Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.396
Filter
1.
Pathologica ; 116(3): 163-169, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38979590

ABSTRACT

The 5th WHO classification of thoracic tumours includes thoracic SMARCA4-deficient undifferentiated tumour (SMARCA4-UT) among the "other epithelial tumours of the lung" chapter. Herein, we present a case of undifferentiated thoracic neoplasm with retention of SMARCA4 expression, lack of NUT fusion protein and loss of SMARCB1/INI1 expression. After presenting the clinical and pathological features of the tumour, we carried out a review of the literature on the same topic. Albeit very rare, we believe this entity should be included in the heterogeneous group of undifferentiated neoplasms of the thorax.


Subject(s)
DNA Helicases , SMARCB1 Protein , Thoracic Neoplasms , Transcription Factors , Humans , SMARCB1 Protein/deficiency , SMARCB1 Protein/genetics , Transcription Factors/genetics , Transcription Factors/deficiency , Thoracic Neoplasms/pathology , Thoracic Neoplasms/genetics , DNA Helicases/deficiency , DNA Helicases/genetics , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Male , Female , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Middle Aged , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/diagnosis
2.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000439

ABSTRACT

LIM homeobox 4 (LHX4) is a transcription factor crucial for anterior pituitary (AP) development. Patients with LHX4 mutation suffer from combined pituitary hormone deficiency (CPHD), short statures, reproductive and metabolic disorders and lethality in some cases. Lhx4-knockout (KO) mice fail to develop a normal AP and die shortly after birth. Here, we characterize a zebrafish lhx4-KO model to further investigate the importance of LHX4 in pituitary gland development and regulation. At the embryonic and larval stages, these fish express lower levels of tshb mRNA compared with their wildtype siblings. In adult lhx4-KO fish, the expressions of pituitary hormone-encoding transcripts, including growth hormone (gh), thyroid stimulating hormone (tshb), proopiomelanocortin (pomca) and follicle stimulating hormone (fshb), are reduced, the pomca promoter-driven expression in corticotrophs is dampened and luteinizing hormone (lhb)-producing gonadotrophs are severely depleted. In contrast to Lhx4-KO mice, Lhx4-deficient fish survive to adulthood, but with a reduced body size. Importantly, lhx4-KO males reach sexual maturity and are reproductively competent, whereas the females remain infertile with undeveloped ovaries. These phenotypes, which are reminiscent of those observed in CPHD patients, along with the advantages of the zebrafish for developmental genetics research, make this lhx4-KO fish an ideal vertebrate model to study the outcomes of LHX4 mutation.


Subject(s)
Hypopituitarism , LIM-Homeodomain Proteins , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/deficiency , Hypopituitarism/genetics , Hypopituitarism/metabolism , Male , Female , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/deficiency , Gene Knockout Techniques , Pituitary Gland/metabolism , Disease Models, Animal , Animals, Genetically Modified
3.
Orphanet J Rare Dis ; 19(1): 237, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877473

ABSTRACT

BACKGROUND: SMARCA4, as one of the subunits of the SWI/SNF chromatin remodeling complex, drives SMARCA4-deficient tumors. Gastric SMARCA4-deficient tumors may include gastric SMARCA4-deficient carcinoma and gastric SMARCA4-deficient undifferentiated tumor (SMARCA4-UT). Gastric SMARCA4-UT is rare and challenging to diagnose in clinical practice. The present report aims to provide insight into the clinicopathological characteristics and genetic alterations of gastric SMARCA4-UTs. RESULTS: We retrospectively reported four rare cases of gastric SMARCA4-UTs. All four cases were male, aged between 61 and 82 years. These tumors presented as ulcerated and transmural masses with infiltration, staged as TNM IV in cases 1, 2 and 4, and TNM IIIA in case 3. Pathologically, four cases presented solid architecture with undifferentiated morphology. Cases 2 and 3 showed focal necrosis and focal rhabdoid morphology. Immunohistochemical staining showed negative expression of epithelial markers and deficient expression of SMARCA4. Furthermore, positivity for Syn (cases 1, 2 and 3) and SALL4 (cases 1 and 2) were observed. Mutant p53 expression occurred in four cases, resulting in strong and diffuse staining of p53 expression in cases 1, 2 and 4, and complete loss in case 3. The Ki67 proliferative index exceeded 80%. 25% (1/4, case 4) of cases had mismatch repair deficiency (dMMR). Two available cases (cases 1 and 3) were detected with SMRACA4 gene alterations. The response to neoadjuvant therapy was ineffective in case 1. CONCLUSIONS: Gastric SMARCA4-UT is a rare entity of gastric cancer with a poor prognosis, predominantly occurs in male patients. The tumors are typically diagnosed at advanced stages and shows a solid architecture with undifferentiated morphology. Negative expression of epithelial markers and complete loss of SMARCA4 immunoexpression are emerging as a useful diagnostic tool for rare gastric SMARCA4-UTs.


