Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 270
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.
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
3.
Front Immunol ; 15: 1371379, 2024.
Article in English | MEDLINE | ID: mdl-38881888

ABSTRACT

SMARCA4-deficient undifferentiated tumor (SMARCA4-dUT) is a devastating subtype of thoracic tumor with SMARCA4 inactivation and is characterized by rapid progression, poor prognosis, and high risk of postoperative recurrence. However, effective treatments for SMARCA4-dUT are lacking. Herein, we describe a patient with SMARCA4-dUT who exhibited an impressive response to the anti-programmed cell death protein-1 (PD-1) antibody (tislelizumab) in combination with conventional chemotherapy (etoposide and cisplatin). To the best of our knowledge, this is the first case of SMARCA4-dUT treated with chemotherapy, comprising etoposide and cisplatin, combined with anti-PD-1 inhibitors. Immunotherapy combined with etoposide and cisplatin may be a promising strategy to treat SMARCA4-dUT.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , DNA Helicases , Transcription Factors , Humans , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , DNA Helicases/genetics , DNA Helicases/deficiency , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Transcription Factors/genetics , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Etoposide/therapeutic use , Etoposide/administration & dosage , Male , Cisplatin/therapeutic use , Cisplatin/administration & dosage , Treatment Outcome , Female
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.
Lung Cancer ; 192: 107818, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38763102

ABSTRACT

INTRODUCTIONS: The 2021 WHO Classification of Thoracic Tumors recognized SMARCA4-deficient undifferentiated thoracic tumors (SMARCA4-dUT) as a distinct entity that shows a striking overlap in demographic and molecular profiles with SMARCA4-deficient non-small lung cancer (SMARCA4-dNSCLC). The implications of SMARCA4 deficiency based on immunohistochemistry remain unclear. We aimed to investigate molecular characteristics of SMARCA4-deficient thoracic tumors (SDTT) and explore optimal therapeutics. METHODS: From June.15, 2018, to Nov.15, 2023, a large cohort including patients diagnosed with SMARCA4-deficient (N = 196) and SMARCA4-intact (N = 438) thoracic tumors confirmed by immunohistochemistry at SYSUCC were screened. Clinicopathologic and molecular characteristics were identified and compared. External SRRSH cohort (N = 34) was combined into a pooled cohort to compare clinical outcome of first-line therapy efficacy. RESULTS: SDTT is male predominance with smoking history, high tumor burden, and adrenal metastases. The relationship between SMARCA4 mutation and protein expression is not completely parallel. The majority of SMARCA4-deficient patients harbor truncating (Class-I) SMARCA4 mutations, whereas class-II alterations and wild-type also exist. Compared with SMARCA4-intact thoracic tumors, patients with SDTT displayed a higher tumor mutation burden (TMB) and associated with a shorter median OS (16.8 months vs. Not reached; P < 0.001). Notably, SMARCA4 protein deficiency, rather than genetic mutations, played a decisive role in these differences. SDTT is generally resistant to chemotherapy, while sensitive to chemoimmunotherapy (median PFS: 7.5 vs. 3.5 months, P < 0.001). In particular, patients with SMARCA4 deficient thoracic tumors treated with paclitaxel-based chemoimmunotherapy achieved a longer median PFS than those with pemetrexed-based chemoimmunotherapy (10.0 vs. 7.3 months, P = 0.028). CONCLUSIONS: SMARCA4 protein deficiency, rather than genetic mutations, played a decisive role in its characteristics of higher TMB and poor prognosis. Chemoimmunotherapy serves as the optimal option in the current treatment regimen. Paclitaxel-based chemoimmunotherapy performed better than those with pemetrexed-based chemoimmunotherapy.


Subject(s)
DNA Helicases , Lung Neoplasms , Nuclear Proteins , Thoracic Neoplasms , Transcription Factors , Humans , DNA Helicases/genetics , DNA Helicases/deficiency , Transcription Factors/genetics , Male , Female , Thoracic Neoplasms/genetics , Thoracic Neoplasms/pathology , Thoracic Neoplasms/drug therapy , Thoracic Neoplasms/therapy , Middle Aged , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Aged , Mutation , Prognosis , Adult , Biomarkers, Tumor/genetics
6.
FEBS Open Bio ; 14(7): 1087-1100, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38720471

