Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Pathol ; 262(1): 50-60, 2024 01.
Article in English | MEDLINE | ID: mdl-37792634

ABSTRACT

Spermatocytic tumor (ST) is a rare type of germ cell tumor that occurs exclusively in the postpubertal testis and typically affects elderly men. Most STs are benign, but rare cases exhibit aggressive clinical behavior, often in association with transition to sarcomatoid histology. Limited molecular analyses have been performed on STs; therefore, their genomic and epigenomic features remain incompletely described. Twenty-seven samples from 25 individual patients were analyzed with a combination of DNA sequencing panels, genomic methylation profiling, SNP array, isochromosome (12p) [i(12p)] FISH, and immunohistochemistry. The series included five metastasizing tumors (three with sarcomatoid transformation, one anaplastic, and one conventional) and 20 non-metastasizing tumors (14 anaplastic and six conventional). Anaplastic tumors comprised a monomorphic population of intermediate-sized neoplastic cells, as previously described. Multiomic analyses demonstrated that there were two genomic subgroups of STs: one with diploid genomes and hotspot RAS/RAF variants and the other with global ploidy shift and absence of recurrent mutations. Relative gain of chromosome 9 was a consistent finding in both subgroups. A comparison of metastasizing and non-metastasizing cases demonstrated that aggressive behavior was associated with the acquisition of pathogenic TP53 mutations and/or relative gains of 12p/i(12p). In cases with sarcomatoid transformation, TP53 mutations seem to underlie the transition to sarcomatoid histology. Genomic methylation analysis demonstrated that aggressive cases with gains of 12p cluster closer to pure seminomas than to STs without gains of 12p. In conclusion, STs include two genomic subgroups, characterized by global ploidy shifts without recurrent mutations and diploid genomes with RAS/RAF hotspot mutations, respectively. Biologic progression was associated with relative gains of 12p and TP53 mutations. The findings in STs with relative gains of 12p suggest that they may exhibit biologic characteristics akin to those seen in germ cell neoplasia in situ-related germ cell tumors rather than non-germ cell neoplasia in situ-derived STs. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Biological Products , Neoplasms, Germ Cell and Embryonal , Seminoma , Testicular Neoplasms , Male , Humans , Aged , Seminoma/genetics , Testicular Neoplasms/metabolism , Neoplasms, Germ Cell and Embryonal/genetics , Genomics , Chromosomes, Human, Pair 12/metabolism
2.
Dig Dis Sci ; 68(4): 1306-1315, 2023 04.
Article in English | MEDLINE | ID: mdl-36348128

ABSTRACT

BACKGROUND AND AIMS: Little is known about the role of chromosome 12 open reading frame 49 (C12ORF49)-induced metabolic signal transduction in tumor growth. We investigated the relationship between C12ORF49 expression and prognosis in colorectal cancer (CRC) patients. METHODS: C12ORF49 protein expression was measured in CRC tissues by Western blot and immunohistochemistry staining. Knock out of C12ORF49 in CRC cells was then performed, and the role of C12ORF49 in CRC cell proliferation and growth was examined. The expression of C12ORF49 in CRC was analyzed in Gene Expression Profiling Interactive Analysis (GEPIA) databases. A prognosis model with 11 C12ORF49-associated genes (CAGs) was generated by TCGA databases. RESULTS: C12ORF49 expression was significantly higher in CRC tumor tissue than in non-tumor tissue. Furthermore, in vitro and in vivo loss-of-function experiments, showed that C12ORF49 plays critical roles in promoting tumor cell growth. There was a significant correlation between C12ORF49 protein and the presence of tumor necrosis. C12ORF49 is critical for its interaction with SREBF1, TMEM41A, and S1PR3 in the poor prognosis of CRC. CONCLUSIONS: Our results suggest that C12ORF49 plays a key role in CRC tumor growth.


Subject(s)
Colorectal Neoplasms , Humans , Colorectal Neoplasms/pathology , Cell Line, Tumor , Chromosomes, Human, Pair 12/metabolism , Open Reading Frames , Prognosis , Cell Proliferation , Gene Expression Regulation, Neoplastic
3.
J Clin Invest ; 131(15)2021 08 02.
Article in English | MEDLINE | ID: mdl-34166228