Subject(s)
DNA Helicases , Nuclear Proteins , Stomach Neoplasms , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/deficiency , Male , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , DNA Helicases/genetics , DNA Helicases/deficiency , DNA Helicases/metabolism , Middle Aged , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/deficiency , Aged, 80 and over , Retrospective Studies , Aged
4.
Cancer Rep (Hoboken) ; 7(6): e2127, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923369

ABSTRACT

BACKGROUND: SMARCA4 is a component gene of the SWI/SNF (SWItch/Sucrose NonFermentable) chromatin remodeling complex; undifferentiated tumors associated with its functional deletion have been described in several organs. However, no established treatment for these tumors currently exists. CASE: In this study, we report a case of a SMARCA4-deficient undifferentiated urothelial carcinoma with high PD-L1 expression that was effectively treated with nivolumab after early relapse following treatment for non-invasive bladder cancer. The histological morphology of the rhabdoid-like undifferentiated tumor of unknown primary led us to suspect a SWI/SNF-deficient tumor, and subsequent immunostaining led to the diagnosis of a SMARCA4-deficient undifferentiated tumor. This effort also led to the identification of the developmental origin of this SMARCA4-deficient undifferentiated tumor as a non-invasive bladder cancer. We also carried out a detailed immune phenotypic assay on peripheral T cells. In brief, a phenotypic change of CD8+T cells from naive to terminally differentiated effector memory cells was observed. CONCLUSION: Regardless of the organ of cancer origin or cancer type, SWI/SNF-deficient tumors should be suspected in undifferentiated and dedifferentiated tumors, and immune checkpoint inhibitors may be considered as a promising treatment option for this type of tumor. The pathogenesis of SMARCA4-deficient anaplastic tumors awaits further elucidation for therapeutic development.


Subject(s)
B7-H1 Antigen , DNA Helicases , Nivolumab , Nuclear Proteins , Transcription Factors , Urinary Bladder Neoplasms , Humans , DNA Helicases/genetics , DNA Helicases/deficiency , Transcription Factors/genetics , Transcription Factors/deficiency , Transcription Factors/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/diagnosis , B7-H1 Antigen/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Nivolumab/therapeutic use , Male , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/pathology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/diagnosis , Aged , Treatment Outcome
5.
Nucleic Acids Res ; 52(10): 5698-5719, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38587186

ABSTRACT

AT-rich interaction domain protein 1A (ARID1A), a SWI/SNF chromatin remodeling complex subunit, is frequently mutated across various cancer entities. Loss of ARID1A leads to DNA repair defects. Here, we show that ARID1A plays epigenetic roles to promote both DNA double-strand breaks (DSBs) repair pathways, non-homologous end-joining (NHEJ) and homologous recombination (HR). ARID1A is accumulated at DSBs after DNA damage and regulates chromatin loops formation by recruiting RAD21 and CTCF to DSBs. Simultaneously, ARID1A facilitates transcription silencing at DSBs in transcriptionally active chromatin by recruiting HDAC1 and RSF1 to control the distribution of activating histone marks, chromatin accessibility, and eviction of RNAPII. ARID1A depletion resulted in enhanced accumulation of micronuclei, activation of cGAS-STING pathway, and an increased expression of immunomodulatory cytokines upon ionizing radiation. Furthermore, low ARID1A expression in cancer patients receiving radiotherapy was associated with higher infiltration of several immune cells. The high mutation rate of ARID1A in various cancer types highlights its clinical relevance as a promising biomarker that correlates with the level of immune regulatory cytokines and estimates the levels of tumor-infiltrating immune cells, which can predict the response to the combination of radio- and immunotherapy.