ABSTRACT

Helicase POLQ-like (HELQ) is a DNA helicase essential for the maintenance of genome stability. A recent study identified two HELQ missense mutations in some cases of infertile men. However, the functions of HELQ in the process of germline specification are not well known and whether its function is conserved between mouse and human remains unclear. Here, we revealed that Helq knockout (Helq-/-) could significantly reduce the efficiency of mouse primordial germ cell-like cell (PGCLC) induction. In addition, Helq-/- embryonic bodies exhibited a severe apoptotic phenotype on day 6 of mouse PGCLC induction. p53 inhibitor treatment could partially rescue the generation of mouse PGCLCs from Helq mutant mouse embryonic stem cells. Finally, the genetic ablation of HELQ could also significantly impede the induction of human PGCLCs. Collectively, our study sheds light on the involvement of HELQ in the induction of both mouse and human PGCLCs, providing new insights into the mechanisms underlying germline differentiation and the genetic studies of human fertility.


Subject(s)
Germ Cells , Animals , Mice , Germ Cells/metabolism , Humans , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Helicases/deficiency , Cell Differentiation/genetics , Mice, Knockout , Male , Apoptosis/genetics
7.
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
8.
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
9.
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
10.
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
11.
Oncologist ; 27(6): 501-511, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35278076

ABSTRACT

BACKGROUND: Thoracic SMARCA4-deficient undifferentiated tumors (SMARCA4-UT) are aggressive neoplasms. Data linking BAF alterations with tumor microenvironment (TME) and efficacy of immune checkpoint inhibitors (ICI) are contradictory. The TME of SMARCA4-UT and their response to ICI are unknown. MATERIALS AND METHODS: Patients diagnosed with SMARCA4-UT in our institution were included. Immunostainings for tertiary lymphoid structures (TLS), immune cell markers, and checkpoints were assessed. Validation was performed using an independent transcriptome dataset including SMARCA4-UT, non-small cell lung cancers (NSCLC) with/without SMARCA4 mutations, and unclassified thoracic sarcomas (UTS). CXCL9 and PD-L1 expressions were assessed in NSCLC and thoracic fibroblast cell lines, with/without SMARCA4 knockdown, treated with/without interferon gamma. RESULTS: Nine patients were identified. All samples but one showed no TLS, consistent with an immune desert TME phenotype. Four patients received ICI as part of their treatment, but the only one who responded, had a tumor with a TLS and immune-rich TME. Unsupervised clustering of the validation cohort using immune cell scores identified 2 clusters associated with cell ontogeny and immunity (cluster 1 enriched for NSCLC independently of SMARCA4 status (n = 9/10; P = .001); cluster 2 enriched for SMARCA4-UT (n = 11/12; P = .005) and UTS (n = 5/5; P = .0005). SMARCA4 loss-of-function experiments revealed interferon-induced upregulation of CXCL9 and PD-L1 expression in the NSCLC cell line with no effect on the thoracic fibroblast cell line. CONCLUSION: SMARCA4-UT mainly have an immune desert TME with limited efficacy to ICI. TME of SMARCA4-driven tumors varies according to the cell of origin questioning the interplay between BAF alterations, cell ontogeny and immunity.


Subject(s)
Carcinoma, Non-Small-Cell Lung , DNA Helicases , Immune Checkpoint Inhibitors , Lung Neoplasms , Nuclear Proteins , Sarcoma , Soft Tissue Neoplasms , Thoracic Neoplasms , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Biomarkers, Tumor/immunology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , DNA Helicases/deficiency , DNA Helicases/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Nuclear Proteins/deficiency , Nuclear Proteins/immunology , Sarcoma/drug therapy , Sarcoma/immunology , Sarcoma/pathology , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/immunology , Soft Tissue Neoplasms/pathology , Thoracic Neoplasms/drug therapy , Thoracic Neoplasms/immunology , Thoracic Neoplasms/pathology , Transcription Factors/immunology , Tumor Microenvironment/immunology
12.
J Clin Invest ; 132(2)2022 01 18.
Article in English | MEDLINE | ID: mdl-35040435

