Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Gastroenterology ; 145(3): 540-3.e22, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23684749

ABSTRACT

Microsatellite instability can be found in approximately 15% of all colorectal cancers. To detect new oncogenes we sequenced the exomes of 25 colorectal tumors and respective healthy colon tissue. Potential mutation hot spots were confirmed in 15 genes; ADAR, DCAF12L2, GLT1D1, ITGA7, MAP1B, MRGPRX4, PSRC1, RANBP2, RPS6KL1, SNCAIP, TCEAL6, TUBB6, WBP5, VEGFB, and ZBTB2; these were validated in 86 tumors with microsatellite instability. ZBTB2, RANBP2, and PSRC1 also were found to contain hot spot mutations in the validation set. The form of ZBTB2 associated with colorectal cancer increased cell proliferation. The mutation hot spots might be used to develop personalized tumor profiling and therapy.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Microsatellite Instability , Oncogenes , Aged , Case-Control Studies , Female , Genetic Markers , Humans , Male , Sequence Analysis, DNA
2.
Gastroenterology ; 144(7): 1402-9, 1409.e1-5, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23399955

ABSTRACT

BACKGROUND & AIMS: Gastrointestinal polyposis is a common clinical problem, yet there is no consensus on how to best manage patients with moderate-load polyposis. Identifying genetic features of this disorder could improve management and especially surveillance of these patients. We sought to determine the prevalence of hamartomatous polyposis-associated mutations in the susceptibility genes PTEN, BMPR1A, SMAD4, ENG, and STK11 in individuals with ≥5 gastrointestinal polyps, including at least 1 hamartomatous or hyperplastic/serrated polyp. METHODS: We performed a prospective, referral-based study of 603 patients (median age: 51 years; range, 2-89 years) enrolled from June 2006 through January 2012. Genomic DNA was extracted from peripheral lymphocytes and analyzed for specific mutations and large rearrangements in PTEN, BMPR1A, SMAD4, and STK11, as well as mutations in ENG. Recursive partitioning analysis was used to determine cutoffs for continuous variables. The prevalence of mutations was compared using Fisher's exact test. Logistic regression analyses were used to determine univariate and multivariate risk factors. RESULTS: Of 603 patients, 119 (20%) had a personal history of colorectal cancer and most (n = 461 [76%]) had <30 polyps. Seventy-seven patients (13%) were found to have polyposis-associated mutations, including 11 in ENG (1.8%), 13 in PTEN (2.2%), 13 in STK11 (2.2%), 20 in BMPR1A (3.3%), and 21 in SMAD4 (3.5%). Univariate clinical predictors for risk of having these mutations included age at presentation younger than 40 years (19% vs 10%; P = .008), a polyp burden of ≥30 (19% vs 11%; P = .014), and male sex (16% vs 10%; P = .03). Patients who had ≥1 ganglioneuroma (29% vs 2%; P < .001) or presented with polyps of ≥3 histologic types (20% vs 2%; P = .003) were more likely to have germline mutations in PTEN. CONCLUSIONS: Age younger than 40 years, male sex, and specific polyp histologies are significantly associated with risk of germline mutations in hamartomatous-polyposis associated genes. These associations could guide clinical decision making and further investigations.


Subject(s)
Antigens, CD/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Colonic Polyps/genetics , Germ-Line Mutation , PTEN Phosphohydrolase/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Cell Surface/genetics , Smad4 Protein/genetics , AMP-Activated Protein Kinase Kinases , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Endoglin , Female , Genotype , Humans , Male , Middle Aged , Peutz-Jeghers Syndrome/genetics , Prospective Studies , Young Adult
3.
Gastroenterology ; 143(6): 1482-1491.e3, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960657