Subject(s)
Chromatin , DNA Repair , DNA-Binding Proteins , Immunity , Transcription Factors , Humans , Cell Line, Tumor , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Homologous Recombination/genetics , Immunity/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/immunology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Trans-Activators , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Clin Cancer Res ; 30(9): 1708-1711, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38416596

ABSTRACT

Small-cell lung carcinoma (SCLC) cell lines have been widely utilized as a preclinical model of this highly aggressive disease. However, since their creation decades ago, novel tumor entities have been defined that might clinicopathologically mimic SCLC, which notably includes thoracic SMARCA4-deficient undifferentiated tumor (SMARCA4-UT). Multiomic reassessment of the presumed SCLC cell lines with high YAP1 expression reveals that nearly all of these tumors represent unsuspected SMARCA4-UT. See related article by Ng et al., p. 1846.


Subject(s)
DNA Helicases , Lung Neoplasms , Nuclear Proteins , Small Cell Lung Carcinoma , Transcription Factors , Humans , DNA Helicases/genetics , DNA Helicases/deficiency , Small Cell Lung Carcinoma/diagnosis , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Transcription Factors/genetics , Transcription Factors/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Diagnosis, Differential , Thoracic Neoplasms/diagnosis , Thoracic Neoplasms/genetics , Thoracic Neoplasms/pathology , Cell Line, Tumor
7.
Pathology ; 56(4): 504-515, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38413251

ABSTRACT

SMARCA4 mutation has emerged as a marker of poor prognosis in lung cancer and has potential predictive value in cancer treatment, but recommendations for which patients require its investigation are lacking. We comprehensively studied SMARCA4 alterations and the clinicopathological significance in a large cohort of immunohistochemically-subtyped non-small cell lung cancer (NSCLC). A total of 1416 patients was studied for the presence of SMARCA4 deficiency by immunohistochemistry (IHC). Thereafter, comprehensive sequencing of tumours was performed for 397 of these patients to study the mutational spectrum of SWI/SNF and SMARCA4 aberrations. IHC evidence of SMARCA4 deficiency was found in 2.9% of NSCLC. Of the sequenced tumours, 38.3% showed aberration in SWI/SNF complex, and 9.3% had SMARCA4 mutations. Strikingly, SMARCA4 aberrations were much more prevalent in large cell carcinoma (LCC) than other histological tumour subtypes. SMARCA4-deficient and SMARCA4-mutated tumours accounted for 40.5% and 51.4% of all LCC, respectively. Multivariable analyses confirmed SMARCA4 mutation was an independent prognostic factor in lung cancer. The immunophenotype of a subset of these tumours frequently showed TTF1 negativity and HepPAR1 positivity. SMARCA4 mutation or its deficiency was associated with positive smoking history and poor prognosis. It also demonstrated mutual exclusion with EGFR mutation. Taken together, the high incidence of SMARCA4 aberrations in LCC may indicate its diagnostic and prognostic value. Our study established the necessity of SMARCA4 IHC in the identification of SMARCA4-aberrant tumours, and this may be of particular importance in LCC and tumours without known driver events.


Subject(s)
Carcinoma, Large Cell , Carcinoma, Non-Small-Cell Lung , DNA Helicases , Nuclear Proteins , Transcription Factors , Female , Humans , Male , Biomarkers, Tumor/genetics , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , DNA Helicases/genetics , DNA Helicases/deficiency , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Prognosis , Transcription Factors/genetics , Transcription Factors/deficiency
8.
Turk Patoloji Derg ; 40(2): 128-133, 2024.
Article in English | MEDLINE | ID: mdl-38265099

ABSTRACT

OBJECTIVE: SMARCA4-deficient undifferentiated tumor (SMARCA4-UT) is a highly malignant neoplasm with an undifferentiated or rhabdoid phenotype, posing a diagnostic challenge. This case report aims to create awareness about this rare neoplasm while dealing with cases presenting with undifferentiated morphology. CASE REPORT: A 55-year-old gentleman with constitutional symptoms and lymphadenopathy. Imaging revealed a mass lesion in the right upper lobe of the lung. A biopsy of the cervical lymph node showed diffusely effaced architecture replaced by sheets of undifferentiated pleomorphic cells with vesicular nuclei, prominent nucleoli, eosinophilic cytoplasm, and multiple necrotic foci. An extensive immunohistochemistry (IHC) panel was applied, which showed positivity for synaptophysin, vimentin, and focal CD34 and EMA expression. Other markers like pan-cytokeratin, p40, TTF1, CD56, INSM1, calretinin, CD45, SOX10, S100, CD30, CD117, SMA, and Desmin were negative, with INI1 retained. The IHC panel excluded the morphological differentials of carcinoma, lymphoma, rhabdomyosarcoma, melanoma, and germ cell tumor. Further literature review led to the possibility of the SMARCA4-UT entity, which had a morphology and IHC profile similar to the present case. Testing for SMARCA4 (BRG-1) by IHC showed a complete loss in the tumor cells, favoring the diagnosis of Thoracic SMARCA4-deficient undifferentiated tumor (SMARCA4-UT). CONCLUSION: SMARCA4-UTs are rare, highly aggressive, and poorly differentiated thoracic tumors. Recognizing them is vital as there is potential for therapeutic interventions such as immunotherapy and SMARCA4-targeted therapies, offering promising prospects for the future.


