis , Smooth Muscle Tumor/genetics , Terminology as Topic , Vena Cava, Inferior/pathology
4.
Sci Rep ; 7(1): 1015, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28432313

ABSTRACT

Up to 86% of uterine leiomyomas harbour somatic mutations in mediator complex subunit 12 (MED12). These mutations have been associated with conventional histology, smaller tumour size, and larger number of tumours within the uterus. Prior studies, with limited sample sizes, have failed to detect associations between other clinical features and MED12 mutations. Here, we prospectively collected 763 uterine leiomyomas and the corresponding normal myometrial tissue from 244 hysterectomy patients, recorded tumour characteristics, collected clinical data from medical records, and screened the tissue samples for MED12 mutations to assess potential associations between clinical variables and mutation status. Out of 763 leiomyomas, 599 (79%) harboured a MED12 mutation. In the analysis of tumour characteristics, positive MED12-mutation status was significantly associated with smaller tumour size, conventional histology, and subserous location, relative to intramural. In the analysis of clinical variables, the number of MED12-mutation-positive tumours showed an inverse association with parity, and the number of mutation-negative tumours showed a positive association with a history of pelvic inflammatory disease. This study confirmed the previously reported differences and discovered novel differentiating features for MED12-mutation-positive and -negative leiomyomas. These findings emphasise the relevance of specific driver mutations in genesis and presentation of uterine leiomyomas.


Subject(s)
Leiomyoma/pathology , Mediator Complex/genetics , Mutation , Uterine Neoplasms/pathology , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Hysterectomy , Leiomyoma/genetics , Leiomyoma/surgery , Middle Aged , Parity , Prospective Studies , Uterine Neoplasms/genetics , Uterine Neoplasms/surgery
5.
Br J Cancer ; 114(12): 1405-11, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27187686

ABSTRACT

BACKGROUND: Uterine leiomyomas from hereditary leiomyomatosis and renal cell cancer (HLRCC) patients are driven by fumarate hydratase (FH) inactivation or occasionally by mediator complex subunit 12 (MED12) mutations. The aim of this study was to analyse whether MED12 mutations and FH inactivation are mutually exclusive and to determine the contribution of MED12 mutations on HLRCC patients' myomagenesis. METHODS: MED12 exons 1 and 2 mutation screening and 2SC immunohistochemistry indicative for FH deficiency was performed on a comprehensive series of HLRCC patients' (122 specimens) and sporadic (66 specimens) tumours. Gene expression analysis was performed using Affymetrix GeneChip Human Exon Arrays (Affymetrix, Santa Clara, CA, USA). RESULTS: Nine tumours from HLRCC patients harboured a somatic MED12 mutation and were negative for 2SC immunohistochemistry. All remaining successfully analysed lesions (107/116) were deficient for FH. Of sporadic tumours, 35/64 were MED12 mutation positive and none displayed a FH defect. In global gene expression analysis FH-deficient tumours clustered together, whereas HLRCC patients' MED12 mutation-positive tumours clustered together with sporadic MED12 mutation-positive tumours. CONCLUSIONS: Somatic MED12 mutations and biallelic FH inactivation are mutually exclusive in both HLRCC syndrome-associated and sporadic uterine leiomyomas. The great majority of HLRCC patients' uterine leiomyomas are caused by FH inactivation, but incidental tumours driven by somatic MED12 mutations also occur. These MED12 mutation-positive tumours display similar expressional profiles with their sporadic counterparts and are clearly separate from FH-deficient tumours.


Subject(s)
Biomarkers, Tumor/genetics , Fumarate Hydratase/metabolism , Leiomyoma/enzymology , Leiomyoma/genetics , Mediator Complex/genetics , Uterine Neoplasms/enzymology , Uterine Neoplasms/genetics , Enzyme Activation , Female , Germ-Line Mutation , Humans , Immunohistochemistry , Mediator Complex/metabolism , Mutation , Transcriptome
6.
Proc Natl Acad Sci U S A ; 113(5): 1315-20, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787895

ABSTRACT

Uterine leiomyomas are common benign smooth muscle tumors that impose a major burden on women's health. Recent sequencing studies have revealed recurrent and mutually exclusive mutations in leiomyomas, suggesting the involvement of molecularly distinct pathways. In this study, we explored transcriptional differences among leiomyomas harboring different genetic drivers, including high mobility group AT-hook 2 (HMGA2) rearrangements, mediator complex subunit 12 (MED12) mutations, biallelic inactivation of fumarate hydratase (FH), and collagen, type IV, alpha 5 and collagen, type IV, alpha 6 (COL4A5-COL4A6) deletions. We also explored the transcriptional consequences of 7q22, 22q, and 1p deletions, aiming to identify possible target genes. We investigated 94 leiomyomas and 60 corresponding myometrial tissues using exon arrays, whole genome sequencing, and SNP arrays. This integrative approach revealed subtype-specific expression changes in key driver pathways, including Wnt/ß-catenin, Prolactin, and insulin-like growth factor (IGF)1 signaling. Leiomyomas with HMGA2 aberrations displayed highly significant up-regulation of the proto-oncogene pleomorphic adenoma gene 1 (PLAG1), suggesting that HMGA2 promotes tumorigenesis through PLAG1 activation. This was supported by the identification of genetic PLAG1 alterations resulting in expression signatures as seen in leiomyomas with HMGA2 aberrations. RAD51 paralog B (RAD51B), the preferential translocation partner of HMGA2, was up-regulated in MED12 mutant lesions, suggesting a role for this gene in the genesis of leiomyomas. FH-deficient leiomyomas were uniquely characterized by activation of nuclear factor erythroid 2-related factor 2 (NRF2) target genes, supporting the hypothesis that accumulation of fumarate leads to activation of the oncogenic transcription factor NRF2. This study emphasizes the need for molecular stratification in leiomyoma research and possibly in clinical practice as well. Further research is needed to determine whether the candidate biomarkers presented herein can provide guidance for managing the millions of patients affected by these lesions.