ABSTRACT

BACKGROUND & AIMS: Familial visceral myopathy (FVM) is a rare inherited form of myopathic pseudo-obstruction; little is known about the genetic factors that cause this disorder. FVM is characterized by impaired functions of enteric smooth muscle cells, resulting in abnormal intestinal motility, severe abdominal pain, malnutrition, and even death. We searched for genetic factors that might cause this disorder. METHODS: We performed whole-exome sequence analysis of blood samples from 2 individuals in a family that had 7 members diagnosed with FVM. Sanger sequencing was used to analyze additional family members and 280 individuals without this disorder (controls). Intestinal tissue samples from 4 patients and 2 controls were analyzed by immunohistochemistry. Functional studies, including immunofluorescence, cell contractility, and actomyosin structure analyses, were performed using CRL-1976 and U2OS sarcoma cell lines. RESULTS: Whole-exome sequence analysis of DNA from 2 siblings identified 83 gene variants that were shared between the siblings and considered as possible disease-causing changes. A heterozygous variant, R148S in enteric smooth muscle actin γ-2 (ACTG2), segregated with disease phenotype. Intestinal smooth muscle (muscularis propria) from individuals with FVM had reduced levels of cytoplasmic ACTG2 and abnormal accumulation of the protein into intracellular inclusions compared with controls. Sarcoma cells that expressed exogenous ACTG2(R148S) incorporated reduced amounts of this protein into actin filaments compared with cells expressing ACTG2(wt) (P < .001). ACTG2(R148S) also interfered with actin cytoskeleton organization and the contractile activities of the cells, indicating a dominant-negative effect. These findings, along with the site of the variation in the protein, indicate that ACTG2 R148S interferes with actin filament assembly. CONCLUSIONS: We identified the R148S variant in ACTG2 as a cause of FVM in one family. The altered ACTG2 protein appears to aggregate, rather than form actin filaments, in intestinal smooth muscle tissue. This defect could impair contraction of the visceral smooth muscle cells and reduce bowel motility.


Subject(s)
Actins/genetics , Actins/metabolism , Intestinal Mucosa/metabolism , Intestinal Pseudo-Obstruction/genetics , Intestinal Pseudo-Obstruction/metabolism , Muscle, Smooth/metabolism , Mutation, Missense/genetics , Adult , Chromosome Segregation/genetics , Exome/genetics , Female , Finland , Gastrointestinal Motility/physiology , Genome-Wide Association Study , Heterozygote , Humans , Intestinal Pseudo-Obstruction/physiopathology , Intestines/physiopathology , Male , Middle Aged , Muscle Contraction/physiology , Muscle, Smooth/physiopathology , Myenteric Plexus/abnormalities , Myenteric Plexus/metabolism , Myenteric Plexus/physiopathology , Pedigree , Phenotype
4.
Int J Cancer ; 130(7): 1558-66, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-21544814

ABSTRACT

Defects in the mismatch repair system lead to microsatellite instability (MSI), a feature observed in ∼ 15% of all colorectal cancers (CRCs). Microsatellite mutations that drive tumourigenesis, typically inactivation of tumour suppressors, are selected for and are frequently detected in MSI cancers. Here, we evaluated somatic mutations in microsatellite repeats of 790 genes chosen based on reduced expression in MSI CRC and existence of a coding mononucleotide repeat of 6-10 bp in length. All the repeats were initially sequenced in 30 primary MSI CRC samples and whenever frameshift mutations were identified in >20%, additional 70 samples were sequenced. To distinguish driver mutations from passengers, we similarly analyzed the occurrence of frameshift mutations in 121 intronic control repeats and utilized a statistical regression model to determine cut-off mutation frequencies for repeats of all types (A/T and C/G, 6-10 bp). Along with several know target genes, including TGFBR2, ACVR2, and MSH3, six novel candidate driver genes emerged that harbored significantly more mutations than identical control repeats. The mutation frequencies in 100 MSI CRC samples were 51% in G8 of GLYR1, 47% in T9 of ABCC5, 43% in G8 of WDTC1, 33% in A8 of ROCK1, 30% in T8 of OR51E2, and 28% in A8 of TCEB3. Immunohistochemical staining of GLYR1 revealed defective protein expression in tumors carrying biallelic mutations, supporting a loss of function hypothesis. This is a large scale, unbiased effort to identify genes that when mutated are likely to contribute to MSI CRC development.


Subject(s)
Colorectal Neoplasms/genetics , DNA, Neoplasm/genetics , Microsatellite Instability , Cell Line, Tumor , Frameshift Mutation , HCT116 Cells , Humans , Immunohistochemistry/methods , Microsatellite Repeats , Mutation Rate , Regression Analysis
5.
Science ; 334(6053): 252-5, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21868628

ABSTRACT

Uterine leiomyomas, or fibroids, are benign tumors that affect millions of women worldwide and that can cause considerable morbidity. To study the genetic basis of this tumor type, we examined 18 uterine leiomyomas derived from 17 different patients by exome sequencing and identified tumor-specific mutations in the mediator complex subunit 12 (MED12) gene in 10. Through analysis of 207 additional tumors, we determined that MED12 is altered in 70% (159 of 225) of tumors from a total of 80 patients. The Mediator complex is a 26-subunit transcriptional regulator that bridges DNA regulatory sequences to the RNA polymerase II initiation complex. All mutations resided in exon 2, suggesting that aberrant function of this region of MED12 contributes to tumorigenesis.