ABSTRACT

Inborn errors of nucleic acid metabolism often cause aberrant activation of nucleic acid sensing pathways, leading to autoimmune or autoinflammatory diseases. The SKIV2L RNA exosome is cytoplasmic RNA degradation machinery that was thought to be essential for preventing the self-RNA-mediated interferon (IFN) response. Here, we demonstrate the physiological function of SKIV2L in mammals. We found that Skiv2l deficiency in mice disrupted epidermal and T cell homeostasis in a cell-intrinsic manner independently of IFN. Skiv2l-deficient mice developed skin inflammation and hair abnormality, which were also observed in a SKIV2L-deficient patient. Epidermis-specific deletion of Skiv2l caused hyperproliferation of keratinocytes and disrupted epidermal stratification, leading to impaired skin barrier with no appreciable IFN activation. Moreover, Skiv2l-deficient T cells were chronically hyperactivated and these T cells attacked lesional skin as well as hair follicles. Mechanistically, SKIV2L loss activated the mTORC1 pathway in both keratinocytes and T cells. Both systemic and topical rapamycin treatment of Skiv2l-deficient mice ameliorated epidermal hyperplasia and skin inflammation. Together, we demonstrate that mTORC1, a classical nutrient sensor, also senses cytoplasmic RNA quality control failure and drives autoinflammatory disease. We also propose SKIV2L-associated trichohepatoenteric syndrome (THES) as a new mTORopathy for which sirolimus may be a promising therapy.


Subject(s)
Autoimmune Diseases/immunology , Cytoplasm/immunology , Diarrhea, Infantile/immunology , Fetal Growth Retardation/immunology , Hair Diseases/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , RNA Stability/immunology , RNA/immunology , Animals , Autoimmune Diseases/genetics , Cytoplasm/genetics , DNA Helicases/deficiency , DNA Helicases/immunology , Diarrhea, Infantile/genetics , Facies , Fetal Growth Retardation/genetics , Hair Diseases/genetics , Inflammation/genetics , Inflammation/immunology , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Knockout , RNA/genetics , RNA Stability/genetics
13.
Pathol Oncol Res ; 27: 1610003, 2021.
Article in English | MEDLINE | ID: mdl-34970085

ABSTRACT

Small cell carcinoma of hypercalcemic type (SCCOHT) is a rare gynaecological neoplasm, originating mostly in the ovaries. Cervical origin of this very aggressive malignancy with unknown histogenesis is an extremely rare condition, without published management recommendations. Alterations in SMARCA4 gene are supposed to play the major role in SCCOHT oncogenesis and their identification is crucial for the diagnosis. Adequate genetic counselling of the patients and their families seems to be of great importance. Optimal management and treatment approaches are not known yet but may extremely influence the prognosis of young female patients that suffer from this very resistant disease. Nowadays, a translational research seems to be the key for the further diagnostic and treatment strategies of SCCOHT. The purpose of the case report is to provide practical information and useful recommendations on the diagnosis, management, and treatment of SMARCA4-deficient carcinoma of the uterine cervix resembling SCCOHT.


Subject(s)
Carcinoma, Small Cell/metabolism , DNA Helicases/deficiency , Hypercalcemia/metabolism , Nuclear Proteins/deficiency , Transcription Factors/deficiency , Uterine Cervical Neoplasms/metabolism , Adolescent , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Carcinoma, Small Cell/diagnosis , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/therapy , DNA Helicases/genetics , Fatal Outcome , Female , Humans , Hypercalcemia/diagnosis , Hypercalcemia/genetics , Hypercalcemia/therapy , Mutation , Nuclear Proteins/genetics , Transcription Factors/genetics , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/therapy
14.
Virchows Arch ; 479(6): 1209-1219, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34350470

ABSTRACT

Mimickers of neuroendocrine neoplasms (NEN) include a number of important pitfall tumors. Here, we describe our experience with mesenchymal mimics of NENs to illustrate their spectrum and draw the attention particularly to a group of mesenchymal/non-epithelial neoplasms (MN) that combine epithelioid histology with neuroendocrine (NE-) features and peculiar genetic abnormalities. In a consultation series of 4498 cases collected between 2009 and 2021, 2099 neoplasms expressing synaptophysin and/or chromograninA were reviewed and analyzed. A total of 364 (18%) were diagnosed as non-NENs, while the remaining tumors were NEN. The group of mesenchymal/non-epithelial neoplasms with NE-features (MN-NE) included 31/364 (8%) cases. These mostly malignant neoplasms showed an epithelioid morphology. While all but one tumor expressed synaptophysin, mostly patchy, only 10/29 (34%) co-expressed chromograninA. A total of 13/31 (42%) of the MN-NE showed EWSR1-related gene fusions (6 Ewing sarcomas, 5 clear cell sarcomas, and 1 desmoplastic small round cell tumor, 1 neoplasm with FUS-CREM gene fusion) and 7 (23%) were SWI/SNF (SMARCB1 or SMARCA4)-deficient neoplasms. The remaining MN-NE included synovial sarcoma, sclerosing epithelioid mesenchymal neoplasm, melanoma, alveolar soft part sarcoma, solitary fibrous tumor, and chordoma. A total of 27/31 MN-NE were from the last 8 years, and 6 of them were located in the pancreas. Eleven MN-NE were initially diagnosed as neuroendocrine carcinomas (NECs). MN-NE with epithelioid features play an increasing role as mimickers of NECs. They mostly belong to tumors with gene fusions involving the EWSR1 gene, or with SWI/SNF complex deficiency. Synaptophysin expression is mostly patchy and chromograninA expression is infrequent in MN-NE of this series and data extracted from literature.


