Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Cell ; 186(22): 4936-4955.e26, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37788668

ABSTRACT

Intrinsically disordered regions (IDRs) represent a large percentage of overall nuclear protein content. The prevailing dogma is that IDRs engage in non-specific interactions because they are poorly constrained by evolutionary selection. Here, we demonstrate that condensate formation and heterotypic interactions are distinct and separable features of an IDR within the ARID1A/B subunits of the mSWI/SNF chromatin remodeler, cBAF, and establish distinct "sequence grammars" underlying each contribution. Condensation is driven by uniformly distributed tyrosine residues, and partner interactions are mediated by non-random blocks rich in alanine, glycine, and glutamine residues. These features concentrate a specific cBAF protein-protein interaction network and are essential for chromatin localization and activity. Importantly, human disease-associated perturbations in ARID1B IDR sequence grammars disrupt cBAF function in cells. Together, these data identify IDR contributions to chromatin remodeling and explain how phase separation provides a mechanism through which both genomic localization and functional partner recruitment are achieved.


Subject(s)
Chromatin Assembly and Disassembly , Multiprotein Complexes , Nuclear Proteins , Humans , Chromatin , DNA-Binding Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism
2.
Genes Chromosomes Cancer ; 63(1): e23198, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37658696

ABSTRACT

Composite hemangioendothelioma is a rare, locally aggressive, and rarely metastasizing vascular neoplasm which affects both children and adults. Recently, a number of gene fusions including YAP1::MAML2, PTBP1::MAML2, and EPC1::PHC2 have been detected in a small subset of cases with or without neuroendocrine expression. Herein, we present four additional cases with novel in-frame fusions. The cohort comprises two females and two males with a wide age range at diagnosis (24-80 years). Two tumors were deep involving the right brachial plexus and mediastinum, while the remaining were superficial (right plantar foot and abdominal wall). The size ranged from 1.5 to 4.8 cm in greatest dimension. Morphologically, all tumors had an admixture of at least two architectural patterns including retiform hemangioendothelioma, hemangioma, epithelioid hemangioendothelioma, or angiosarcoma. The tumors were positive for endothelial markers CD31 (3/3), ERG (4/4), and D2-40 (1/4, focal), while SMA was expressed in 2/3 highlighting the surrounding pericytes. Synaptophysin showed immunoreactivity in 2/3 cases. One patient had a local recurrence after 40 months, while two patients had no evidence of disease 4 months post-resection. Targeted RNA sequencing detected novel in-frame fusions in each of the cases: HSPG2::FGFR1, YAP1::FOXR1, ACTB::MAML2, and ARID1B::MAML2. The two cases with neuroendocrine expression occurred as superficial lesions and harbored YAP1::FOXR1 and ARID1B::MAML2 fusions. Our study expands on the molecular spectrum of this enigmatic tumor, further enhancing our current understanding of the disease.


Subject(s)
Hemangioendothelioma, Epithelioid , Hemangioendothelioma , Hemangioma , Adult , Male , Child , Female , Humans , Young Adult , Middle Aged , Aged , Aged, 80 and over , Hemangioendothelioma/pathology , Hemangioendothelioma, Epithelioid/genetics , Base Sequence , Diagnosis, Differential , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Heterogeneous-Nuclear Ribonucleoproteins , Polypyrimidine Tract-Binding Protein
3.
Mod Pathol ; 37(1): 100374, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37925057

ABSTRACT

Dedifferentiated and undifferentiated ovarian carcinomas (DDOC/UDOC) are rare neoplasms defined by the presence of an undifferentiated carcinoma. In this study, we detailed the clinical, pathological, immunohistochemical, and molecular features of a series of DDOC/UDOC. We collected a multi-institutional cohort of 23 DDOC/UDOC and performed immunohistochemistry for core switch/sucrose nonfermentable (SWI/SNF) complex proteins (ARID1A, ARID1B, SMARCA4, and SMARCB1), mismatch repair (MMR) proteins, and p53. Array-based genome-wide DNA methylation and copy number variation analyses were performed on a subset of cases with comparison made to a previously reported cohort of undifferentiated endometrial carcinoma (UDEC), small cell carcinoma of the ovary, hypercalcemic type (SCCOHT), and tubo-ovarian high-grade serous carcinoma (HGSC). The age of all 23 patients with DDOC/UDOC ranged between 22 and 71 years (with an average age of 50 years), and a majority of them presented with extraovarian disease (16/23). Clinical follow-up was available for 19 patients. Except for 2 patients, the remaining 17 patients died from disease, with rapid disease progression resulting in mortality within a year in stage II-IV settings (median disease-specific survival of 3 months). Eighteen of 22 cases with interpretable immunohistochemistry results showed loss of expression of core SWI/SNF protein(s) that are expected to result in SWI/SNF complex inactivation as 10 exhibited coloss of ARID1A and ARID1B, 7 loss of SMARCA4, and 1 loss of SMARCB1. Six of 23 cases were MMR-deficient. Two of 20 cases exhibited mutation-type p53 immunoreactivity. Methylation profiles showed coclustering of DDOC/UDOC with UDEC, which collectively were distinct from SCCOHT and HGSC. However, DDOC/UDOC showed an intermediate degree of copy number variation, which was slightly greater, compared with SCCOHT but much less compared with HGSC. Overall, DDOC/UDOC, like its endometrial counterpart, is highly aggressive and is characterized by frequent inactivation of core SWI/SNF complex proteins and MMR deficiency. Its molecular profile overlaps with UDEC while being distinct from SCCOHT and HGSC.