ABSTRACT

The 12q13-q14 chromosomal region is recurrently amplified in 25% of fusion-positive (FP) rhabdomyosarcoma (RMS) cases and is associated with a poor prognosis. To identify amplified oncogenes in FP RMS, we compared the size, gene composition, and expression of 12q13-q14 amplicons in FP RMS with those of other cancer categories (glioblastoma multiforme, lung adenocarcinoma, and liposarcoma) in which 12q13-q14 amplification frequently occurs. We uncovered a 0.2 Mb region that is commonly amplified across these cancers and includes CDK4 and 6 other genes that are overexpressed in amplicon-positive samples. Additionally, we identified a 0.5 Mb segment that is only recurrently amplified in FP RMS and includes 4 genes that are overexpressed in amplicon-positive RMS. Among these genes, only serine hydroxymethyltransferase 2 (SHMT2) was overexpressed at the protein level in an amplicon-positive RMS cell line. SHMT2 knockdown in amplicon-positive RMS cells suppressed growth, transformation, and tumorigenesis, whereas overexpression in amplicon-negative RMS cells promoted these phenotypes. High SHMT2 expression reduced sensitivity of FP RMS cells to SHIN1, a direct SHMT2 inhibitor, but sensitized cells to pemetrexed, an inhibitor of the folate cycle. In conclusion, our study demonstrates that SHMT2 contributes to tumorigenesis in FP RMS and that SHMT2 amplification predicts differential response to drugs targeting this metabolic pathway.


Subject(s)
Carcinogenesis , Chromosomes, Human, Pair 12 , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glycine Hydroxymethyltransferase , Neoplasm Proteins , Rhabdomyosarcoma , Carcinogenesis/genetics , Carcinogenesis/metabolism , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Female , Glycine Hydroxymethyltransferase/biosynthesis , Glycine Hydroxymethyltransferase/genetics , Humans , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Rhabdomyosarcoma/enzymology , Rhabdomyosarcoma/genetics
4.
Protein Pept Lett ; 27(12): 1192-1203, 2020.
Article in English | MEDLINE | ID: mdl-32533814

ABSTRACT

Triple-A Syndrome (TAS) is a rare autosomal recessive disorder characterized by three cardinal symptoms: alacrimia, achalasia and adrenal insufficiency due to ACTH insensitivity. Various progressive neurological abnormalities and skin changes have been described in association with the syndrome. The disease is caused by mutation in the AAAS gene on chromosome 12q13. Mutations in AAAS were identified in more than 90% of individuals and families with TAS. The protein encoded by AAAS was termed ALADIN and is part of the WD repeat family of proteins, that have been found to be involved in many different functions such as protein-protein interaction, RNA processing, cytoskeleton assembly, control of cell division, signal transduction and apoptosis. Immunohistochemical analysis showed that mutated or truncated ALADIN localizes to the cytoplasm rather than to the nuclear pore complex. The exact function of ALADIN and the mechanisms that lead to the ACTH-resistant adrenal phenotype remains largely unknown. Nonetheless, recent studies provided some insights on the role of ALADIN as a member of the Nuclear Pore Complex not only implicated in the import of proteins involved in DNA repair and oxidative stress homeostasis but also in the strengthening of the mitotic spindle assembly. Early identification of the syndrome is challenging, given the rarity of the condition and high phenotypic heterogeneity even among members of the same family. In this review, we aim to summarize the current knowledge of clinical and molecular profile of patients with TAS and recommendations for the diagnosis, management, and follow-up of patients.


Subject(s)
Adrenal Insufficiency , Chromosomes, Human, Pair 12 , Esophageal Achalasia , Genetic Association Studies , Mutation , Nerve Tissue Proteins , Nuclear Pore Complex Proteins , Adrenal Insufficiency/diagnosis , Adrenal Insufficiency/genetics , Adrenal Insufficiency/metabolism , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , DNA Repair/genetics , Esophageal Achalasia/diagnosis , Esophageal Achalasia/genetics , Esophageal Achalasia/metabolism , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Oxidative Stress/genetics
5.
Diagn Cytopathol ; 47(9): 930-934, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31120625