Subject(s)
Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Carcinoma, Neuroendocrine/genetics , DNA Helicases/deficiency , Gene Fusion , Neoplasms, Connective Tissue/genetics , Nuclear Proteins/deficiency , RNA-Binding Protein EWS/genetics , SMARCB1 Protein/deficiency , Transcription Factors/deficiency , Carcinoma, Neuroendocrine/chemistry , Carcinoma, Neuroendocrine/pathology , Chromogranin A/analysis , Cyclic AMP Response Element Modulator/genetics , Decision Support Techniques , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Neoplasms, Connective Tissue/chemistry , Neoplasms, Connective Tissue/pathology , Predictive Value of Tests , RNA-Binding Protein FUS/genetics , Synaptophysin/analysis
15.
Cells ; 10(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34440689

ABSTRACT

Lung cancers are ranked third among the cancer incidence in France in the year 2020, with adenocarcinomas being the commonest sub-type out of ~85% of non-small cell lung carcinomas. The constant evolution of molecular genotyping, which is used for the management of lung adenocarcinomas, has led to the current focus on tumor suppressor genes, specifically the loss of function mutation in the SMARCA4 gene. SMARCA4-deficient adenocarcinomas are preponderant in younger aged male smokers with a predominant solid morphology. The importance of identifying SMARCA4-deficient adenocarcinomas has gained interest for lung cancer management due to its aggressive behavior at diagnosis with vascular invasion and metastasis to the pleura seen upon presentation in most cases. These patients have poor clinical outcome with short overall survival rates, regardless of the stage of disease. The detection of SMARCA4 deficiency is possible in most pathology labs with the advent of sensitive and specific immunohistochemical antibodies. The gene mutations can be detected together with other established lung cancer molecular markers based on the current next generation sequencing panels. Sequencing will also allow the identification of associated gene mutations, notably KRAS, KEAP1, and STK11, which have an impact on the overall survival and progression-free survival of the patients. Predictive data on the treatment with anti-PD-L1 are currently uncertain in this high tumor mutational burden cancer, which warrants more groundwork. Identification of target drugs is also still in pre-clinical testing. Thus, it is paramount to identify the SMARCA4-deficient adenocarcinoma, as it carries worse repercussions on patient survival, despite having an exceptionally low prevalence. Herein, we discuss the pathophysiology of SMARCA4, the clinicopathological consequences, and different detection methods, highlighting the perspectives and challenges in the assessment of SMARCA4 deficiency for the management of non-small cell lung cancer patients. This is imperative, as the contemporary shift on identifying biomarkers associated with tumor suppressor genes such as SMARCA4 are trending; hence, awareness of pathologists and clinicians is needed for the SMARCA4-dNSCLC entity with close follow-up on new management strategies to overcome the poor possibilities of survival in such patients.


Subject(s)
Adenocarcinoma of Lung/metabolism , DNA Helicases/deficiency , DNA Helicases/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Adenocarcinoma of Lung/genetics , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , DNA Helicases/analysis , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mutation , Nuclear Proteins/analysis , Sequence Analysis, DNA , Transcription Factors/analysis
16.
Nat Commun ; 12(1): 4319, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34262032

