Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 650
Filter
1.
Pathol Oncol Res ; 30: 1611730, 2024.
Article in English | MEDLINE | ID: mdl-39165647

ABSTRACT

Introduction: Mesenchymal chondrosarcoma (MCS) is a rare subtype of chondrosarcoma that occurs at widespread anatomical locations, such as bone, soft tissue, and intracranial sites. The central nervous system (CNS) is one of the most common origins of extraosseous MCS. However, alternative HEY1::NCOA2 fusions have not been reported in this tumor. Case report: We report a case of intracranial MCS with HEY1::NCOA2 rearrangement. A 52-year-old woman presented with a 15-mm calcified mass around the sella turcica. She initially underwent transsphenoidal surgery for tumor resection and then additional resections for five local recurrences over 5 years. Histologically, the tumor was composed of small round to spindle-shaped cells admixed with well-differentiated hyaline cartilaginous islands. A hemangiopericytoma-like vascular pattern and small sinusoid-like vessels were also observed. RNA sequencing using RNA extracted from formalin-fixed paraffin-embedded samples from the last operation revealed two alternative variants of the HEY1::NCOA2 fusion: HEY1(ex4)::NCOA2 (ex13) and HEY1(ex4)::NCOA2(ex14). Both variants were confirmed as in-frame fusions using reverse transcription-polymerase chain reaction. Discussion: Cartilaginous components were often not apparent during the recurrences. In addition to the non-typical pathological finding, the correct diagnosis was hampered by the poor RNA quality of the surgical specimens and non-specific STAT6 nuclear staining. Conclusion: This is the first reported case of intracranial MCS with an alternative HEY1::NCOA2 fusion.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cell Cycle Proteins , Chondrosarcoma, Mesenchymal , Nuclear Receptor Coactivator 2 , Sella Turcica , Humans , Female , Middle Aged , Chondrosarcoma, Mesenchymal/genetics , Chondrosarcoma, Mesenchymal/pathology , Chondrosarcoma, Mesenchymal/surgery , Basic Helix-Loop-Helix Transcription Factors/genetics , Sella Turcica/pathology , Nuclear Receptor Coactivator 2/genetics , Cell Cycle Proteins/genetics
2.
Medicine (Baltimore) ; 103(33): e39123, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39151492

ABSTRACT

RATIONALE: Uterine tumors resembling ovarian sex cord tumors (UTROSCT) with rhabdoid features are uncommon mesenchymal neoplasms exhibiting diverse histological patterns, including significant rhabdoid morphology. A thorough comprehension of their clinicopathologic features is crucial for precise diagnosis and effective management. PATIENT CONCERNS: This study presents 4 cases of UTROSCT with rhabdoid features, diagnosed in patients aged 31 to 58. Varied recurrence patterns were observed, including similar recurrent lesions to the primary tumors with subsequent mortality, initial invasion and lymph node metastasis, and presence of only primary tumor. DIAGNOSES: Histopathological examination revealed diverse morphological patterns, prominently featuring rhabdoid differentiation. Immunohistochemical analysis showed expression of hormone receptors, sex cord, smooth muscle, and epithelial markers, notably WT1, CD56, and CD99. Molecular analysis identified ESR1-NCOA2 fusions and ESR1 and NCOA2/3 rearrangements, indicating a potential association between these genetic alterations and extensive rhabdoid differentiation. INTERVENTIONS: Various treatments were administered post-recurrence, including chemotherapy and targeted therapies. However, poor clinical outcomes were observed in all cases. OUTCOMES: Despite aggressive treatments, including chemotherapy and targeted therapies, poor clinical outcomes were observed, highlighting the aggressive nature of UTROSCT with significant rhabdoid differentiation. LESSONS: This case series emphasizes the importance of detailed pathological reporting, comprehensive molecular testing, and thorough tumor staging in UTROSCT cases with rhabdoid features. Enhanced understanding of the clinicopathologic characteristics of UTROSCT with rhabdoid differentiation is crucial for accurate diagnosis, prognostication, and management strategies.