Subject(s)
Leiomyoma/genetics , Mediator Complex/genetics , Uterine Neoplasms/genetics , Codon , Exons , Female , Gene Expression Profiling , Humans , INDEL Mutation , Introns , Leiomyoma/metabolism , Mutation , Mutation, Missense , Signal Transduction , Uterine Neoplasms/metabolism
6.
J Pathol ; 225(1): 4-11, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21630274

ABSTRACT

Germline mutations in the FH gene encoding the Krebs cycle enzyme fumarate hydratase predispose to hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome. FH-deficient cells and tissues accumulate high levels of fumarate, which may act as an oncometabolite and contribute to tumourigenesis. A recently proposed role for fumarate in the covalent modification of cysteine residues to S-(2-succinyl) cysteine (2SC) (termed protein succination) prompted us to assess 2SC levels in our existing models of HLRCC. Herein, using a previously characterized antibody against 2SC, we show that genetic ablation of FH causes high levels of protein succination. We next hypothesized that immunohistochemistry for 2SC would serve as a metabolic biomarker for the in situ detection of FH-deficient tissues. Robust detection of 2SC was observed in Fh1 (murine FH)-deficient renal cysts and in a retrospective series of HLRCC tumours (n = 16) with established FH mutations. Importantly, 2SC was undetectable in normal tissues (n = 200) and tumour types not associated with HLRCC (n = 1342). In a prospective evaluation of cases referred for genetic testing for HLRCC, the presence of 2SC-modified proteins (2SCP) correctly predicted genetic alterations in FH in every case. In two series of unselected type II papillary renal cancer (PRCC), prospectively analysed by 2SCP staining followed by genetic analysis, the biomarker accurately identified previously unsuspected FH mutations (2/33 and 1/36). The investigation of whether metabolites in other tumour types produce protein modification signature(s) that can be assayed using similar strategies will be of interest in future studies of cancer.


Subject(s)
Carcinoma, Renal Cell/diagnosis , Fumarate Hydratase/deficiency , Kidney Neoplasms/diagnosis , Leiomyomatosis/diagnosis , Neoplastic Syndromes, Hereditary/diagnosis , Adult , Aged , Animals , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/genetics , Disease Models, Animal , Female , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Kidney Neoplasms/genetics , Leiomyomatosis/genetics , Loss of Heterozygosity , Male , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Neoplastic Syndromes, Hereditary/genetics , Prospective Studies , Sensitivity and Specificity , Succinic Acid/metabolism
7.
Blood ; 118(3): 493-8, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21562039

ABSTRACT

A strong clustering of Hodgkin lymphoma in certain families has been long acknowledged. However, the genetic factors in the background of familial Hodgkin lymphoma are largely unknown. We have studied a family of 4 cousins with a rare subtype of the disease, nodular lymphocyte predominant Hodgkin lymphoma. We applied exome sequencing together with genome-wide linkage analysis to this family and identified a truncating germline mutation in nuclear protein, ataxia-telangiectasia locus (NPAT) gene, which segregated in the family. We also studied a large number of samples from other patients with Hodgkin lymphoma, and a germline variation leading to the deletion of serine 724 was found in several cases suggesting an elevated risk for the disease (odds ratio = 4.11; P = .018). NPAT is thus far the first gene implicated in nodular lymphocyte predominant Hodgkin lymphoma predisposition.