ABSTRACT

Despite the genetic inactivation of SMARCA4, a core component of the SWI/SNF-complex commonly found in cancer, there are no therapies that effectively target SMARCA4-deficient tumours. Here, we show that, unlike the cells with activated MYC oncogene, cells with SMARCA4 inactivation are refractory to the histone deacetylase inhibitor, SAHA, leading to the aberrant accumulation of H3K27me3. SMARCA4-mutant cells also show an impaired transactivation and significantly reduced levels of the histone demethylases KDM6A/UTX and KDM6B/JMJD3, and a strong dependency on these histone demethylases, so that its inhibition compromises cell viability. Administering the KDM6 inhibitor GSK-J4 to mice orthotopically implanted with SMARCA4-mutant lung cancer cells or primary small cell carcinoma of the ovary, hypercalcaemic type (SCCOHT), had strong anti-tumour effects. In this work we highlight the vulnerability of KDM6 inhibitors as a characteristic that could be exploited for treating SMARCA4-mutant cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , DNA Helicases/deficiency , Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Neoplasms/drug therapy , Nuclear Proteins/deficiency , Transcription Factors/deficiency , Animals , Antineoplastic Agents/pharmacology , Benzazepines/pharmacology , Benzazepines/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , DNA Helicases/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Neoplasms/metabolism , Nuclear Proteins/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Transcription Factors/metabolism , Transcriptional Activation
17.
Nat Commun ; 12(1): 3520, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112784

ABSTRACT

The Immunodeficiency Centromeric Instability Facial Anomalies (ICF) 4 syndrome is caused by mutations in LSH/HELLS, a chromatin remodeler promoting incorporation of histone variant macroH2A. Here, we demonstrate that LSH depletion results in degradation of nascent DNA at stalled replication forks and the generation of genomic instability. The protection of stalled forks is mediated by macroH2A, whose knockdown mimics LSH depletion and whose overexpression rescues nascent DNA degradation. LSH or macroH2A deficiency leads to an impairment of RAD51 loading, a factor that prevents MRE11 and EXO1 mediated nascent DNA degradation. The defect in RAD51 loading is linked to a disbalance of BRCA1 and 53BP1 accumulation at stalled forks. This is associated with perturbed histone modifications, including abnormal H4K20 methylation that is critical for BRCA1 enrichment and 53BP1 exclusion. Altogether, our results illuminate the mechanism underlying a human syndrome and reveal a critical role of LSH mediated chromatin remodeling in genomic stability.


Subject(s)
DNA Helicases/metabolism , DNA Replication , Genomic Instability , Histones/metabolism , Rad51 Recombinase/metabolism , Animals , BRCA1 Protein/metabolism , Cell Line, Tumor , Cell Survival/genetics , Chromatin Assembly and Disassembly/genetics , Chromatin Immunoprecipitation Sequencing , DNA Helicases/deficiency , DNA Helicases/genetics , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA Replication/genetics , Epigenesis, Genetic , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Genomic Instability/genetics , Histones/deficiency , Histones/genetics , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Methylation , Mice , RNA, Small Interfering , Rad51 Recombinase/genetics , Up-Regulation
18.
Immunotherapy ; 13(10): 799-806, 2021 07.
Article in English | MEDLINE | ID: mdl-34030451

ABSTRACT

SMARCA4-deficient thoracic sarcoma (DTS) is a recently noted progressive thoracic malignancy. We recently experienced three cases of SMARCA4-DTS who were treated with atezolizumab in combination with bevacizumab, paclitaxel and carboplatin (ABCP) as the first-line therapy. Immunohistopathological analysis revealed absent expression of SMARCA4 in all cases. The tumor mutational burden was over 11/Mb and mutations in SMARCA4 and TP53 were detected in all three cases. Partial response to ABCP treatment was observed in all three cases, with a progression-free survival of approximately 6 months or longer and a continuous response of 1 year or longer in one case. The first-line ABCP treatment demonstrated durable efficacy in SMARCA4-DTS regardless of the degree of PD-L1 expression.


Lay abstract Lung cancer is the leading cause of cancer-related death worldwide. Among them, SMARCA4-deficient thoracic sarcoma (DTS), which lacks SMARCA4 expression and exhibits an undifferentiated carcinoma histology, is a recently identified subtype of lung cancer. It tends to occur in younger people with heavy smoking status and has been reported to recur quickly and have a poor prognosis even after chemotherapy, radiation therapy or surgery. There is no effective molecularly targeted agent for SMARCA4-DTS and the identification of an effective therapy is required. Here, we report the clinical features and genomic information of three SMARCA4-DTS cases in which atezolizumab with bevacizumab, paclitaxel and carboplatin treatment was effective. This report suggests the efficacy of atezolizumab with bevacizumab, paclitaxel and carboplatin treatment compared with conventional chemotherapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab/therapeutic use , Carboplatin/therapeutic use , DNA Helicases/deficiency , Lung Neoplasms/drug therapy , Nuclear Proteins/deficiency , Paclitaxel/therapeutic use , Sarcoma/drug therapy , Transcription Factors/deficiency , Aged , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Drug Therapy, Combination/methods , Female , Humans , Lung Neoplasms/immunology , Male , Middle Aged , Sarcoma/immunology , Treatment Outcome
19.
J Immunol ; 206(10): 2453-2467, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33941659