Subject(s)
Biomarkers, Tumor , DNA Helicases , Nuclear Proteins , Transcription Factors , Humans , Male , Transcription Factors/genetics , Transcription Factors/deficiency , Middle Aged , DNA Helicases/deficiency , DNA Helicases/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Immunohistochemistry , Thoracic Neoplasms/pathology , Thoracic Neoplasms/genetics , Thoracic Neoplasms/chemistry
9.
Int J Gynecol Pathol ; 43(4): 354-361, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38113031

ABSTRACT

SMARCA4 gene encodes BRG1 , a member of the SWItch/sucrose non-fermentable protein family involved in epigenetic transcriptional regulation of important cellular processes. In the uterine corpus, SMARCA4 / BRG1 deficiency is associated with a novel class of undifferentiated uterine sarcomas, characterized by younger age onset, rhabdoid histology, focal phyllodiform architecture, high-risk pathologic findings, and dismal prognosis. Herein, we report a case of a 34-year-old Asian woman with a SMARCA4 / BRG1 -deficient uterine tumor fulfilling the clinicopathologic features of an undifferentiated uterine sarcoma. However, the tumor exhibited several unique features that have not been previously emphasized, including (1) conspicuous phyllodiform architecture recapitulating conventional adenosarcoma, (2) rhabdoid tumor cells forming cords and keratin-positive cohesive epithelial islands, and (3) cooccurrence with a spatially distinct and discrete endometrial complex atypical hyperplasia from the rest of the proliferation. By immunohistochemistry, the tumor cells were diffusely positive for synaptophysin, whereas BRG1 was lost. Pertinent molecular findings included frameshift mutations in the SMARCA4 gene, mutations in histone modification and chromatin remodeling genes, including KMT2C , ARID1B , KAT6A , and NCOR1 , and mutations in Wnt signaling involving APC and CTNNB1 . Copy number gain in MDM2 and CDK4 were also identified. The tumor mutation burden was intermediate (6.8/MB) and it was microsatellite stable. On balance, our case exhibited morphologic and molecular features that overlap with (1) an undifferentiated uterine sarcoma, (2) an adenosarcoma with sarcomatous overgrowth, and (3) a mixed adenosarcoma and undifferentiated endometrial carcinoma. These hybrid features further expand the molecular-morphologic spectrum of SMARCA4 / BRG1 -deficient uterine neoplasms.


Subject(s)
Adenosarcoma , DNA Helicases , Nuclear Proteins , Transcription Factors , Uterine Neoplasms , Humans , Female , DNA Helicases/genetics , DNA Helicases/deficiency , Transcription Factors/genetics , Transcription Factors/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Adult , Adenosarcoma/pathology , Adenosarcoma/genetics , Uterine Neoplasms/pathology , Uterine Neoplasms/genetics , Immunohistochemistry , Carcinoma/pathology , Carcinoma/genetics
10.
Nature ; 620(7976): 1047-1053, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37459895