ABSTRACT

Primary thyroid teratomas are rare, usually benign, and typically occur in children. We report the unusual occurrence of a malignant thyroid teratoma in a young man. Initial ultrasound and CT studies revealed an 8.5 heterogeneous mass involving the entire right thyroid lobe causing tracheal compression and deviation. Fine-needle aspiration (FNA) revealed malignant cells with possible neuroendocrine features. Similar findings have been previously reported, with an occasional interpretation as possible medullary thyroid carcinoma. In no report, as with our case, has the correct diagnosis been suggested with FNA. The surgical specimen contained abundant primitive neuroepithelium with a very minor component of mature ectodermal tissue in one area. Like this case, an abundance of immature neuroepithelium has been reported in essentially all previous reports of primary malignant thyroid teratoma, sometimes creating a challenge to find another type of germ cell tissue. Array comparative genomic hybridization studies in this case revealed a markedly complex karyotype including gain of chromosome 12 and loss of 17p. Amplification of MYCN, EWSR1 rearrangement and isochromosome 12p were not identified, providing no evidence for neuroblastoma or Ewing sarcoma/peripheral neuroectodermal tumor, both of which have also rarely been reported as primary thyroid tumors. With the use of cisplatinum-based chemotherapy combined with radiation, survival times have increased dramatically. Our patient is now disease free and back to his normal activities after relatively short follow-up. Although rare, it is important to be aware that teratomas may present as a thyroid nodule. Recognition by FNA is challenging, and requires multiple modalities for full identification.


Subject(s)
Chemoradiotherapy , Cisplatin/administration & dosage , Teratoma , Thyroid Neoplasms , Adolescent , Biopsy, Fine-Needle , Chromosome Deletion , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 17/metabolism , Humans , Male , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Smith-Magenis Syndrome/genetics , Smith-Magenis Syndrome/metabolism , Smith-Magenis Syndrome/pathology , Smith-Magenis Syndrome/therapy , Teratoma/genetics , Teratoma/metabolism , Teratoma/pathology , Teratoma/therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Thyroid Neoplasms/therapy
6.
Exp Hematol ; 73: 50-63.e2, 2019 05.
Article in English | MEDLINE | ID: mdl-30986496

ABSTRACT

The most frequently occurring genetic abnormality in pediatric B-lymphocyte-lineage acute lymphoblastic leukemia is the t(12;21) chromosomal translocation that results in a ETV6-RUNX1 (also known as TEL-AML1) fusion gene. Expression of ETV6-RUNX1 induces a preleukemic condition leading to acquisition of secondary driver mutations, but the mechanism is poorly understood. SPI-B (encoded by SPIB) is an important transcriptional activator of B-cell development and differentiation. We hypothesized that SPIB is directly transcriptionally repressed by ETV6-RUNX1. Using chromatin immunoprecipitation, we identified a regulatory region in the first intron of SPIB that interacts with ETV6-RUNX1. Mutation of the RUNX1 binding site in SPIB intron 1 prevented transcriptional repression in transient transfection assays. Next, we sought to determine to what extent gene expression in REH cells can be altered by ectopic SPI-B expression. SPI-B expression was forced using CRISPR-mediated gene activation and also using a retroviral vector. Forced expression of SPI-B resulted in altered gene expression and, at high levels, impaired cell proliferation and induced apoptosis. Finally, we identified CARD11 and CDKN1A (encoding p21) as transcriptional targets of SPI-B involved in regulation of proliferation and apoptosis. Taken together, this study identifies SPIB as an important target of ETV6-RUNX1 in regulation of B-cell gene expression in t(12;21) leukemia.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Leukemic , Introns , Oncogene Proteins, Fusion/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Response Elements , Transcription Factors/biosynthesis , Apoptosis/genetics , CARD Signaling Adaptor Proteins/biosynthesis , CARD Signaling Adaptor Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 21/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/genetics , Guanylate Cyclase/biosynthesis , Guanylate Cyclase/genetics , Humans , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transcription Factors/genetics , Translocation, Genetic
7.
Exp Hematol ; 73: 7-12.e4, 2019 05.
Article in English | MEDLINE | ID: mdl-30825516

ABSTRACT

The TEL-AML1 fusion gene, generated by the t(12;21) chromosome translocation, arises in a progenitor/stem cell and could induce clonal expansion of a persistent preleukemic B-cell clone which, on acquisition of secondary alterations, may turn into full-blown leukemia. During infections, deregulated cytokine signaling, including transforming growth factor ß (TGF-ß), can further accelerate this process by creating a protumoral bone marrow (BM) microenvironment. Here, we show that activin A, a member of the TGF-ß family induced under inflammatory conditions, inhibits the proliferation of normal progenitor B cells but not that of preleukemic TEL-AML1-positive clones, thereby providing a selective advantage to the latter. Finally, we find that activin A inhibits BM-derived mesenchymal stromal cell-mediated secretion of CXCL12, a major chemoattractant in the BM compartment, thereby contributing to shape a leukemia-promoting environment. Overall, our findings indicate that activin A, in concert with TGF-ß, could play an important role in the creation of a pro-oncogenic BM microenvironment and provide novel mechanistic insights into TEL-AML1-associated leukemogenesis.