Subject(s)
Cell Cycle Proteins/genetics , Germ-Line Mutation , Hodgkin Disease/epidemiology , Hodgkin Disease/genetics , Nuclear Proteins/genetics , Cell Line, Tumor , DNA Mutational Analysis , Family Health , Female , Finland/epidemiology , Genetic Linkage , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Humans , Lymphoma, Follicular/epidemiology , Lymphoma, Follicular/genetics , Male , Pedigree , Risk Factors , Young Adult
8.
Fam Cancer ; 10(2): 397-411, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21404119

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC, also known as multiple cutaneous and uterine leiomyomatosis, MCUL) is a highly penetrant autosomal dominant tumor predisposition syndrome characterized by benign leiomyomas of the skin and the uterus. Renal cell carcinomas, occurring in a subset of the HLRCC families, are exceptionally aggressive. Therefore careful, frequent surveillance strategies are recommended. Association of malignant smooth-muscle tumors, leiomyosarcomas, with HLRCC has been observed but the risk appears to be smaller than initially estimated. To date inactivating heterozygous mutations in the fumarate hydratase (FH, fumarase) gene, predisposing to HLRCC, have been found in approximately 180 families worldwide. The most extensively studied hypothesis on molecular mechanisms of HLRCC tumorigenesis is activation of the hypoxia pathway due to aberrant stabilization of the HIF1 transcription factor. HIF1 regulates transcription of genes relevant for vascularization, glucose transport and glycolysis, processes that facilitate tumor growth. However, additional mechanisms underlying tumor formation are likely to exist.


Subject(s)
Kidney Neoplasms/genetics , Leiomyomatosis/congenital , Female , Fumarate Hydratase/genetics , Genetic Counseling , Heterozygote , Humans , Hypoxia-Inducible Factor 1/genetics , Kidney Neoplasms/diagnosis , Kidney Neoplasms/etiology , Kidney Neoplasms/therapy , Leiomyomatosis/diagnosis , Leiomyomatosis/etiology , Leiomyomatosis/genetics , Leiomyomatosis/therapy , Neoplastic Syndromes, Hereditary , Skin Neoplasms/genetics , Uterine Neoplasms/genetics
9.
Carcinogenesis ; 32(3): 305-11, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21163887

ABSTRACT

Frameshift mutations frequently accumulate in microsatellite-unstable colorectal cancers (MSI CRCs) typically leading to downregulation of the target genes due to nonsense-mediated messenger RNA decay. However, frameshift mutations that occur in the 3' end of the coding regions can escape decay, which has largely been ignored in previous works. In this study, we characterized nonsense-mediated decay-escaping frameshift mutations in MSI CRC in an unbiased, genome wide manner. Combining bioinformatic search with expression profiling, we identified genes that were predicted to escape decay after a deletion in a microsatellite repeat. These repeats, located in 258 genes, were initially sequenced in 30 MSI CRC samples. The mitotic checkpoint kinase TTK was found to harbor decay-escaping heterozygous mutations in exon 22 in 59% (105/179) of MSI CRCs, which is notably more than previously reported. Additional novel deletions were found in exon 5, raising the mutation frequency to 66%. The exon 22 of TTK contains an A(9)-G(4)-A(7) locus, in which the most common mutation was a mononucleotide deletion in the A(9) (c.2560delA). When compared with identical non-coding repeats, TTK was found to be mutated significantly more often than expected without selective advantage. Since TTK inhibition is known to induce override of the mitotic spindle assembly checkpoint (SAC), we challenged mutated cancer cells with the microtubule-stabilizing drug paclitaxel. No evidence of checkpoint weakening was observed. As a conclusion, heterozygous TTK mutations occur at a high frequency in MSI CRCs. Unexpectedly, the plausible selective advantage in tumourigenesis does not appear to be related to SAC.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Colorectal Neoplasms/genetics , Frameshift Mutation/genetics , Microsatellite Instability , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus , Adenocarcinoma/pathology , Aged , Blotting, Western , Colorectal Neoplasms/pathology , Computational Biology , DNA, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Male , Microsatellite Repeats/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein-Tyrosine Kinases , Tumor Cells, Cultured
10.
Cancer Res ; 70(22): 9153-65, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20978192