ABSTRACT

Zygotic genome activation (ZGA) activates the quiescent genome to enable the maternal-to-zygotic transition1,2. However, the identity of transcription factors that underlie mammalian ZGA in vivo remains elusive. Here we show that OBOX, a PRD-like homeobox domain transcription factor family (OBOX1-OBOX8)3-5, are key regulators of mouse ZGA. Mice deficient for maternally transcribed Obox1/2/5/7 and zygotically expressed Obox3/4 had a two-cell to four-cell arrest, accompanied by impaired ZGA. The Obox knockout defects could be rescued by restoring either maternal and zygotic OBOX, which suggests that maternal and zygotic OBOX redundantly support embryonic development. Chromatin-binding analysis showed that Obox knockout preferentially affected OBOX-binding targets. Mechanistically, OBOX facilitated the 'preconfiguration' of RNA polymerase II, as the polymerase relocated from the initial one-cell binding targets to ZGA gene promoters and distal enhancers. Impaired polymerase II preconfiguration in Obox mutants was accompanied by defective ZGA and chromatin accessibility transition, as well as aberrant activation of one-cell polymerase II targets. Finally, ectopic expression of OBOX activated ZGA genes and MERVL repeats in mouse embryonic stem cells. These data thus demonstrate that OBOX regulates mouse ZGA and early embryogenesis.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Genome , Homeodomain Proteins , Transcription Factors , Zygote , Animals , Mice , Chromatin/genetics , Chromatin/metabolism , Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Genome/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Mutation , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Zygote/metabolism
11.
Article in English | MEDLINE | ID: mdl-35289322

ABSTRACT

BACKGROUND: SMARCA2 (SWI/SNF Related, Matrix Associated, Actin Dependent Regulator of Chromatin, Subfamily A, Member 2) is an important ATPase catalytic subunit in the switch-sucrose nonfermenting (SWI/SNF) complex. However, its relationship with the pathological features of NSCLC and its prognosis remain unclear. METHODS: We retrospectively reviewed 2390 patients with surgically resected NSCLC, constructed tissue microarrays (TMAs) and performed immunohistochemical assays. We analyzed the correlation of SAMRCA2 with clinicopathological features and evaluated its prognostic value. RESULTS: Among 2390 NSCLC cases, the negative expression ratios of SAMRCA2, SMARCA4, ARID1A, ARID1B and INI1 were 9.3%, 1.8%, 1.2%, 0.4% and 0%, respectively. In NSCLC, male sex, T3 and T4 stage, moderate and poor differentiation, tumor ≥ 2 cm, Ki67 ≥ 15%, SOX-2 negative expression, middle lobe lesion and adenocarcinoma were relative risk factors affecting SMARCA2-negative expression. In lung adenocarcinomas, high-grade nuclei, histological morphology of acinar and papillary, solid and micropapillary and TTF-1-negative expression were relative risk factors affecting SMARCA2-negative expression. Kaplan-Meier survival analysis showed that the OS was shorter in the SMARCA2-negative group. Multivariate survival analysis revealed that SMARCA2-negative expression was an independent factor correlated with a poor prognosis in NSCLC. CONCLUSION: In conclusion, SMARCA2-negative expression is an independent predictor of a poor outcome of NSCLC and is a potential target for NSCLC treatment.


Subject(s)
Adenosine Triphosphatases , Carcinoma, Non-Small-Cell Lung/genetics , Transcription Factors , Adenosine Triphosphatases/metabolism , Humans , Male , Retrospective Studies , Transcription Factors/deficiency , Transcription Factors/genetics
12.
Front Immunol ; 13: 829239, 2022.
Article in English | MEDLINE | ID: mdl-35185921

ABSTRACT

Introduction: Hyper IgE syndromes (HIES) are a group of rare primary immunodeficiency characterized by high levels of serum IgE, cold abscesses, pulmonary infections, and eczema. ZNF341 deficiency was described in 2018 in 11 patients clinically diagnosed previously with HIES. Eight of those patients, all offspring of consanguineous couples, are from three families who live in a Muslim village in Israel which has approximately 15,000 residents. Objective: Our study aimed to evaluate the prevalence of ZNF341 mutation in the population of the village. Methods: Three hundred DNA samples of females were included in the study. The samples belong to females that were referred to the Meir Medical Center for prenatal genetic testing before pregnancy, during 2017-2019: 200 samples were from the village, and 100 samples of Muslim females were from other villages.All samples were tested by Sanger sequencing for the ZNF341 mutation (c.904C>T, NM_001282933.1). Results: Heterozygous nonsense mutation in ZNF341 was found in ten samples (5%) of the study group compared to zero in the control group (p<0.01). Conclusion: The carrier frequency of the mutation in ZNF341 in the studied village population is 1:20. This high frequency is probably due to founder mutation and consanguineous marriages.


Subject(s)
Job Syndrome/epidemiology , Job Syndrome/genetics , Transcription Factors/genetics , Carrier State , Codon, Nonsense , Eczema , Female , Humans , Immunoglobulin E/immunology , Islam , Israel/epidemiology , Job Syndrome/immunology , Population , Transcription Factors/deficiency
13.
Development ; 149(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35179180

ABSTRACT

Specification of primordial germ cells requires a proportion of the cells in the posterior of the epiblast to reacquire pluripotency. A new paper in Development describes how OVOL2 is involved in regulating the balance between mesodermal fate and germ cell fate during gastrulation. We caught up with the first author, Yuki Naitou, and corresponding author, Katsuhiko Hayashi (Osaka University), to find out more about the paper and their future research.