Subject(s)
Activins/metabolism , Bone Marrow/metabolism , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 21/genetics , Leukemia/metabolism , Mesenchymal Stem Cells/metabolism , Precancerous Conditions/metabolism , Stem Cell Niche , Translocation, Genetic , Activins/genetics , Bone Marrow/pathology , Cell Line , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Chromosomes, Human, Pair 12/metabolism , Chromosomes, Human, Pair 21/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Humans , Leukemia/genetics , Leukemia/pathology , Mesenchymal Stem Cells/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/pathology
8.
Leuk Res ; 73: 86-94, 2018 10.
Article in English | MEDLINE | ID: mdl-30248580

ABSTRACT

BACKGROUND: Chromosomal aberrations at the ETV6 gene locus on 12p13.2 are common in bone marrow samples involved by blastic plasmacytoid dendritic cell neoplasm (BPDCN). However, their pathogenic role, incidence in cutaneous BPDCN lesions, and clinical significance have not been assessed systematically. RESULTS: The study group included 30 BPDCN patients, 25 men and 5 women, with a median age of 64 years. Conventional cytogenetic analysis demonstrated karyotypic aberrancies in 15 cases, of which 8 had chromosomal lesions involving 12p. In addition, 2 cases with normal diploid karyotype had cryptic 12p/ETV6 deletion by ETV6 FISH test. Notably, 2 bone marrow samples with ETV6 rearrangement had no detectable BPDCN involvement, but otherwise dynamic changes in the detection of 12p/ETV6 aberrations correlated with the presence of morphologically and/or immunophenotypically detectable disease. Tissue specimens from 6 patients with cutaneous BPDCN all tested positive for homozygous or heterozygous ETV6 deletions. CONCLUSION: We demonstrate that monoallelic and biallelic 12p/ETV6 deletions are highly prevalent in BPDCN, and their detection is enhanced by the use of FISH and aCGH. In addition, 12p/ETV6 may be present in the bone marrow of BPDCN patients in the absence of detectable disease suggesting that such alterations might represent an early pathogenic event.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 12/genetics , Dendritic Cells , Gene Deletion , Hematologic Neoplasms/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Chromosomes, Human, Pair 12/metabolism , Female , Hematologic Neoplasms/metabolism , Humans , Male , Middle Aged , Prevalence , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , ETS Translocation Variant 6 Protein
9.
Pediatr Blood Cancer ; 65(4)2018 Apr.
Article in English | MEDLINE | ID: mdl-29286563

ABSTRACT

BACKGROUND: Congenital mesoblastic nephroma (MN) is a rare pediatric renal tumor representing approximately 5% of all pediatric renal tumors. Three different types of MN are distinguished histologically: classical, cellular, and mixed. A frequent genetic alteration is the translocation t(12;15) resulting in a fusion of the ETV6 gene on 12p13 and the NTRK3 gene on 15p15 that occurs almost exclusively in cellular MN. The aim of this study was to determine translocation status of a large cohort of MN with respect to tumor subtype and outcome. PROCEDURE: In total, clinical data from 111 patients were available. Sixty-seven tumors were classical MN (51%), 29 cellular MN (31%), and 15 were mixed MN (18%). From these 111 cases, 79 were analyzed by FISH and RT-PCR. RESULTS: All classical and mixed MN were translocation negative. Seventeen out of 29 (58%) cellular MN harbored the ETV6-NTRK3 translocation. Five-year relapse-free survival (RFS) and overall survival (OS) were 93.2% and 96.8% for the complete cohort. All seven relapses occurred in translocation negative tumors. Five-year RFS was significantly inferior for cellular and mixed MN compared to classic MN (89%, 80%, and 98%), whereas 5-year OS was similar (93%, 96%, and 98%). Within the group of cellular MN, patients having translocation-positive tumors had a significantly superior RFS (5-year RFS: 100% vs. 73%). CONCLUSION: The majority of cellular MNs harbor the ETV6-NTKR3 gene fusion, whereas all classic- and mixed-type MNs were translocation negative. Within the cellular subgroup, patients having translocation-positive tumors had a significantly superior RFS.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 15 , Nephroma, Mesoblastic , Oncogene Proteins, Fusion , Translocation, Genetic , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Disease-Free Survival , Female , Humans , In Situ Hybridization, Fluorescence , Male , Nephroma, Mesoblastic/genetics , Nephroma, Mesoblastic/metabolism , Nephroma, Mesoblastic/mortality , Nephroma, Mesoblastic/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
10.
J Clin Invest ; 128(1): 427-445, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29227286