Subject(s)
Brain Neoplasms , Carcinoma, Small Cell , Carcinoma , Colorectal Neoplasms , Endometrial Neoplasms , Neoplastic Syndromes, Hereditary , Ovarian Neoplasms , Female , Humans , Middle Aged , Young Adult , Adult , Aged , Tumor Suppressor Protein p53/genetics , DNA Copy Number Variations , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Carcinoma/pathology , Carcinoma, Ovarian Epithelial , Endometrial Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Ann Hematol ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990294

ABSTRACT

The MEF2D rearrangement is a recurrent chromosomal abnormality detected in approximately 2.4-5.3% of patients with acute B-cell lymphoblastic leukemia (B-ALL). Currently, MEF2D-rearranged B-ALL is not classified as an independent subtype in the WHO classification. Consequently, the clinical significance of MEF2D rearrangement in B-ALL remains largely unexplored. In this study, we retrospectively screened 260 B-ALL patients with RNA sequencing data collected between November 2018 and December 2022. Among these, 10 patients were identified with MEF2D rearrangements (4 with MEF2D::HNRNPUL1, 3 with MEF2D::BCL9, 1 with MEF2D::ARID1B, 1 with MEF2D::DAZAP1 and 1 with MEF2D::HNRNPM). Notably, HNRNPM and ARID1B are reported as MEF2D fusion partners for the first time. The patient with the MEF2D::HNRNPM fusion was resistant to chemotherapy and chimeric antigen receptor T-cell therapy and relapsed early after allogenic stem cell transplantation. The patient with MEF2D::ARID1B experienced early extramedullary relapse after diagnosis. All 10 patients achieved complete remission after induction chemotherapy. However, 9/10 (90%) of whom experienced relapse. Three of the 9 patients relapsed with aberrant expression of myeloid antigens. The median overall survival of these patients was only 11 months. This small cohort showed a high incidence of early relapse and short survival in patients with MEF2D rearrangements.

5.
Genes Dev ; 30(19): 2187-2198, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27737960

ABSTRACT

Oncogene-induced senescence (OIS) is a potent tumor suppressor mechanism. To identify senescence regulators relevant to cancer, we screened an shRNA library targeting genes deleted in hepatocellular carcinoma (HCC). Here, we describe how knockdown of the SWI/SNF component ARID1B prevents OIS and cooperates with RAS to induce liver tumors. ARID1B controls p16INK4a and p21CIP1a transcription but also regulates DNA damage, oxidative stress, and p53 induction, suggesting that SWI/SNF uses additional mechanisms to regulate senescence. To systematically identify SWI/SNF targets regulating senescence, we carried out a focused shRNA screen. We discovered several new senescence regulators, including ENTPD7, an enzyme that hydrolyses nucleotides. ENTPD7 affects oxidative stress, DNA damage, and senescence. Importantly, expression of ENTPD7 or inhibition of nucleotide synthesis in ARID1B-depleted cells results in re-establishment of senescence. Our results identify novel mechanisms by which epigenetic regulators can affect tumor progression and suggest that prosenescence therapies could be employed against SWI/SNF-mutated cancers.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cellular Senescence/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Apyrase/metabolism , Carcinoma, Hepatocellular/enzymology , Cell Line , Cell Line, Tumor , Epigenesis, Genetic/genetics , Female , Humans , Liver Neoplasms/enzymology , Male , Mice , Mice, Inbred C57BL , Mutation , RNA, Small Interfering/genetics
6.
Am J Med Genet C Semin Med Genet ; 193(3): e32056, 2023 09.
Article in English | MEDLINE | ID: mdl-37654076