Subject(s)
Ovarian Neoplasms , Sex Cord-Gonadal Stromal Tumors , Uterine Neoplasms , Humans , Female , Adult , Sex Cord-Gonadal Stromal Tumors/pathology , Sex Cord-Gonadal Stromal Tumors/genetics , Sex Cord-Gonadal Stromal Tumors/diagnosis , Middle Aged , Uterine Neoplasms/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/diagnosis , Diagnosis, Differential , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Rhabdoid Tumor/genetics , Rhabdoid Tumor/diagnosis , Rhabdoid Tumor/pathology , Nuclear Receptor Coactivator 2/genetics , CD56 Antigen/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Biomarkers, Tumor/genetics , 12E7 Antigen/genetics , 12E7 Antigen/metabolism , WT1 Proteins/genetics
3.
Proc Natl Acad Sci U S A ; 121(26): e2405905121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38889153

ABSTRACT

Aberrant regulation of chromatin modifiers is a common occurrence across many cancer types, and a key priority is to determine how specific alterations of these proteins, often enzymes, can be targeted therapeutically. MOZ, a histone acyltransferase, is recurrently fused to coactivators CBP, p300, and TIF2 in cases of acute myeloid leukemia (AML). Using either pharmacological inhibition or targeted protein degradation in a mouse model for MOZ-TIF2-driven leukemia, we show that KAT6 (MOZ/MORF) enzymatic activity and the MOZ-TIF2 protein are necessary for indefinite proliferation in cell culture. MOZ-TIF2 directly regulates a small subset of genes encoding developmental transcription factors, augmenting their high expression. Furthermore, transcription levels in MOZ-TIF2 cells positively correlate with enrichment of histone H3 propionylation at lysine 23 (H3K23pr), a recently appreciated histone acylation associated with gene activation. Unexpectedly, we also show that MOZ-TIF2 and MLL-AF9 regulate transcription of unique gene sets, and their cellular models exhibit distinct sensitivities to multiple small-molecule inhibitors directed against AML pathways. This is despite the shared genetic pathways of wild-type MOZ and MLL. Overall, our data provide insight into how aberrant regulation of MOZ contributes to leukemogenesis. We anticipate that these experiments will inform future work identifying targeted therapies in the treatment of AML and other diseases involving MOZ-induced transcriptional dysregulation.


Subject(s)
Histone Acetyltransferases , Histones , Animals , Mice , Histones/metabolism , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Humans , Disease Models, Animal , Nuclear Receptor Coactivator 2/metabolism , Nuclear Receptor Coactivator 2/genetics , Gene Expression Regulation, Leukemic , Gene Expression Regulation, Developmental , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/genetics
4.
Genes Chromosomes Cancer ; 63(5): e23244, 2024 05.
Article in English | MEDLINE | ID: mdl-38747338

ABSTRACT

We describe a case of a pleomorphic adenoma (PA) arising from the para-tracheal accessory salivary gland in a 44-year-old male harboring a novel WWTR1::NCOA2 gene fusion. To our knowledge, this novel gene fusion has not been described previously in salivary gland tumors. The patient presented with hoarseness of voice. The radiological exam revealed a mass in the upper third of the trachea involving the larynx. Histologically, the tumor consisted of bland-looking monocellular eosinophilic epithelial cells arranged in cords and sheets separated by thin fibrous stroma, focally forming a pseudo-tubular pattern. In immunohistochemistry, the tumor cells demonstrated positivity for CK7, PS100, SOX10, and HMGA2; and negativity for CK5/6, p40 p63, and PLAG1. In addition, the clustering analysis clearly demonstrates a clustering of tumors within the PA group. In addition to reporting this novel fusion in the PA spectrum, we discuss the relevant differential diagnoses and briefly review of NCOA2 and WWTR1 gene functions in normal and neoplastic contexts.