ABSTRACT

As new generations of targeted therapies emerge and tumor genome sequencing discovers increasingly comprehensive mutation repertoires, the functional relationships of mutations to tumor phenotypes remain largely unknown. Here, we measured ex vivo sensitivity of 246 blood cancers to 63 drugs alongside genome, transcriptome, and DNA methylome analysis to understand determinants of drug response. We assembled a primary blood cancer cell encyclopedia data set that revealed disease-specific sensitivities for each cancer. Within chronic lymphocytic leukemia (CLL), responses to 62% of drugs were associated with 2 or more mutations, and linked the B cell receptor (BCR) pathway to trisomy 12, an important driver of CLL. Based on drug responses, the disease could be organized into phenotypic subgroups characterized by exploitable dependencies on BCR, mTOR, or MEK signaling and associated with mutations, gene expression, and DNA methylation. Fourteen percent of CLLs were driven by mTOR signaling in a non-BCR-dependent manner. Multivariate modeling revealed immunoglobulin heavy chain variable gene (IGHV) mutation status and trisomy 12 as the most important modulators of response to kinase inhibitors in CLL. Ex vivo drug responses were associated with outcome. This study overcomes the perception that most mutations do not influence drug response of cancer, and points to an updated approach to understanding tumor biology, with implications for biomarker discovery and cancer care.


Subject(s)
Antineoplastic Agents/therapeutic use , Databases, Factual , Hematologic Neoplasms , Leukemia, Lymphocytic, Chronic, B-Cell , Models, Biological , Signal Transduction , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Female , Hematologic Neoplasms/classification , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/classification , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Trisomy/genetics
11.
Gene ; 603: 27-33, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27956168

ABSTRACT

BACKGROUND: CASC18 along with APPL2, OCC-1 and NUAK1 flanking genes are located in 12q23.3 locus which is known as a potential cancer predisposition locus. Only an uncharacterized EST was initially reported for CASC18 and it was crucial to find its full length sequence and function. METHODS AND RESULTS: In an attempt to search for the CASC18's full-length gene sequence, other related ESTs were bioinformatically collected and four novel splice variants (designated as; CASC18-A, -B, -C and -D) were deduced and some were experimentally validated. Two transcription start sites and two alternative polyadenylation sites were deduced for CASC18 gene, using EST data mining and RACE method. CASC18-A and CASC18-D were exclusively expressed in neural cell lines and CASC18-D expression level was gradually increased during the NT2 differentiation to the neuron-like cells. Consistently, overexpression of CASC18-D variant in NT2 cells resulted in remarkable up-regulation of PAX6 neural differentiation marker, suggesting a crucial role of this variant in neural differentiation. CONCLUSION: Here, we introduced seven novel transcription variants for human CASC18 gene in which CASC18-D has the potential of being used as a neural cell differentiation marker.


Subject(s)
Alternative Splicing , Cell Differentiation/genetics , Genetic Loci , Genome, Human , Neural Stem Cells/metabolism , RNA, Long Noncoding/genetics , Cell Line, Tumor , Chromosome Mapping , Chromosomes, Human, Pair 12/chemistry , Chromosomes, Human, Pair 12/metabolism , Expressed Sequence Tags , Humans , Neural Stem Cells/cytology , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , RNA, Long Noncoding/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
12.
Nat Commun ; 7: 12098, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27384883

ABSTRACT

Genome-wide association studies have identified multiple renal cell carcinoma (RCC) susceptibility loci. Here, we use regional imputation and bioinformatics analysis of the 12p12.1 locus to identify the single-nucleotide polymorphism (SNP) rs7132434 as a potential functional variant. Luciferase assays demonstrate allele-specific regulatory activity and, together with data from electromobility shift assays, suggest allele-specific differences at rs7132434 for AP-1 transcription factor binding. In an analysis of The Cancer Genome Atlas data, SNPs highly correlated with rs7132434 show allele-specific differences in BHLHE41 expression (trend P value=6.3 × 10(-7)). Cells overexpressing BHLHE41 produce larger mouse xenograft tumours, while RNA-seq analysis reveals that constitutively increased BHLHE41 induces expression of IL-11. We conclude that the RCC risk allele at 12p12.1 maps to rs7132434, a functional variant in an enhancer that upregulates BHLHE41 expression which, in turn, induces IL-11, a member of the IL-6 cytokine family.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Renal Cell/genetics , Chromosomes, Human, Pair 12/chemistry , Genetic Loci , Genetic Predisposition to Disease , Interleukin-11/genetics , Kidney Neoplasms/genetics , Alleles , Animals , Atlases as Topic , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Chromosomes, Human, Pair 12/metabolism , Computational Biology , Humans , Interleukin-11/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Neoplasm Transplantation , Polymorphism, Single Nucleotide , Protein Binding , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
14.
Blood ; 123(26): 4101-10, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24829201