ABSTRACT

Heterozygous ARID1B variants result in Coffin-Siris syndrome. Features may include hypoplastic nails, slow growth, characteristic facial features, hypotonia, hypertrichosis, and sparse scalp hair. Most reported cases are due to ARID1B loss of function variants. We report a boy with developmental delay, feeding difficulties, aspiration, recurrent respiratory infections, slow growth, and hypotonia without a clinical diagnosis, where a previously unreported ARID1B missense variant was classified as a variant of uncertain significance. The pathogenicity of this variant was refined through combined methodologies including genome-wide methylation signature analysis (EpiSign), Machine Learning (ML) facial phenotyping, and LIRICAL. Trio exome sequencing and EpiSign were performed. ML facial phenotyping compared facial images using FaceMatch and GestaltMatcher to syndrome-specific libraries to prioritize the trio exome bioinformatic pipeline gene list output. Phenotype-driven variant prioritization was performed with LIRICAL. A de novo heterozygous missense variant, ARID1B p.(Tyr1268His), was reported as a variant of uncertain significance. The ACMG classification was refined to likely pathogenic by a supportive methylation signature, ML facial phenotyping, and prioritization through LIRICAL. The ARID1B genotype-phenotype has been expanded through an extended analysis of missense variation through genome-wide methylation signatures, ML facial phenotyping, and likelihood-ratio gene prioritization.


Subject(s)
Abnormalities, Multiple , Hand Deformities, Congenital , Intellectual Disability , Micrognathism , Male , Humans , DNA-Binding Proteins/genetics , Muscle Hypotonia/pathology , Transcription Factors/genetics , Face/pathology , Abnormalities, Multiple/diagnosis , Micrognathism/genetics , Intellectual Disability/pathology , Hand Deformities, Congenital/genetics , Neck/pathology
7.
J Cell Sci ; 134(4)2021 02 26.
Article in English | MEDLINE | ID: mdl-33443092

ABSTRACT

The ARID1B (BAF250b) subunit of the human SWI/SNF chromatin remodeling complex is a canonical nuclear tumor suppressor. We employed in silico prediction, intracellular fluorescence and cellular fractionation-based subcellular localization analyses to identify the ARID1B nuclear localization signal (NLS). A cytoplasm-restricted ARID1B-NLS mutant was significantly compromised in its canonical transcription activation and tumor suppressive functions, as expected. Surprisingly however, cytoplasmic localization appeared to induce a gain of oncogenic function for ARID1B, as evidenced from several cell line- and mouse xenograft-based assays. Mechanistically, cytoplasm-localized ARID1B could bind c-RAF (RAF1) and PPP1CA causing stimulation of RAF-ERK signaling and ß-catenin (CTNNB1) transcription activity. ARID1B harboring NLS mutations derived from tumor samples also exhibited aberrant cytoplasmic localization and acquired a neo-morphic oncogenic function via activation of RAF-ERK signaling. Furthermore, immunohistochemistry on a tissue microarray revealed significant correlation of ARID1B cytoplasmic localization with increased levels of active forms of ERK1 and ERK2 (also known as MAPK3 and MAPK1) and of ß-catenin, as well as with advanced tumor stage and lymph node positivity in human primary pancreatic tumor tissues. ARID1B therefore promotes oncogenesis through cytoplasm-based gain-of-function mechanisms in addition to dysregulation in the nucleus.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Carcinogenesis , DNA-Binding Proteins , MAP Kinase Signaling System , Transcription Factors , Carcinogenesis/genetics , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Humans , Protein Phosphatase 1 , Signal Transduction , Transcription Factors/genetics , beta Catenin/genetics , beta Catenin/metabolism
8.
Am J Med Genet A ; 191(1): 22-28, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36177969

ABSTRACT

Coffin-Siris syndrome (CSS, MIM135900) is a rare multiple congenital anomaly syndrome caused by pathogenic variants in the BAF complex; up to 28% of patients have previously been reported to have seizures, however, a comprehensive review of epilepsy has not been undertaken in this population. The International CSS Patient Report Database was queried for patients with self-reported seizures, epilepsy, and EEG results. Data gathered included demographic data, pathogenic gene variants, seizure characteristics and treatments, and EEG findings. In addition, a PubMed search was performed using keywords "Coffin-Siris syndrome" and "epilepsy," "seizures," or "EEG." Results from relevant papers are reported. Twenty-four (7.2%) of 334 patients in the database reported having seizures, EEG abnormalities, and/or epilepsy. Median age of seizure onset was 2. 7 years. Fifteen of the 23 patients with seizures or epilepsy had an ARID1B causative variant. Seventeen patients (5.1%) reported EEG abnormalities, the majority of which were described as focal or multifocal (87.5%). In all but one patient, seizures were controlled on antiseizure medications (ASMs). The literature review yielded 311 unique CSS patients, 82 of which (26.4%) carried diagnoses of seizures or epilepsy. Details on seizure type(s), EEG findings, and response to treatment were limited.