ABSTRACT

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is caused by mutations in the Krebs cycle enzyme fumarate hydratase (FH). It has been proposed that "pseudohypoxic" stabilization of hypoxia-inducible factor-α (HIF-α) by fumarate accumulation contributes to tumorigenesis in HLRCC. We hypothesized that an additional direct consequence of FH deficiency is the establishment of a biosynthetic milieu. To investigate this hypothesis, we isolated primary mouse embryonic fibroblast (MEF) lines from Fh1-deficient mice. As predicted, these MEFs upregulated Hif-1α and HIF target genes directly as a result of FH deficiency. In addition, detailed metabolic assessment of these MEFs confirmed their dependence on glycolysis, and an elevated rate of lactate efflux, associated with the upregulation of glycolytic enzymes known to be associated with tumorigenesis. Correspondingly, Fh1-deficient benign murine renal cysts and an advanced human HLRCC-related renal cell carcinoma manifested a prominent and progressive increase in the expression of HIF-α target genes and in genes known to be relevant to tumorigenesis and metastasis. In accord with our hypothesis, in a variety of different FH-deficient tissues, including a novel murine model of Fh1-deficient smooth muscle, we show a striking and progressive upregulation of a tumorigenic metabolic profile, as manifested by increased PKM2 and LDHA protein. Based on the models assessed herein, we infer that that FH deficiency compels cells to adopt an early, reversible, and progressive protumorigenic metabolic milieu that is reminiscent of that driving the Warburg effect. Targets identified in these novel and diverse FH-deficient models represent excellent potential candidates for further mechanistic investigation and therapeutic metabolic manipulation in tumors.


Subject(s)
Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/cytology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Leiomyomatosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Spectral Karyotyping
11.
Am J Pathol ; 177(4): 1969-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20709796

ABSTRACT

Mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene have been shown to predispose to pituitary adenoma predisposition, a condition characterized by growth hormone (GH)-secreting pituitary tumors. To study AIP-mediated tumorigenesis, we generated an Aip mouse model. Heterozygous mice developed normally but were prone to pituitary adenomas, in particular to those secreting GH. A complete loss of AIP was detected in these lesions, and full penetrance was reached at the age of 15 months. No excess of any other tumor type was found. Ki-67 analysis indicated that Aip-deficient tumors have higher proliferation rates compared with Aip-proficient tumors, suggesting a more aggressive disease. Similar to human AIP-deficient pituitary adenomas, immunohistochemical studies showed that expression of aryl hydrocarbon receptor nuclear translocator 1 or 2 (ARNT or ARNT2) protein was lost in the mouse tumors, suggesting that mechanisms of AIP-related tumorigenesis involve aberrant ARNT function. The Aip(+/-) mouse appears to be an excellent model for the respective human disease phenotype. This model constitutes a tool to further study AIP-associated pituitary tumorigenesis and may be potentially valuable in efforts to develop therapeutic strategies to treat pituitary adenomas.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Growth Hormone-Secreting Pituitary Adenoma/etiology , Growth Hormone-Secreting Pituitary Adenoma/pathology , Intracellular Signaling Peptides and Proteins/physiology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Blotting, Western , Cell Proliferation , Female , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Humans , Immunoenzyme Techniques , Loss of Heterozygosity , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Mol Cancer Res ; 8(7): 952-60, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20551151

ABSTRACT

The circadian clock regulates daily variations in physiologic processes. CLOCK acts as a regulator in the circadian apparatus controlling the expression of other clock genes, including PER1. Clock genes have been implicated in cancer-related functions; in this work, we investigated CLOCK as a possible target of somatic mutations in microsatellite unstable colorectal cancers. Combining microarray gene expression data and public gene sequence information, we identified CLOCK as 1 of 790 putative novel microsatellite instability (MSI) target genes. A total of 101 MSI colorectal carcinomas (CRC) were sequenced for a coding microsatellite in CLOCK. The effect of restoring CLOCK expression was studied in LS180 cells lacking wild-type CLOCK by stably expressing GST-CLOCK or glutathione S-transferase empty vector and testing the effects of UV-induced apoptosis and radiation by DNA content analysis using flow cytometry. Putative novel CLOCK target genes were searched by using ChIP-seq. CLOCK mutations occurred in 53% of MSI CRCs. Restoring CLOCK expression in cells with biallelic CLOCK inactivation resulted in protection against UV-induced apoptosis and decreased G(2)-M arrest in response to ionizing radiation. Using ChIP-Seq, novel CLOCK-binding elements were identified near DNA damage genes p21, NBR1, BRCA1, and RAD50. CLOCK is shown to be mutated in cancer, and altered response to DNA damage provides one plausible mechanism of tumorigenesis.