ABSTRACT

The leukocyte adhesion cascade is important in chronic lymphocytic leukemia (CLL), as it controls migration of malignant cells into the pro-survival lymph node microenvironment. Circulating trisomy 12 CLL cells have increased expression of the integrins CD11a and CD49d, as well as CD38, but the tissue expression of these and other molecules, and the functional and clinical sequelae of these changes have not been described. Here, we demonstrate that circulating trisomy 12 CLL cells also have increased expression of the integrins CD11b, CD18, CD29, and ITGB7, and the adhesion molecule CD323. Notably, there was reduced expression of CD11a, CD11b, and CD18 in trisomy 12 cases with NOTCH1 mutations compared with wild type. Trisomy 12 cells also exhibit upregulation of intracellular integrin signaling molecules CALDAG-GEFI, RAP1B, and Ras-related protein ligand, resulting in enhanced very late antigen-4 [VLA-4] directed adhesion and motility. CD38 expression in CLL has prognostic significance, but the increased CD38 expression in trisomy 12 CLL cells must be taken into account in this subgroup, and the threshold of CD38 positivity should be raised to 40% for this marker to retain its prognostic value. In conclusion, trisomy 12 CLL cells exhibit functional upregulation of integrin signaling, with ß2-integrin expression being modulated by NOTCH1 mutation status.


Subject(s)
Gene Expression Regulation, Leukemic , Integrins/biosynthesis , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mutation , Neoplasm Proteins/metabolism , Neoplastic Cells, Circulating/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Up-Regulation , Aged , Cell Movement/genetics , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Integrins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplastic Cells, Circulating/pathology , Receptor, Notch1/genetics , Trisomy/genetics , Trisomy/pathology , rap GTP-Binding Proteins/genetics , rap GTP-Binding Proteins/metabolism
15.
J Proteome Res ; 13(7): 3160-5, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24831074

ABSTRACT

Following an official announcement of the Chromosome-centric Human Proteome Project (C-HPP), the Chromosome 12 (Ch12) Consortium has been established by five representative teams from five Asian countries including Thailand (Siriraj Hospital, Mahidol University), Singapore (National University of Singapore), Taiwan (Academia Sinica), Hong Kong (The Chinese University of Hong Kong), and India (Institute of Bioinformatics). We have worked closely together to extensively and systematically analyze all missing and known proteins encoded by Ch12 for their tissue/cellular/subcellular localizations. The target organs/tissues/cells include kidney, brain, gastrointestinal tissues, blood/immune cells, and stem cells. In the later phase, post-translational modifications and functional significance of Ch12-encoded proteins as well as their associations with human diseases (i.e., immune diseases, metabolic disorders, and cancers) will be defined. We have collaborated with other chromosome teams, Human Kidney and Urine Proteome Project (HKUPP), AOHUPO Membrane Proteomics Initiative, and other existing HUPO initiatives in the Biology/Disease-Based Human Proteome Project (B/D-HPP) to delineate functional roles and medical implications of Ch12-encoded proteins. The data set to be obtained from this multicountry consortium will be an important piece of the jigsaw puzzle to fulfill the missions and goals of the C-HPP and the global Human Proteome Project (HPP).


Subject(s)
Chromosomes, Human, Pair 12/genetics , Proteome/genetics , Chromosomes, Human, Pair 12/metabolism , Humans , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Organ Specificity , Proteome/metabolism , Research Design
16.
Am J Clin Nutr ; 97(3): 545-51, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23364009

ABSTRACT

BACKGROUND: Alcohol consumption is heritable, but genetic susceptibility to drinking behavior has not been investigated widely in genome-wide association studies. OBJECTIVE: We aimed to identify susceptibility loci for drinking behavior (drinkers compared with nondrinkers) in Han Chinese. DESIGN: We performed 2 genome-wide association studies including 1420 drinkers and 3590 nondrinkers in discovery, followed by a de novo replication analysis comprising 4896 drinkers and 13,293 nondrinkers. DNA samples of the subjects were collected for genotyping. RESULTS: The association results of drinking behavior (drinkers or nondrinkers) showed a cluster of single nucleotide polymorphisms at 12q24 in discovery (P < 5 × 10(-8)), with the strongest association for rs11066280 near C12orf51 (P-combined = 3.26 × 10(-215)). Moreover, we observed the association with drinking behavior for a functional variant in ALDH2 at 12q24 (rs671, P-discovery = 5.17 × 10(-35)). We also identified the association between rs11066280 and daily alcohol intake among drinkers (P-combined = 4.01 × 10(-21)). CONCLUSION: Our data indicate that common variants at 12q24 may contribute to the susceptibility of drinking behavior in Han Chinese.