Subject(s)
Abnormalities, Multiple , Epilepsy , Hand Deformities, Congenital , Intellectual Disability , Micrognathism , Humans , Micrognathism/diagnosis , Micrognathism/genetics , Micrognathism/pathology , Hand Deformities, Congenital/complications , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/genetics , DNA-Binding Proteins/genetics , Intellectual Disability/diagnosis , Face/abnormalities , Neck/abnormalities , Genetic Association Studies , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/epidemiology , Abnormalities, Multiple/genetics , Epilepsy/complications , Epilepsy/diagnosis , Epilepsy/epidemiology , Seizures/epidemiology , Seizures/genetics , Seizures/pathology
9.
Am J Med Genet A ; 191(2): 605-611, 2023 02.
Article in English | MEDLINE | ID: mdl-36416235

ABSTRACT

Coffin-Siris Syndrome (CSS) is a rare multi-system dominant condition with a variable clinical presentation mainly characterized by hypoplasia/aplasia of the nail and/or distal phalanx of the fifth digit, coarse facies, hirsutism/hypertrichosis, developmental delay and intellectual disability of variable degree and growth impairment. Congenital anomalies may include cardiac, genitourinary and central nervous system malformations whereas congenital diaphragmatic hernia (CDH) is rarely reported. The genes usually involved in CSS pathogenesis are ARID1B (most frequently), SMARCA4, SMARCB1, ARID1A, SMARCE1, DPF2, and PHF6. Here, we present two cases of CSS presenting with CDH, for whom Whole Exome Sequencing (WES) identified two distinct de novo heterozygous causative variants, one in ARID1B (case 1) and one in SMARCA4 (case 2). Due to the rarity of CDH in CSS, in both cases the occurrence of CDH did not represent a predictive sign of CSS but, on the other hand, prompted genetic testing before (case 1) or independently (case 2) from the clinical hypothesis of CSS. We provide further evidence of the association between CSS and CDH, reviewed previous cases from literature and discuss possible functional links to related conditions.


Subject(s)
Abnormalities, Multiple , Hand Deformities, Congenital , Hernias, Diaphragmatic, Congenital , Intellectual Disability , Micrognathism , Humans , Face/abnormalities , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Micrognathism/diagnosis , Micrognathism/genetics , Micrognathism/pathology , Hernias, Diaphragmatic, Congenital/diagnosis , Hernias, Diaphragmatic, Congenital/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/genetics , Hand Deformities, Congenital/pathology , Neck/abnormalities , DNA Helicases/genetics , Nuclear Proteins , Transcription Factors/genetics , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins/genetics
10.
Cell Biol Int ; 47(7): 1222-1228, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36883912

ABSTRACT

In a few reports, ARID1B/A mutation was found in neuroblastoma. We analyzed the clinical characteristics, clinical efficacy, and prognosis of three children with high-risk refractory neuroblastoma (NB) with somatic ARID1B gene mutation. The whole exon sequencing results showed that there were involved in transcription, DNA synthesis, and repair of ARID1B gene mutations. All mutation sites were located in the promoter region of the exon: ARID1B (p.A460) mutation was found in cases 1 and 2, and ARID1B (p.V215G) mutation was found in cases 1 and 3. The nucleic acid site of ARID1B (p.A460) mutation was c.1379 (exon1) C > G, and the nucleic acid site of ARID1B (p.V215G) mutation was c.644 (exon1) T > G. The meningeal metastasis in case 1 turned negative after 4 cycles of intrathecal injection combined with chemotherapy. However, the child died of agranulocytosis combined with sepsis during the 5th cycle of chemotherapy. Case 2 achieved complete remission (CR). Case 3 achieved CR after chemotherapy, surgery, metaiodobenzylguanidine, and 3F-8 (Naxitamab) immunotherapy after the initial diagnosis. The mediastinum and lymph node metastasis occurred during the 6-month observation period after stopping treatment. He achieved very good partial remission after individualized chemotherapy and surgical treatment. ARID1B is a component protein of the SWI/SNF chromatin-remodeling complex that participates in the occurrence of a variety of tumors by regulating DNA repair and synthesis. ARID1B nucleic acid mutation (p.A460, p.V215G) in the promoter region of three children may contribute to the poor prognosis of NB children.