Subject(s)
HMGA2 Protein , Nuclear Receptor Coactivator 2 , Trans-Activators , Humans , Male , Nuclear Receptor Coactivator 2/genetics , Nuclear Receptor Coactivator 2/metabolism , Adult , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Trans-Activators/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Oncogene Proteins, Fusion/genetics , Myoepithelioma/genetics , Myoepithelioma/pathology , Myoepithelioma/metabolism
5.
Diagn Pathol ; 19(1): 65, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678288

ABSTRACT

BACKGROUND: MEIS1::NCOA2 is a rare fusion gene that has been recently described in a subset of spindle cell rhabdomyosarcomas and multiple low-grade undifferentiated spindle cell sarcomas predominantly arising in the genitourinary and gynecologic tracts with no specific line of differentiation. We present the first documented case of this neoplasm arising as a lung primary tumor. CASE PRESENTATION: A 74-year-old woman with a 40-year smoking history presented with a 2.1 × 1.7 cm lung nodule discovered on computed tomography (CT) scan. A biopsy and subsequent lobe resection were performed, as well as an extensive metastatic work up, which revealed no additional masses. No specific line of differentiation was found by immunohistochemical staining, and an RNA-based fusion panel revealed a MEIS1::NCOA2 fusion, at which point a diagnosis of Low-Grade Undifferentiated Sarcoma with MEIS1::NCOA2-Rearrangement was rendered. CONCLUSIONS: This report represents the first diagnosis of this tumor primary to the lung, and provides additional insight into the origin and localization of these rare tumors.


Subject(s)
Lung Neoplasms , Myeloid Ecotropic Viral Integration Site 1 Protein , Nuclear Receptor Coactivator 2 , Sarcoma , Humans , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Female , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Aged , Sarcoma/genetics , Sarcoma/pathology , Nuclear Receptor Coactivator 2/genetics , Gene Rearrangement , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis
6.
Nat Metab ; 6(5): 825-836, 2024 May.
Article in English | MEDLINE | ID: mdl-38622413

ABSTRACT

Nuclear receptor corepressors (NCoRs) function in multiprotein complexes containing histone deacetylase 3 (HDAC3) to alter transcriptional output primarily through repressive chromatin remodelling at target loci1-5. In the liver, loss of HDAC3 causes a marked hepatosteatosis largely because of de-repression of genes involved in lipid metabolism6,7; however, the individual roles and contribution of other complex members to hepatic and systemic metabolic regulation are unclear. Here we show that adult loss of both NCoR1 and NCoR2 (double knockout (KO)) in hepatocytes phenocopied the hepatomegalic fatty liver phenotype of HDAC3 KO. In addition, double KO livers exhibited a dramatic reduction in glycogen storage and gluconeogenic gene expression that was not observed with hepatic KO of individual NCoRs or HDAC3, resulting in profound fasting hypoglycaemia. This surprising HDAC3-independent activation function of NCoR1 and NCoR2 is due to an unexpected loss of chromatin accessibility on deletion of NCoRs that prevented glucocorticoid receptor binding and stimulatory effect on gluconeogenic genes. These studies reveal an unanticipated, non-canonical activation function of NCoRs that is required for metabolic health.


Subject(s)
Gluconeogenesis , Histone Deacetylases , Liver , Mice, Knockout , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Receptors, Glucocorticoid , Gluconeogenesis/genetics , Animals , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 1/genetics , Mice , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Liver/metabolism , Hepatocytes/metabolism , Nuclear Receptor Coactivator 2/metabolism , Nuclear Receptor Coactivator 2/genetics
8.
Cancer Genet ; 284-285: 5-11, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38471404