Subject(s)
Biomarkers, Tumor/metabolism , Leiomyoma/classification , Uterine Neoplasms/classification , Biomarkers, Tumor/genetics , Female , Gene Expression Profiling , Humans , Leiomyoma/genetics , Mutation , Proto-Oncogene Mas , Uterine Neoplasms/genetics
7.
Hum Mol Genet ; 24(15): 4407-16, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25964426

ABSTRACT

Uterine leiomyomas are extremely frequent benign smooth muscle tumors often presenting as multiple concurrent lesions and causing symptoms such as abnormal menstrual bleeding, abdominal pain and infertility. While most leiomyomas are believed to arise independently, a few studies have encountered separate lesions harboring identical genetic changes, suggesting a common clonal origin. To investigate the frequency of clonally related leiomyomas, genome-wide tools need to be utilized, and thus little is known about this phenomenon. Using MED12 sequencing and SNP arrays, we searched for clonally related uterine leiomyomas in a set of 103 tumors from 14 consecutive patients who entered hysterectomy owing to symptomatic lesions. Whole-genome sequencing was also utilized to study the genomic architecture of clonally related tumors. This revealed four patients to have two or more tumors that were clonally related, all of which lacked MED12 mutations. Furthermore, some tumors were composed of genetically distinct subclones, indicating a nonlinear, branched model of tumor evolution. DEPDC5 was discovered as a novel tumor suppressor gene playing a role in the progression of uterine leiomyomas. Perhaps counterintuitively­considering Knudson's two-hit hypothesis­a large shared deletion was followed by different truncating DEPDC5 mutations in four clonally related leiomyomas. This study provides insight into the intratumor heterogeneity of these tumors and suggests that a shared clonal origin is a common feature of leiomyomas that do not carry an MED12 mutation. These observations also offer one explanation to the common occurrence of multiple concurrent lesions.


Subject(s)
Leiomyoma/genetics , Mediator Complex/genetics , Neoplasms/genetics , Repressor Proteins/genetics , Uterine Neoplasms/genetics , Carcinogenesis/genetics , Clone Cells , Female , GTPase-Activating Proteins , Genetic Predisposition to Disease , Genome, Human , Humans , Leiomyoma/pathology , Mutation , Neoplasms/pathology , Polymorphism, Single Nucleotide , Uterine Neoplasms/pathology
8.
Fertil Steril ; 102(4): 1137-42, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25108465

ABSTRACT

OBJECTIVE: To determine the frequency of mediator complex subunit 12 (MED12) mutations in well-documented, prospectively collected, unselected series of sporadic uterine leiomyomas to better understand the contribution of MED12 mutations in leiomyoma genesis. DESIGN: Mutation analysis of two prospectively collected sample series. SETTING: Department of gynecology in university hospital and medical genetics research laboratory. PATIENT(S): 164 uterine leiomyomas from 28 patients (13 consecutive and 15 unselected patients) undergoing hysterectomy. INTERVENTION(S): MED12 mutation screening by direct sequencing, and clinical data collection. MAIN OUTCOME MEASURE(S): MED12 mutation status and various clinical variables. RESULT(S): MED12 mutations were found in 73 (83.0%) of 88 and 65 (85.5%) of 76 of uterine leiomyomas from the consecutive and unselected patient series, respectively. Smaller tumor size and a larger number of tumors correlated with positive MED12 mutation status. CONCLUSION(S): The frequency of MED12 mutations in our prospectively collected uterine leiomyoma sets was higher than in previous works. This is in keeping with the concept that MED12 mutation-positive tumors tend to be smaller in size than MED12 mutation-negative tumors. The results highlight the central role of MED12 mutations in uterine leiomyoma genesis.