Subject(s)
Adenocarcinoma/genetics , CLOCK Proteins/genetics , Colorectal Neoplasms/genetics , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Frameshift Mutation , Gene Expression Profiling , HCT116 Cells , Humans , Microsatellite Repeats , Molecular Sequence Data , Mutation
13.
Am J Pathol ; 175(6): 2501-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19850893

ABSTRACT

Germline mutations in the aryl hydrocarbon receptor interacting protein (AIP) gene predispose to the development of pituitary adenomas. Here, we characterized AIP mutation positive (AIPmut+) and AIP mutation negative (AIPmut-) pituitary adenomas by immunohistochemistry. The expressions of the AIP-related proteins aryl hydrocarbon receptor (AHR), AHR nuclear translocator (ARNT), cyclin-dependent kinase inhibitor 1B encoding p27(Kip1), and hypoxia-inducible factor 1-alpha were examined in 14 AIPmut+ and 53 AIPmut- pituitary adenomas to detect possible expression differences. In addition, the expression of CD34, an endothelial and hematopoietic stem cell marker, was analyzed. We found ARNT to be less frequently expressed in AIPmut+ pituitary adenomas (P = 0.001), suggesting that AIP regulates the ARNT levels. AIP small interfering RNA-treated HeLa, HEK293, or Aip-null mouse embryonic fibroblast cells did not show lowered expression of ARNT. Instead, in the pituitary adenoma cell line GH3, Aip silencing caused a partial reduction of Arnt and a clear increase in cell proliferation. We also observed a trend for increased expression of nuclear AHR in AIPmut+ samples, although the difference was not statistically significant (P = 0.06). The expressions of p27(Kip1), hypoxia-inducible factor 1-alpha, or CD34 did not differ between tumor types. The present study shows that the expression of ARNT protein is significantly reduced in AIPmut+ tumors. We suggest that the down-regulation of ARNT may be connected to an imbalance in AHR/ARNT complex formation arising from aberrant cAMP signaling.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Pituitary Neoplasms/metabolism , Signal Transduction/physiology , Animals , Antigens, CD34/biosynthesis , Antigens, CD34/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Blotting, Western , Cyclin-Dependent Kinase Inhibitor p27 , Down-Regulation , Gene Expression , Gene Expression Profiling , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mutation , Pituitary Neoplasms/genetics , RNA, Small Interfering , Receptors, Aryl Hydrocarbon/biosynthesis , Receptors, Aryl Hydrocarbon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
14.
Genes Chromosomes Cancer ; 48(7): 544-51, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19373782

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a tumor predisposition syndrome with cutaneous and uterine leiomyomatosis as well as renal cell cancer (RCC) as its clinical manifestations. HLRCC is caused by heterozygous germline mutations in the fumarate hydratase (fumarase) gene. In this study, we used array comparative genomic hybridization to identify the specific copy number changes characterizing the HLRCC-associated RCCs. The study material comprised formalin-fixed paraffin-embedded renal tumors obtained from Finnish patients with HLRCC. All 11 investigated tumors displayed the papillary type 2 histopathology typical for HLRCC renal tumors. The most frequent copy number changes detected in at least 3/11 (27%) of the tumors were gains in chromosomes 2, 7, and 17, and losses in 13q12.3-q21.1, 14, 18, and X. These findings provide genetic evidence for a distinct copy number profile in HLRCC renal tumors compared with sporadic RCC tumors of the same histopathological subtype, and delineate chromosomal regions that associate with this very aggressive form of RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Dosage , Kidney Neoplasms/genetics , Leiomyomatosis/genetics , Adult , Aged , Chromosome Aberrations , Comparative Genomic Hybridization , Female , Gene Deletion , Humans , Male , Middle Aged , Mutagenesis, Insertional
15.
Cancer Genet Cytogenet ; 183(2): 83-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18503824

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a syndrome predisposing to cutaneous and uterine leiomyomatosis as well as renal cell cancer and uterine leiomyosarcoma. Heterozygous germline mutations in the fumarate hydratase (FH, fumarase) gene are known to cause HLRCC. On occasion, no FH mutation is detected by direct sequencing, despite the evident HLRCC phenotype in a family. In the present study, to investigate whole gene or exonic deletions and amplifications in FH mutation-negative patients, we used multiplex ligation-dependent probe amplification technology. The study material comprised 7 FH mutation-negative HLRCC patients and 12 patients affected with HLRCC-associated phenotypes, including papillary RCC, early-onset RCC, uterine leiomyomas, or uterine leiomyosarcoma. A novel FH mutation, a deletion of FH exon 1 that encodes the mitochondrial signal peptide, was detected in one of the HLRCC patients (1/7). The patient with the FH mutation displayed numerous painful cutaneous leiomyomas and papillary type renal cell cancer. Our finding, together with the two patients with whole FH gene deletion who had been detected previously, suggests that exonic or whole-gene FH deletions are not a frequent cause of HLRCC syndrome.