Subject(s)
DNA-Binding Proteins , Neuroblastoma , Male , Child , Humans , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Mutation/genetics , Exons
11.
J Pathol ; 257(2): 140-145, 2022 06.
Article in English | MEDLINE | ID: mdl-35218556

ABSTRACT

SWI/SNF (SWItch/Sucrose Non-Fermentable) complex deficiency has been reported in a wide variety of cancers and is often associated with an undifferentiated phenotype. In the gynecologic tract SWI/SNF-deficient cancers are diagnostically challenging and little is known about their cellular origins. Here we show that undifferentiated endometrial carcinoma (UDEC), SMARCA4-deficient uterine sarcoma (SDUS), and ovarian small cell carcinoma, hypercalcemic type (SCCOHT) harbor distinct DNA methylation signatures despite shared morphology and SWI/SNF inactivation. Our results indicate that the cellular context is an important determinant of the epigenetic landscape, even in the setting of core SWI/SNF deficiency, and therefore methylation profiling may represent a useful diagnostic tool in undifferentiated, SWI/SNF-deficient cancers. Furthermore, applying copy number analyses and group-wise differential methylation analyses including endometrioid endometrial carcinomas and extracranial malignant rhabdoid tumors, we uncover analogous molecular features in SDUS and SCCOHT in contrast to UDEC. These results suggest that SDUS and SCCOHT represent chromosomally stable SWI/SNF-deficient cancers of the gynecologic tract, which are within the broader spectrum of malignant rhabdoid tumors. © 2022 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)
Carcinoma, Endometrioid , Carcinoma, Small Cell , Endometrial Neoplasms , Hypercalcemia , Lung Neoplasms , Rhabdoid Tumor , Small Cell Lung Carcinoma , Carcinoma, Endometrioid/genetics , DNA Helicases/genetics , DNA Methylation , Endometrial Neoplasms/genetics , Female , Humans , Nuclear Proteins/genetics , Transcription Factors/genetics , United Kingdom
12.
Genet Med ; 24(8): 1753-1760, 2022 08.
Article in English | MEDLINE | ID: mdl-35579625

ABSTRACT

PURPOSE: Genome-wide sequencing is increasingly being performed during pregnancy to identify the genetic cause of congenital anomalies. The interpretation of prenatally identified variants can be challenging and is hampered by our often limited knowledge of prenatal phenotypes. To better delineate the prenatal phenotype of Coffin-Siris syndrome (CSS), we collected clinical data from patients with a prenatal phenotype and a pathogenic variant in one of the CSS-associated genes. METHODS: Clinical data was collected through an extensive web-based survey. RESULTS: We included 44 patients with a variant in a CSS-associated gene and a prenatal phenotype; 9 of these patients have been reported before. Prenatal anomalies that were frequently observed in our cohort include hydrocephalus, agenesis of the corpus callosum, hypoplastic left heart syndrome, persistent left vena cava, diaphragmatic hernia, renal agenesis, and intrauterine growth restriction. Anal anomalies were frequently identified after birth in patients with ARID1A variants (6/14, 43%). Interestingly, pathogenic ARID1A variants were much more frequently identified in the current prenatal cohort (16/44, 36%) than in postnatal CSS cohorts (5%-9%). CONCLUSION: Our data shed new light on the prenatal phenotype of patients with pathogenic variants in CSS genes.


Subject(s)
Hand Deformities, Congenital , Intellectual Disability , Micrognathism , Abnormalities, Multiple , Chromosomal Proteins, Non-Histone/genetics , Face/abnormalities , Genetic Association Studies , Hand Deformities, Congenital/genetics , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Micrognathism/genetics , Neck/abnormalities , Phenotype
13.
Histopathology ; 79(2): 160-167, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33432679

ABSTRACT

AIMS: Genomic inactivation of ARID1B in ARID1A-inactivated tumour and genomic inactivation of SMARCB1 represent two recurrent mechanisms, core SWItch/sucrose non-fermentable (SWI/SNF) complex inactivation, that are associated with de-differentiation in endometrial carcinoma. Approximately one-third of dedifferentiated/undifferentiated endometrial carcinomas (DDEC/UEC) show loss of ARID1B expression with a minor subset showing loss of SMARCB1 expression, but little is known regarding the specificity of ARID1B or SMARCB1 loss in gynaecological tract tumours in general. The aim of this study was to examine the frequency of ARID1B and SMARCB1 loss by immunohistochemistry in a series of gynaecological tract epithelial/mesenchymal neoplasms. METHODS AND RESULTS: We evaluated 1849 tumours that included 748 endometrial carcinomas, 101 uterine carcinosarcomas/adenosarcomas, 64 uterine sarcomas, 221 cervical carcinomas and 715 ovarian carcinomas/borderline tumours by tissue microarrays (TMA). We observed ARID1B loss in 35 of 86 (41%) and SMARCB1 loss in three of 86 (3%) DDEC/UEC, but not in any other uterine tumour types examined. ARID1B-deficient DDEC/UEC also showed concurrent loss of ARID1A expression. All SMARCB1-deficient tumours showed loss of MLH1 and PMS2, while 29 of 35 ARID1B-deficient tumours showed loss of MLH1 and PMS2 or loss of MSH6. All ovarian carcinomas/borderline tumours and cervical carcinomas showed intact expression of ARID1B and SMARCB1. CONCLUSION: Our findings indicate that the loss of expression of ARID1B or SMARCB1 by immunohistochemistry is highly specific for undifferentiated carcinoma among tumours of the upper gynaecological tract and cervix, and therefore can be used to identify these highly aggressive malignant tumours.