Subject(s)
Germ Cells/metabolism , Research Personnel/psychology , Transcription Factors/metabolism , Animals , Authorship , Epithelial-Mesenchymal Transition , Gastrulation , Germ Cells/cytology , Germ Layers/cytology , Germ Layers/metabolism , Humans , Male , Mesoderm/cytology , Mesoderm/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
14.
Cell Rep ; 38(7): 110384, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172142

ABSTRACT

Impaired production of thymic regulatory T cells (Tregs) is implicated in the development of Aire-dependent autoimmunity. Because Tregs require agonistic T cell receptor stimuli by self-antigens to develop, reduced expression of self-antigens from medullary thymic epithelial cells (mTECs) has been considered to play a major role in the reduced Treg production in Aire deficiency. Here, we show that mTECs abnormally express co-inhibitory receptor CTLA-4 if Aire is non-functional. Upon binding with CD80/CD86 ligands expressed on thymic dendritic cells (DCs), the ectopically expressed CTLA-4 from Aire-deficient mTECs removes the CD80/CD86 ligands from the DCs. This attenuates the ability of DCs to provide co-stimulatory signals and to present self-antigens transferred from mTECs, both of which are required for Treg production. Accordingly, impaired production of Tregs and organ-specific autoimmunity in Aire-deficient mice are rescued by the depletion of CTLA-4 expression from mTECs. Our studies illuminate the significance of mTEC-DC interaction coordinated by Aire for the establishment of thymic tolerance.


Subject(s)
Autoimmunity , CTLA-4 Antigen/metabolism , Thymus Gland/cytology , Animals , Antigens, CD/metabolism , Autoantigens/metabolism , Epithelial Cells/metabolism , Ligands , Mice, Inbred BALB C , Mice, Inbred C57BL , Organ Specificity , Stromal Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism , AIRE Protein
15.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166348, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35032595

ABSTRACT

ARPKD is a genetically inherited kidney disease that manifests by bilateral enlargement of cystic kidneys and liver fibrosis. It shows a range of severity, with 30% of individuals dying early on and the majority having good prognosis if they survive the first year of life. The reasons for this variability remain unclear. Two genes have been shown to cause ARPKD when mutated, PKHD1, mutations in which lead to most of ARPKD cases and DZIP1L, which is associated with moderate ARPKD. This mini review will explore the genetics of ARPKD and discuss potential genetic modifiers and phenocopies that could affect diagnosis.


Subject(s)
Polycystic Kidney, Autosomal Recessive/pathology , Adaptor Proteins, Signal Transducing/genetics , Animals , Humans , Polycystic Kidney, Autosomal Recessive/genetics , Polymorphism, Genetic , Prognosis , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
16.
Nat Commun ; 13(1): 149, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013221

ABSTRACT

Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.


Subject(s)
Cachexia/genetics , Endomyocardial Fibrosis/genetics , Heart Failure/genetics , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Transcription Factors/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/metabolism , Aged , Aged, 80 and over , Animals , Cachexia/metabolism , Cachexia/physiopathology , Cachexia/prevention & control , Case-Control Studies , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/physiopathology , Endomyocardial Fibrosis/prevention & control , Female , Gene Expression Regulation , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Failure/prevention & control , Heart Function Tests , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/deficiency , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Muscular Atrophy/prevention & control , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency
17.
Development ; 149(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35029669

ABSTRACT

In mammals, primordial germ cells (PGCs), the origin of the germ line, are specified from the epiblast at the posterior region where gastrulation simultaneously occurs, yet the functional relationship between PGC specification and gastrulation remains unclear. Here, we show that OVOL2, a transcription factor conserved across the animal kingdom, balances these major developmental processes by repressing the epithelial-to-mesenchymal transition (EMT) that drives gastrulation and the upregulation of genes associated with PGC specification. Ovol2a, a splice variant encoding a repressor domain, directly regulates EMT-related genes and, consequently, induces re-acquisition of potential pluripotency during PGC specification, whereas Ovol2b, another splice variant missing the repressor domain, directly upregulates genes associated with PGC specification. Taken together, these results elucidate the molecular mechanism underlying allocation of the germ line among epiblast cells differentiating into somatic cells through gastrulation. This article has an associated 'The people behind the papers' interview.