Subject(s)
Carcinoma, Renal Cell/genetics , Exons , Fumarate Hydratase/genetics , Leiomyomatosis/genetics , Mutation , Sequence Deletion , Base Sequence , DNA Primers , Humans , Ligase Chain Reaction
16.
Int J Cancer ; 121(6): 1386-9, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17520677

ABSTRACT

Germline mutations in nuclear genes encoding mitochondrial enzymes fumarate hydratase (FH) and succinate dehydrogenase (subunits SDHB/C/D) have been implicated in the development of tumor syndromes referred to as hereditary leiomyomatosis and renal cell cancer (HLRCC) and hereditary paragangliomatosis (HPGL), respectively. FH and SDH are operating in the tricarboxylic acid cycle (the TCA cycle, the Krebs cycle). In the FH and SDH deficient tumors, accumulation of the substrates, fumarate and succinate, has been shown to cause stabilization of hypoxia inducible factor 1 alpha (HIF1 alpha). According to recent studies, HIF1 alpha could contribute to the hypoxia induced genomic instability seen in many cancers, through repression of mismatch repair (MMR) protein MSH2. In this study, in agreement with previous works, we found HIF1 alpha to be moderately or highly stabilized in 67% (16/24) and 77% (48/62) of HLRCC tumors and SDHB/C/D paragangliomas (PGL) and pheochromocytomas (PHEO), respectively. In addition, a set of 54 other familial and nonfamilial PGLs/PHEOs were studied. Moderately or highly stabilized HIF1 alpha was present in 68% (26/38) of the PGLs but in PHEOs (n = 16) no such pattern was observed. We then analyzed the suggested link between HIF1 alpha stabilization and MSH2 repression, in HLRCC and HPGL tumor material. No microsatellite instability (MSI) or lack of MSH2 expression was, however, observed. Thus we failed to provide in vivo evidence for the proposed link between HIF1 alpha stabilization and functional MMR deficiency, in TCAC deficient tumors.


Subject(s)
Fumarate Hydratase/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Microsatellite Instability , Neoplastic Syndromes, Hereditary/genetics , Succinate Dehydrogenase/deficiency , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Citric Acid Cycle/physiology , Fumarate Hydratase/genetics , Germ-Line Mutation , Humans , Immunohistochemistry , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , MutS Homolog 2 Protein/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Paraganglioma/genetics , Paraganglioma/metabolism , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Succinate Dehydrogenase/genetics , Tissue Array Analysis
17.
Hum Pathol ; 38(5): 793-6, 2007 May.
Article in English | MEDLINE | ID: mdl-17270241

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a tumor predisposition syndrome caused by mutations in the fumarate hydratase (FH) gene. HLRCC is characterized by uterine and cutaneous leiomyomas, renal cell cancer, and uterine leiomyosarcoma. Typically, renal cell cancers in HLRCC are unilateral and display a papillary type 2 or ductal histology. We describe here a 23-year-old patient carrying a novel FH mutation (N330S) with a bilateral renal cell center. Carcinoma of the right kidney showed papillary structure, but the left tumor was diagnosed as a conventional (clear cell) renal carcinoma, a type not previously described in HLRCC. The clear cell renal carcinoma also displayed loss of the normal FH allele and the FH immunostaining. Our finding extends the number of cases in which HLRCC can be suspected, and the FH immunohistochemistry may serve as a useful tool to screen for HLRCC in young individuals with clear cell renal carcinoma.