Subject(s)
Alcohol Drinking/genetics , Asian People/genetics , Chromosomes, Human, Pair 12/genetics , Adult , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Chromosomes, Human, Pair 12/metabolism , Female , Gene Frequency , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Genotype , Humans , Linear Models , Logistic Models , Male , Middle Aged , Multigene Family , Multivariate Analysis , Polymorphism, Single Nucleotide , Risk Factors
17.
Am J Med Genet A ; 158A(12): 3046-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23169773

ABSTRACT

Identification of the isochromosome 12p (i(12p)) associated with Pallister-Killian syndrome is complicated by the low frequency of this supernumerary chromosome in PHA stimulated peripheral blood lymphocytes, and frequently requires cytogenetic analysis of fibroblast cells. Recently, it has been shown that array CGH techniques are able to detect tetrasomy 12p in peripheral blood, even when not identified by traditional cytogenetic techniques. We studied 15 patients with a previous cytogenetic and clinical diagnosis of Pallister-Killian syndrome using genome-wide SNP arrays to investigate the ability of this platform to identify the i(12p) in blood and tissue. Array analysis verified tetrasomy 12p in all samples from fibroblasts, but was only able to detect it in 46% of blood samples. The genotyping information available from the SNP arrays allowed for the detection of as low as 5% mosaicism, as well as suggesting a Meiosis II origin for the isochromosome in the majority of patients. Analysis of the percentage of abnormal cells with patient age at time of study suggests that the frequency of the i(12p) decreased with age in blood, but not in fibroblasts. These highlight the power of SNP arrays in detecting and characterizing the isochromosome 12p in Pallister-Killian syndrome as well as underscoring the important utility of traditional cytogenetic techniques.


Subject(s)
Chromosome Disorders/genetics , Chromosomes, Human, Pair 12 , Meiosis/genetics , Tetrasomy/genetics , Child, Preschool , Chromosome Disorders/diagnosis , Chromosome Disorders/metabolism , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Cohort Studies , Cytogenetic Analysis/methods , Fibroblasts/metabolism , Genotype , Humans , Infant , Infant, Newborn , Isochromosomes/genetics , Mosaicism , Polymorphism, Single Nucleotide , Tetrasomy/diagnosis
18.
J Clin Invest ; 122(11): 3990-4002, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23093776

ABSTRACT

Translocations are chromosomal rearrangements that are frequently associated with a variety of disease states and developmental disorders. We identified 2 families with brachydactyly type E (BDE) resulting from different translocations affecting chromosome 12p. Both translocations caused downregulation of the parathyroid hormone-like hormone (PTHLH) gene by disrupting the cis-regulatory landscape. Using chromosome conformation capturing, we identified a regulator on chromosome 12q that interacts in cis with PTHLH over a 24.4-megabase distance and in trans with the sex-determining region Y-box 9 (SOX9) gene on chromosome 17q. The element also harbored a long noncoding RNA (lncRNA). Silencing of the lncRNA, PTHLH, or SOX9 revealed a feedback mechanism involving an expression-dependent network in humans. In the BDE patients, the human lncRNA was upregulated by the disrupted chromosomal association. Moreover, the lncRNA occupancy at the PTHLH locus was reduced. Our results document what we believe to be a novel in cis- and in trans-acting DNA and lncRNA regulatory feedback element that is reciprocally regulated by coding genes. Furthermore, our findings provide a systematic and combinatorial view of how enhancers encoding lncRNAs may affect gene expression in normal development.