Subject(s)
Embryonic Development/genetics , Gastrulation/genetics , Germ Cells/metabolism , Transcription Factors/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Lineage , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Germ Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Up-Regulation , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
18.
Nature ; 602(7895): 129-134, 2022 02.
Article in English | MEDLINE | ID: mdl-35082446

ABSTRACT

Differentiation proceeds along a continuum of increasingly fate-restricted intermediates, referred to as canalization1,2. Canalization is essential for stabilizing cell fate, but the mechanisms that underlie robust canalization are unclear. Here we show that the BRG1/BRM-associated factor (BAF) chromatin-remodelling complex ATPase gene Brm safeguards cell identity during directed cardiogenesis of mouse embryonic stem cells. Despite the establishment of a well-differentiated precardiac mesoderm, Brm-/- cells predominantly became neural precursors, violating germ layer assignment. Trajectory inference showed a sudden acquisition of a non-mesodermal identity in Brm-/- cells. Mechanistically, the loss of Brm prevented de novo accessibility of primed cardiac enhancers while increasing the expression of neurogenic factor POU3F1, preventing the binding of the neural suppressor REST and shifting the composition of BRG1 complexes. The identity switch caused by the Brm mutation was overcome by increasing BMP4 levels during mesoderm induction. Mathematical modelling supports these observations and demonstrates that Brm deletion affects cell fate trajectory by modifying saddle-node bifurcations2. In the mouse embryo, Brm deletion exacerbated mesoderm-deleted Brg1-mutant phenotypes, severely compromising cardiogenesis, and reveals an in vivo role for Brm. Our results show that Brm is a compensable safeguard of the fidelity of mesoderm chromatin states, and support a model in which developmental canalization is not a rigid irreversible path, but a highly plastic trajectory.


Subject(s)
Cell Differentiation , Cell Lineage , Mesoderm/cytology , Mesoderm/metabolism , Myocytes, Cardiac/cytology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Embryo, Mammalian , Epigenesis, Genetic , Female , Gene Expression Regulation , Male , Mice , Myocardium/metabolism , Neurogenesis , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/metabolism , Octamer Transcription Factor-6/metabolism , Phenotype , Repressor Proteins/metabolism , Stem Cells/cytology , Time Factors , Transcription Factors/deficiency , Transcription Factors/genetics
20.
Exp Cell Res ; 410(2): 112969, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34883111

ABSTRACT

MEL1 (MDS1/EVI1-like gene 1/PRDM16), a zinc finger protein, is located near the chromosomal breakpoint at 1p36 in human acute myeloid leukemia (AML) cells with the t (1; 3) (p36; q21) translocation. Mel1/Prdm16 is not only a causative gene of leukemia, but also has multiple regulatory functions, such as the regulation of fat metabolism. To investigate the function of Mel1/Prdm16, we generated Mel1/Prdm16-deficient mice, but homozygous deficiency (Mel1/Prdm16-/-) was embryonic lethal at E 11.5. Heterozygous mice showed abnormal cartilage and bone formation in the postnatal skull and long bones, suggesting that Mel1/Prdm16 expression plays an important role in bone development. In osteoblast and chondrocyte cell lines, Mel1/Prdm16 promotes the differentiation of chondrocytes and regulates the differentiation of osteoblasts. Transient repression of the master regulator Runx2 is required for chondrocyte differentiation at an early stage of differentiation. However, in Mel1/Prdm16-suppressed ATDC5 cells, the initial suppression of Runx2 was lacking and its expression was upregulated at the beginning of differentiation, suggesting that chondrogenic differentiation is suppressed in Mel1/Prdm16+/- mesenchymal progenitor cells because Runx2 expression is upregulated during the early stage of differentiation. Thus, the Mel1/Prdm16 gene may be involved in the early repression of Runx2 expression during osteochondral differentiation and promote chondrogenic differentiation.


Subject(s)
Bone and Bones/anatomy & histology , Bone and Bones/cytology , Cell Differentiation , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Bone Morphogenetic Protein 2/metabolism , Cartilage/pathology , Core Binding Factor Alpha 1 Subunit/metabolism , DNA-Binding Proteins/deficiency , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Models, Biological , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Signal Transduction , Transcription Factors/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...