Subject(s)
Leiomyoma/genetics , Mediator Complex/genetics , Mutation , Uterine Neoplasms/genetics , Adult , Aged , DNA Mutational Analysis , Female , Gene Frequency , Genetic Predisposition to Disease , Genetic Testing/methods , Hospitals, University , Humans , Leiomyoma/pathology , Leiomyoma/surgery , Middle Aged , Prospective Studies , Risk Factors , Uterine Neoplasms/pathology , Uterine Neoplasms/surgery
9.
Fertil Steril ; 102(3): 621-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25106763

ABSTRACT

Uterine leiomyomas are benign smooth-muscle tumors of extremely low malignant potential. Early work utilizing classical cytogenetics revealed that a subset of uterine leiomyomas harbor recurrent chromosomal rearrangements, such as translocations affecting the HMGA2 gene. Our understanding of the genetics of many tumor types has deepened remarkably with the emergence of next-generation sequencing technologies. Exome sequencing identified that the majority of leiomyomas display highly specific MED12 mutations. Further studies suggest that these MED12 hotspot mutations are also frequent in breast fibroadenomas, but not in other human tumors. Whole-genome sequencing showed that a subset of leiomyomas display complex chromosomal rearrangements resembling chromothripsis. These were formed in a single event of chromosomal breakage and random reassembly involving one or a limited number of chromosomes. Although most leiomyomas have been shown to arise independently, these studies also revealed that distinct nodules within a uterus may display identical genetic changes indicating a common clonal origin. A minority of leiomyomas were also found to display deletions within the COL4A5-COL4A6 genes, leading to upregulation of the adjacent gene IRS4. The findings derived from high-throughput sequencing combined with previous knowledge have led to an emerging molecular classification of leiomyomas, suggesting that there are several distinct pathogenic pathways involved in leiomyoma formation. The evidence points to at least 4 molecular subclasses: leiomyomas with MED12 mutation, FH inactivation, HMGA2 overexpression, and COL4A6-COL4A5 deletion. Elucidating the molecular pathogenesis of leiomyomas should be relevant for developing treatments for this very common disease.


Subject(s)
Genomics/methods , High-Throughput Nucleotide Sequencing , Leiomyoma/genetics , Uterine Neoplasms/genetics , Exome , Female , Genes, Neoplasm , Humans , Leiomyoma/classification , Leiomyoma/pathology , Mediator Complex/genetics , Uterine Neoplasms/classification , Uterine Neoplasms/pathology
10.
N Engl J Med ; 369(1): 43-53, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23738515

ABSTRACT

BACKGROUND: Uterine leiomyomas are benign but affect the health of millions of women. A better understanding of the molecular mechanisms involved may provide clues to the prevention and treatment of these lesions. METHODS: We performed whole-genome sequencing and gene-expression profiling of 38 uterine leiomyomas and the corresponding myometrium from 30 women. RESULTS: Identical variants observed in some separate tumor nodules suggested that these nodules have a common origin. Complex chromosomal rearrangements resembling chromothripsis were a common feature of leiomyomas. These rearrangements are best explained by a single event of multiple chromosomal breaks and random reassembly. The rearrangements created tissue-specific changes consistent with a role in the initiation of leiomyoma, such as translocations of the HMGA2 and RAD51B loci and aberrations at the COL4A5-COL4A6 locus, and occurred in the presence of normal TP53 alleles. In some cases, separate events had occurred more than once in single tumor-cell lineages. CONCLUSIONS: Chromosome shattering and reassembly resembling chromothripsis (a single genomic event that results in focal losses and rearrangements in multiple genomic regions) is a major cause of chromosomal abnormalities in uterine leiomyomas; we propose that tumorigenesis occurs when tissue-specific tumor-promoting changes are formed through these events. Chromothripsis has previously been associated with aggressive cancer; its common occurrence in leiomyomas suggests that it also has a role in the genesis and progression of benign tumors. We observed that multiple separate tumors could be seeded from a single lineage of uterine leiomyoma cells. (Funded by the Academy of Finland Center of Excellence program and others.).


Subject(s)
Chromosome Aberrations , Fumarate Hydratase/deficiency , Leiomyoma/genetics , Mediator Complex/genetics , Uterine Neoplasms/genetics , Chromosome Breakage , Chromosome Deletion , Collagen Type IV/genetics , Female , Fumarate Hydratase/genetics , Gene Expression Profiling , Gene Rearrangement , Genome-Wide Association Study , Humans , Mutation , Myometrium/chemistry , Up-Regulation
11.
Oncotarget ; 2(12): 966-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22182697

ABSTRACT

Uterine leiomyomas, or fibroids, are extremely common tumors. Regardless of their benign nature, fibroids can cause considerable morbidity. Women with African ancestry have a threefold increased risk of developing uterine leiomyomas with a greater symptom severity when compared to white women. Recently, we demonstrated that exon 2 of the MED12 gene is somatically altered in up to 70 per cent of uterine leiomyomas in a series of Finnish (Caucasian) patients. To validate these results in other populations, we sequenced a set of 28 uterine leiomyomas for MED12 exon 2 mutations from 18 different Black African or Coloured South African patients. We observed 14 mutation positive lesions (50%). When corrected by tumor size, these results are very similar to those derived in the Finnish material. This study confirms a major role of MED12 in the genesis of leiomyomas, regardless of ethnicity.


Subject(s)
Leiomyoma/genetics , Mediator Complex/genetics , Uterine Neoplasms/genetics , Adult , Base Sequence , Black People/genetics , DNA Mutational Analysis , Female , Humans , Middle Aged , Mutation , South Africa
SELECTION OF CITATIONS
SEARCH DETAIL
...