Subject(s)
Brachydactyly , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 17 , Gene Expression Regulation , Genetic Loci , RNA, Long Noncoding , Translocation, Genetic , Animals , Brachydactyly/diagnostic imaging , Brachydactyly/genetics , Brachydactyly/metabolism , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Female , Gene Silencing , Humans , Male , Mice , Mice, Transgenic , Parathyroid Hormone-Related Protein/biosynthesis , Parathyroid Hormone-Related Protein/genetics , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , Radiography , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics
19.
J Clin Invest ; 122(11): 4003-11, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23093777

ABSTRACT

The HELLP syndrome is a pregnancy-associated disease inducing hemolysis, elevated liver enzymes, and low platelets in the mother. Although the HELLP symptoms occur in the third trimester in the mother, the origin of the disease can be found in the first trimester fetal placenta. A locus for the HELLP syndrome is present on chromosome 12q23 near PAH. Here, by multipoint nonparametric linkage, pedigree structure allele sharing, and haplotype association analysis of affected sisters and cousins, we demonstrate that the HELLP locus is in an intergenic region on 12q23.2 between PMCH and IGF1. We identified a novel long intergenic noncoding RNA (lincRNA) transcript of 205,012 bases with (peri)nuclear expression in the extravillous trophoblast using strand-specific RT-PCR complemented with RACE and FISH. siRNA-mediated knockdown followed by RNA-sequencing, revealed that the HELLP lincRNA activated a large set of genes that are involved in the cell cycle. Furthermore, blocking potential mutation sites identified in HELLP families decreased the invasion capacity of extravillous trophoblasts. This is the first large noncoding gene to be linked to a Mendelian disorder with autosomal-recessive inheritance.


Subject(s)
Cell Cycle/genetics , Chromosomes, Human, Pair 12 , Genetic Diseases, Inborn , Genetic Loci , HELLP Syndrome , RNA, Long Noncoding , Trophoblasts/metabolism , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Family , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Genome-Wide Association Study , HELLP Syndrome/genetics , HELLP Syndrome/metabolism , HELLP Syndrome/pathology , Humans , Male , Pedigree , Pregnancy , Pregnancy Trimester, First/genetics , Pregnancy Trimester, First/metabolism , Pregnancy Trimester, Third/genetics , Pregnancy Trimester, Third/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Trophoblasts/pathology
20.
Appl Immunohistochem Mol Morphol ; 19(1): 54-61, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20881839

ABSTRACT

BACKGROUND: The human homolog of the mouse double minute 2 (MDM2) oncogene is amplified in about 20% of sarcomas. The measurement of the MDM2 amplification can aid in classification and may provide a predictive value for recently formulated therapies targeting MDM2. We have developed and validated an automated bright field dual-color in situ hybridization application to detect MDM2 gene amplification. DESIGN: A repeat-depleted MDM2 probe was constructed to target the MDM2 gene region at 12q15. A chromosome 12-specific probe (CHR12) was generated from a pα12H8 plasmid. The in situ hybridization assay was developed by using a dinitrophenyl-labeled MDM2 probe and a digoxigenin-labeled CHR12 probe on the Ventana Medical Systems' automated slide-staining platforms. The specificity of the MDM2 and CHR12 probes was shown on metaphase spreads and further validated against controls, including normal human tonsil and known MDM2-amplified samples. The assay performance was evaluated on a cohort of 100 formalin-fixed, paraffin-embedded specimens by using a conventional bright field microscope. RESULTS: Simultaneous hybridization and signal detection for MDM2 and CHR12 showed that both DNA targets were present in the same cells. One hundred soft tissue specimens were stained for MDM2 and CHR12. Although 26 of 29 lipomas were nonamplified and eusomic, MDM2 amplification was noted in 78% of atypical lipomatous tumors or well-differentiated liposarcomas. Five of 6 dedifferentiated liposarcoma cases were amplified for MDM2. MDM2 amplification was observed in 1 of 8 osteosarcomas; 3 showed CHR12 aneusomy. MDM2 amplification was present in 1 of 4 chondrosarcomas. Nine of 10 synovial sarcomas displayed no evidence of MDM2 amplification in most tumor cells. In pleomorphic sarcoma, not otherwise specified (pleomorphic malignant fibrous histiocytoma), MDM2 was amplified in 38% of cases, whereas 92% were aneusomic for CHR12. One alveolar rhabdomyosarcoma and 2 embryonal rhabdomyosarcomas displayed low-level aneusomy of CHR12 without net MDM2 amplifications. CONCLUSIONS: These results show that the use of the ISH MDM2 and CHR12 assay allows simultaneous analyses of the 2 DNA targets within the context of tissue morphology. This method combines the advantages of a fully automated protocol with bright field microscopy and has the potential for routine application in surgical pathology.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Gene Amplification , In Situ Hybridization, Fluorescence/methods , Proto-Oncogene Proteins c-mdm2/genetics , Sarcoma/genetics , Sarcoma/pathology , Aneuploidy , Animals , Chromosomes, Human, Pair 12/metabolism , Humans , In Situ Hybridization , Mice , Oligonucleotide Probes/chemistry , Oligonucleotide Probes/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Sarcoma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...