Subject(s)
Carcinoma/diagnosis , Cell Dedifferentiation , DNA-Binding Proteins/deficiency , Genital Neoplasms, Female/diagnosis , SMARCB1 Protein/deficiency , Transcription Factors/deficiency , Biomarkers, Tumor/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Cell Dedifferentiation/genetics , Cohort Studies , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/pathology , Humans , Immunohistochemistry , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Sarcoma/diagnosis , Sarcoma/metabolism , Sarcoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
14.
Am J Med Genet A ; 185(7): 2250-2261, 2021 07.
Article in English | MEDLINE | ID: mdl-33768696

ABSTRACT

Coffin-Siris syndrome (CSS, MIM# 1359200) is a multisystem congenital disorder characterized by coarse facial features, hypoplasia of the fifth digits and nails, and intellectual disability. It is a genetically heterogeneous condition caused by pathogenic variants in genes encoding proteins of the BAF (BRG1-associated factors) chromatin modeling complex and its downstream transcriptional factor. To date over 220 CSS individuals with pathogenic variants found have been described in the literature. This case series reported 18 molecularly confirmed Chinese individuals (17 with ARIDIB (OMIM*614556) variants and one with SMARCB1 (OMIM*601607) variant) from 17 unrelated families in Hong Kong. The clinical features of these 18 Chinese CSS patients together with two previously reported Chinese patients with ARID1B variants were reviewed. Among the 19 Chinese patients with ARID1B variants, our data suggested a lower prevalence of feeding problem, autistic features, agenesis of corpus callosum (ACC) or partial/hypoplasia of corpus callosum, and sparse hair when compared with previous reports. There was appearing higher prevalence of digital hypoplasia. Digital hypoplasia was observed to become less noticeable with time in some patients. This report highlighted the age-dependent phenotypic presentation of CSS and ethnicity-related effect on ARID1B-CSS phenotype. Moreover, this series included the first family with molecularly confirmed maternal somatic mosaicism of ARID1B variant leading to familial CSS recurrence.


Subject(s)
Abnormalities, Multiple/genetics , DNA-Binding Proteins/genetics , Face/abnormalities , Genetic Predisposition to Disease , Hand Deformities, Congenital/genetics , Intellectual Disability/genetics , Micrognathism/genetics , Neck/abnormalities , Transcription Factors/genetics , Abnormalities, Multiple/physiopathology , Adolescent , Adult , Child , Child, Preschool , Face/physiopathology , Female , Genotype , Hand Deformities, Congenital/physiopathology , Humans , Infant , Intellectual Disability/physiopathology , Male , Micrognathism/physiopathology , Neck/physiopathology , Phenotype , Young Adult
15.
Neurol Sci ; 42(2): 727-729, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33006724

ABSTRACT

Coffin-Siris syndrome is a rare genetic disorder defined by the presence of particular facial traits, congenital malformations, intellectual disability, and speech impairment. Epilepsy in Coffin-Siris syndrome has only occasionally been reported, and its features are poorly defined. We provide a detailed description of the clinical and instrumental findings of three patients with Coffin-Siris syndrome and epilepsy. The clinical diagnosis in our patients was confirmed by molecular analysis, which identified the presence of de novo mutations of ARID1B and SMARCB1 genes, in two patients and one patient, respectively. All the patients presented with epilepsy, with a mean age of seizure onset of 5.5 years. Seizures were brief and had a focal onset with secondary generalization. Electroencephalographic recording documented a unilateral, and less commonly bilateral, paroxysmal activity in the temporal, parietal, and occipital regions. Clinical response to anticonvulsive therapy was satisfactory, with a low rate of seizure recurrence. Our case series contributes to delineate the phenotype of Coffin-Siris syndrome. We wish this report could pave the way for further studies that will better define the prevalence and clinical manifestations of epilepsy in this rare syndrome.


Subject(s)
Epilepsy , Intellectual Disability , Abnormalities, Multiple , Child, Preschool , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins , Epilepsy/genetics , Face/abnormalities , Hand Deformities, Congenital , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Micrognathism , Neck/abnormalities
16.
Reprod Domest Anim ; 56(2): 374-380, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33320378