Subject(s)
Carcinoma, Renal Cell/genetics , Fumarate Hydratase/genetics , Kidney Neoplasms/genetics , Mutation , Neoplasms, Multiple Primary/diagnosis , Uterine Neoplasms/diagnosis , Adult , Base Sequence , Carcinoma, Renal Cell/surgery , Female , Genetic Predisposition to Disease , Humans , Incidental Findings , Kidney Neoplasms/surgery , Leiomyoma/diagnosis
18.
Gene Expr ; 14(2): 59-69, 2007.
Article in English | MEDLINE | ID: mdl-18257390

ABSTRACT

Fumarate hydratase (FH) is an enzyme of the mitochondrial tricarboxylic acid cycle (TCAC). Here we report the characterization of a novel FH variant (FHv) that contains an alternative exon 1b, thus lacking the mitochondrial signal sequence. Distinct from mitochondrial FH, FHv localized to cytosol and nucleus and lacked FH enzyme activity. FHv was expressed ubiquitously in human fetal and adult tissues. Heat shock and prolonged hypoxia increased FHv expression in a cell line (HTB 115) by nine- and fourfold, respectively. These results suggest that FHv has an alternative function outside the TCAC related to cellular stress response.


Subject(s)
Fumarate Hydratase/genetics , Gene Expression Regulation , Stress, Physiological/genetics , Base Sequence , Blotting, Western , Cell Line , DNA, Complementary , Fluorescent Antibody Technique , Humans , Molecular Sequence Data , Protein Biosynthesis , Subcellular Fractions/enzymology
19.
Prostate ; 66(11): 1144-50, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16652384

ABSTRACT

BACKGROUND: Amplification and overexpression of EIF3S3 gene has been demonstrated in breast and prostate cancer. Here, our goal was to study the effect of EIF3S3 on cell growth. METHODS: The effect of EIF3S3 on growth of NIH 3T3 murine fibroblasts as well as breast (SK-Br-3 and ZR-75-1) and prostate (PC-3 and LNCaP) cancer cell lines was examined by using transfection with inducible pTet-Off system and siRNAs. RESULTS: NIH 3T3 cells with overexpression of EIF3S3 grew significantly faster than cells transfected with empty vector and survived longer when grown in soft agar. The EIF3S3 overexpression was associated with increased fraction of cells in S-phase and with phosphorylation of retinoblastoma (Rb) protein. siRNA treatment inhibited significantly (P = 0.0022) the growth of all breast and prostate cancer cell lines studied. CONCLUSIONS: The results suggest that EIF3S3 regulates cell growth and viability, and that overexpression of the gene may provide growth advantage to the cancer cells.


Subject(s)
Cell Division , Eukaryotic Initiation Factor-3/genetics , Gene Expression , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Apoptosis , Cell Line, Tumor , Humans , Male , Mice , NIH 3T3 Cells , Phosphorylation , RNA, Small Interfering/genetics , Retinoblastoma Protein/metabolism , S Phase , Transfection
20.
Int J Cancer ; 119(2): 283-7, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16477632

ABSTRACT

Germline mutations in fumarate hydratase (FH) gene at 1q43 predispose to hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome. In HLRCC, the most common clinical features are leiomyomas of the skin and uterus, and in a subset of the families, renal cell cancer (RCC) and uterine leiomyosarcoma (ULMS) occur frequently at young age. This study was conducted to evaluate the possible contribution of FH mutations in a population-based series of early onset (< or = 45 years) ULMSs. Eighty-one cases were identified through the national cancer registry, and samples from 67 cases (83%) were available for FH mutation screening and analysis of allelic imbalance (AI) at the FH locus. Seventeen percent of tumors showed AI. In the mutation analysis, a novel missense mutation K424R was found. The mutation was also found from the patient's normal tissue. To study whether this variant has functional consequences, FH enzyme activity assay was performed in a cell model. The activity of the mutated protein was significantly reduced as compared to wild type (p = 0.009). This study shows that FH germline mutations can occur in seemingly nonsyndromic cases of ULMS (1/67, 1.5%). It appears that on the population level hereditary FH defects do play a role in pathogenesis of sporadic early onset ULMSs, albeit rarely.


Subject(s)
Fumarate Hydratase/genetics , Germ-Line Mutation , Leiomyosarcoma/enzymology , Leiomyosarcoma/genetics , Mutation, Missense , Uterine Neoplasms/enzymology , Uterine Neoplasms/genetics , Adolescent , Adult , Age of Onset , Aged , Allelic Imbalance , Amino Acid Sequence , Arginine , Child , Child, Preschool , DNA Mutational Analysis , DNA, Neoplasm/analysis , Female , Humans , Infant , Lysine , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...