ABSTRACT

The detection of intracellular nucleic acids is a fundamental mechanism of host defense against infections. The dysregulated nucleic acid sensing, however, is a major cause for a number of autoimmune diseases. In this study, we report that GTPase-activating protein SH3 domain-binding protein 1 (G3BP1) is critical for both intracellular DNA- and RNA-induced immune responses. We found that in both human and mouse cells, the deletion of G3BP1 led to the dampened cGAS activation by DNA and the insufficient binding of RNA by RIG-I. We further found that resveratrol (RSVL), a natural compound found in grape skin, suppressed both intracellular DNA- and RNA-induced type I IFN production through inhibiting G3BP1. Importantly, using experimental mouse models for Aicardi-Goutières syndrome, an autoimmune disorder found in humans, we demonstrated that RSVL effectively alleviated intracellular nucleic acid-stimulated autoimmune responses. Thus, our study demonstrated a broader role of G3BP1 in sensing different kinds of intracellular nucleic acids and presented RSVL as a potential treatment for autoimmune conditions caused by dysregulated nucleic acid sensing.


Subject(s)
Autoimmunity/genetics , DNA Helicases/deficiency , DNA Helicases/metabolism , Intracellular Space/metabolism , Nucleic Acids/metabolism , Poly-ADP-Ribose Binding Proteins/deficiency , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/deficiency , RNA Helicases/metabolism , RNA Recognition Motif Proteins/deficiency , RNA Recognition Motif Proteins/metabolism , Signal Transduction/genetics , A549 Cells , Animals , Autoimmunity/drug effects , Cell Survival/drug effects , DNA Helicases/antagonists & inhibitors , DNA Helicases/genetics , Fibroblasts/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Intracellular Space/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/antagonists & inhibitors , RNA Helicases/genetics , RNA Recognition Motif Proteins/antagonists & inhibitors , RNA Recognition Motif Proteins/genetics , Resveratrol/administration & dosage , Signal Transduction/immunology , Transfection
20.
PLoS Genet ; 17(5): e1008919, 2021 05.
Article in English | MEDLINE | ID: mdl-34003859

ABSTRACT

An essential component of the homologous recombination machinery in eukaryotes, the RAD54 protein is a member of the SWI2/SNF2 family of helicases with dsDNA-dependent ATPase, DNA translocase, DNA supercoiling and chromatin remodelling activities. It is a motor protein that translocates along dsDNA and performs multiple functions in homologous recombination. In particular, RAD54 is an essential cofactor for regulating RAD51 activity. It stabilizes the RAD51 nucleofilament, remodels nucleosomes, and stimulates the homology search and strand invasion activities of RAD51. Accordingly, deletion of RAD54 has dramatic consequences on DNA damage repair in mitotic cells. In contrast, its role in meiotic recombination is less clear. RAD54 is essential for meiotic recombination in Drosophila and C. elegans, but plays minor roles in yeast and mammals. We present here characterization of the roles of RAD54 in meiotic recombination in the model plant Arabidopsis thaliana. Absence of RAD54 has no detectable effect on meiotic recombination in otherwise wild-type plants but RAD54 becomes essential for meiotic DSB repair in absence of DMC1. In Arabidopsis, dmc1 mutants have an achiasmate meiosis, in which RAD51 repairs meiotic DSBs. Lack of RAD54 leads to meiotic chromosomal fragmentation in absence of DMC1. The action of RAD54 in meiotic RAD51 activity is thus mainly downstream of the role of RAD51 in supporting the activity of DMC1. Equivalent analyses show no effect on meiosis of combining dmc1 with the mutants of the RAD51-mediators RAD51B, RAD51D and XRCC2. RAD54 is thus required for repair of meiotic DSBs by RAD51 and the absence of meiotic phenotype in rad54 plants is a consequence of RAD51 playing a RAD54-independent supporting role to DMC1 in meiotic recombination.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/cytology , Arabidopsis/metabolism , DNA Breaks, Double-Stranded , DNA Helicases/metabolism , Meiosis , Rad51 Recombinase/metabolism , Recombinational DNA Repair , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Helicases/deficiency , DNA Helicases/genetics , DNA-Binding Proteins , Genes, Essential , Meiosis/genetics , Mutation , Rad51 Recombinase/genetics , Rec A Recombinases/genetics , Rec A Recombinases/metabolism , Repressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...