ABSTRACT

Crosstalk between the oocyte and surrounding cumulus cells (CCs) is essential for the production of competent oocytes. Previous studies have analysed the relative transcript abundance in oocytes derived from small (SF: <3 mm diameter)- and medium-sized (MF: 3-6 mm diameter) follicles to determine the potential use of SF-derived oocytes in assisted reproductive technologies (ART). The aim of this study was to examine the relative transcript abundance of CCs obtained from cumulus-oocyte complexes (COCs) derived from SF and MF. Nine genes were selected according to their importance for developmental competence: AT-rich interaction domain 1B (ARID1B), bone morphogenic protein receptor 2 (BMPR2), CD44, follicle-stimulating hormone receptor (FSHR), follistatin (FST), inhibin beta-A (INHBA), luteinizing hormone receptor (LHR), nuclear receptor subfamily 2 group F member 6 (NR2F6) and vascular endothelial growth factor A (VEGFA). The expression of these genes was analysed by RT-qPCR. The results pointed to significant differences in five genes, and the relative transcript abundance of SF-derived CCs was lower in the case of INHBA, but higher in FSHR, FST, LHR and NR2F6 compared with MF-derived CCs. We provide information of gene activity in the porcine CCs from different sized follicles, thus improving our understanding of oocyte biology and providing new markers that identify viable and competent oocytes.


Subject(s)
Cumulus Cells/metabolism , Gene Expression Profiling , Ovarian Follicle/physiology , Animals , Female , Oocytes/cytology , Oocytes/physiology , RNA, Messenger/analysis , Sus scrofa/physiology
17.
Hum Mutat ; 41(5): 1012-1024, 2020 05.
Article in English | MEDLINE | ID: mdl-31981384

ABSTRACT

Haploinsufficiency of ARID1B (AT-rich interaction domain 1B) has been involved in autism spectrum disorder, nonsyndromic and syndromic intellectual disability, and corpus callosum agenesis. Growth impairment is a major clinical feature caused by ARID1B mutations; however, the mechanistic link has not been elucidated. Here, we confirm that growth delay is a common characteristic of patients with ARID1B mutations, which may be associated with dysregulation of the Wnt/ß-catenin signaling pathway. An analysis of patients harboring pathogenic variants of ARID1B revealed that nearly half had short stature and nearly all had below-average height. Moreover, the percentage of patients with short stature increased with age. Knockdown of arid1b in zebrafish embryos markedly reduced body length and perturbed the expression of both chondrogenic and osteogenic genes including sox9a, col2a1a, runx2b, and col10a1. Knockout of Arid1b in chondrogenic ATDC5 cells inhibited chondrocyte proliferation and differentiation. Finally, Wnt/ß-catenin signaling was perturbed in Arid1b-depleted zebrafish embryos and Arid1b knockout ATDC5 cells. These data indicate that ARID1B modulates bone growth possibly via regulation of the Wnt/ß-catenin pathway, and may be an appropriate target for gene therapy in disorders of growth and development.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Growth Disorders/diagnosis , Growth Disorders/genetics , Mutation , Transcription Factors/genetics , Wnt Signaling Pathway , Alleles , Animals , Animals, Genetically Modified , Body Weights and Measures , Cell Differentiation/genetics , Child, Preschool , DNA-Binding Proteins/metabolism , Facies , Gene Knockdown Techniques , Gene Silencing , Genetic Association Studies/methods , Genotype , Growth Charts , Growth Disorders/metabolism , Humans , Loss of Function Mutation , Male , Phenotype , Transcription Factors/metabolism , Zebrafish
18.
Mol Med ; 26(1): 78, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32791957

ABSTRACT

INTRODUCTION: Patients with advanced non-small cell lung cancer (NSCLC) benefit from treatment with immune checkpoint inhibitors (ICIs). Biomarkers such as programmed death-ligand 1 (PD-L1), the tumor mutational burden (TMB) and the mismatch repair (MMR) status are used to predict the prognosis of ICIs therapy. Nevertheless, novel biomarkers need to be further investigated, and a systematic prognostic model is needed for the evaluation of the survival risks of ICIs treatment. METHODS: A cohort of 240 patients who received ICIs from the cBioPortal for Cancer Genomics was evaluated in this research. Clinical information and targeted sequencing data were acquired for analyses. The Kaplan-Meier plot method was used to perform survival analyses, and selected variables were then confirmed by a novel nomogram constructed by the "rms" package of R software. RESULTS: Seven percent of the NSCLC patients harbored ARID1A mutations, while 4% of the NSCLC patients harbored ARID1B mutations. Mutations in ARID1A and ARID1B were confirmed to be associated with sensitivity to ICIs. Patients harboring these mutations were found to have a better response to treatment (ARID1A: P = 0.045; ARID1B: P = 0.034) and prolonged progression-free survival (ARID1B: P = 0.032). Here, a novel nomogram was constructed to predict the prognosis of ICIs treatment. Elevation of the TMB, enhanced expression of PD-L1 and activation of the antigen presentation process and cellular immunity were found to be correlated with ARID1A and ARID1B mutations. CONCLUSION: ARID1A and ARID1B could serve as novel biomarkers for the prognosis and sensitivity to ICIs of advanced NSCLC.


Subject(s)
Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/genetics , DNA-Binding Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Protein Subunits/genetics , Transcription Factors/genetics , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Proteins/metabolism , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Male , Mutation , Neoplasm Staging , Nomograms , Prognosis , Protein Subunits/metabolism , Survival Analysis , Transcription Factors/chemistry , Transcription Factors/metabolism , Treatment Outcome
19.
Histopathology ; 77(1): 46-54, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32413172

ABSTRACT

AIMS: Undifferentiated carcinoma refers to an epithelial malignancy that lacks morphological evidence of differentiation. Recent studies have implicated the loss of constitutively expressed switch/sucrose non-fermenting (SWI/SNF) complex subunits in undifferentiated carcinomas of the gastrointestinal tract and other sites. In this study we examine the expression of SWI/SNF and mismatch repair (MMR) proteins in a series of undifferentiated carcinomas from the gastrointestinal tract and the pancreas. METHODS AND RESULTS: We searched pathology databases from four Canadian health centres for primary undifferentiated carcinoma from gastrointestinal and pancreatic resection specimens. Upon review of 31 cases, 19 were confirmed to be undifferentiated carcinomas (eight colonic, six gastric, three pancreatic, one appendiceal and one duodenal). Immunohistochemical analysis of SMARCA4, SMARCA2, SMARCB1, ARID1A, ARID1B, MSH2, MSH6, MLH1 and PMS2 was performed on whole sections. Five of 19 (26%) showed loss of core SWI/SNF proteins (two loss of SMARCA4, one loss of SMARCB1 and two concurrent loss of ARID1A and ARID1B). SMARCA4, SMARCB1, or ARID1A/ARID1B-deficient undifferentiated carcinoma consistently exhibited sheet-like growth pattern, with cellular discohesion and rhabdoid morphology. Nine of 17 undifferentiated carcinomas tested were MMR-deficient by immunohistochemistry. In comparison, none of the 12 poorly differentiated carcinomas that were originally diagnosed as undifferentiated carcinomas showed loss of SMARCA4, SMARCA2, SMARCB1 or ARID1B. CONCLUSIONS: Undifferentiated gastrointestinal/pancreatic carcinomas show frequent loss of expression of SWI/SNF complex proteins. The loss of these core components of SWI/SNF complex may contribute to the arrest of cellular differentiation, resulting in the undifferentiated histology and aggressive clinical behaviour.


Subject(s)
Biomarkers, Tumor/analysis , Gastrointestinal Neoplasms/pathology , Pancreatic Neoplasms/pathology , Transcription Factors/biosynthesis , Adult , Aged , Aged, 80 and over , Female , Gastrointestinal Neoplasms/metabolism , Humans , Male , Middle Aged , Pancreatic Neoplasms/metabolism , Transcription Factors/analysis , Pancreatic Neoplasms
20.
Genet Med ; 21(6): 1295-1307, 2019 06.
Article in English | MEDLINE | ID: mdl-30349098

ABSTRACT

PURPOSE: Pathogenic variants in ARID1B are one of the most frequent causes of intellectual disability (ID) as determined by large-scale exome sequencing studies. Most studies published thus far describe clinically diagnosed Coffin-Siris patients (ARID1B-CSS) and it is unclear whether these data are representative for patients identified through sequencing of unbiased ID cohorts (ARID1B-ID). We therefore sought to determine genotypic and phenotypic differences between ARID1B-ID and ARID1B-CSS. In parallel, we investigated the effect of different methods of phenotype reporting. METHODS: Clinicians entered clinical data in an extensive web-based survey. RESULTS: 79 ARID1B-CSS and 64 ARID1B-ID patients were included. CSS-associated dysmorphic features, such as thick eyebrows, long eyelashes, thick alae nasi, long and/or broad philtrum, small nails and small or absent fifth distal phalanx and hypertrichosis, were observed significantly more often (p < 0.001) in ARID1B-CSS patients. No other significant differences were identified. CONCLUSION: There are only minor differences between ARID1B-ID and ARID1B-CSS patients. ARID1B-related disorders seem to consist of a spectrum, and patients should be managed similarly. We demonstrated that data collection methods without an explicit option to report the absence of a feature (such as most Human Phenotype Ontology-based methods) tended to underestimate gene-related features.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Abnormalities, Multiple/genetics , Adolescent , Adult , Child , Child, Preschool , Chromosomal Proteins, Non-Histone/genetics , Exome , Face/abnormalities , Female , Genetic Association Studies/methods , Genetic Variation/genetics , Hand Deformities, Congenital/genetics , Humans , Infant , Infant, Newborn , Intellectual Disability/genetics , Male , Micrognathism/genetics , Middle Aged , Mutation , Neck/abnormalities , Penetrance
SELECTION OF CITATIONS
SEARCH DETAIL