ABSTRACT

Systemic mastocytosis (SM) is a myeloproliferative neoplasm displaying abnormal mast cell proliferation. It is subdivided into different forms, including aggressive systemic mastocytosis (ASM) and systemic mastocytosis with an associated hematologic neoplasm (SM-AHN). Oncogenic genetic alterations include point mutations, mainly the KIT D816V, conferring poor prognosis and therapy resistance, and fusion genes, with those involving PDGFRA/PDGFRB as the most recurrent events. We here describe an ASM case negative to the KIT D816V and JAK2 V617F alterations but showing a RUNX1 frameshift heterozygous mutation and the co-occurrence of three fusion transcripts. The first one, PRKG2::PDGFRB, was generated by a balanced t(4;5)(q24;q32) translocation as the sole abnormality. Other two novel chimeras, KAT6A::NCOA2 and RXRA::NOTCH1, originated from cryptic intra-chromosomal abnormalities. The patient rapidly evolved towards SM-AHN, characterized by the persistence of the PRKG2::PDGFRB chimera, due to the presence of an extra copy of the der(5)t(4;5)(q24;q34) chromosome and an increase in the RUNX1 mutation allelic frequency. The results indicated that the transcriptional landscape and the mutational profile of SM deserve attention to predict the evolution and prognosis of this complex disease, whose classification criteria are still a matter of debate.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Frameshift Mutation , Mastocytosis, Systemic , Oncogene Proteins, Fusion , Humans , Core Binding Factor Alpha 2 Subunit/genetics , Mastocytosis, Systemic/genetics , Oncogene Proteins, Fusion/genetics , Receptor, Notch1/genetics , Nuclear Receptor Coactivator 2/genetics , Male , Heterozygote , Female , Middle Aged , Histone Acetyltransferases
11.
Genes Chromosomes Cancer ; 63(1): e23203, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37724942

ABSTRACT

Spindle cell mesenchymal neoplasms are a diverse and often challenging diagnostic group. While morphological impression is sufficient for some diagnoses, increasingly immunohistochemical and even molecular data is required to render an accurate diagnosis, which can lead to the characterization of new entities. We describe five cases of novel mesenchymal neoplasms with rearrangements in the NCOA2 and NCOA3 genes partnered with either CTCF or CRTC1. Three tumors occurred in the head and neck (palate, auditory canal), while the other two were in visceral organs (lung, urinary bladder). All cases occurred in adults (range 33-86) with a median age of 42 and fairly even sex distribution = (male-to-female = 3:2). Morphologically, they had similar features consisting of monotonous, bland spindle to ovoid cells with fascicular and reticular arrangements in a myxohyaline to collagenous stroma. However, immunophenotypically they had essentially a null phenotype, with only two tumors staining partially for CD34 and smooth muscle actin. Targeted RNA sequencing detected in-frame CTCF::NCOA2 (one case), CRTC1::NCOA2 (two cases), and CTCF::NCOA3 (two cases) fusions. Treatment was surgical resection in all cases. Local recurrence and/or distant metastases were not observed in any case (median follow-up, 7.5 months; range, 2-19 months). Given their morphologic, immunohistochemical, and molecular similarities, we believe that these cases may represent an emerging family of low-grade NCOA2/3-rearranged fibroblastic spindle cell neoplasms.


Subject(s)
Neoplasms, Connective and Soft Tissue , Soft Tissue Neoplasms , Adult , Humans , Male , Female , Fibroblasts/pathology , Base Sequence , Neoplasms, Connective and Soft Tissue/genetics , Phenotype , Biomarkers, Tumor/genetics , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/pathology , Nuclear Receptor Coactivator 2/genetics
12.
Genes Chromosomes Cancer ; 63(1): e23209, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37870842

ABSTRACT

Smooth muscle tumors are the most common mesenchymal tumors of the female genital tract, including the vulva. Since vulvar smooth muscle tumors are rare, our understanding of them compared to their uterine counterparts continues to evolve. Herein, we present two cases of morphologically distinct myxoid epithelioid smooth muscle tumors of the vulva with novel MEF2D::NCOA2 gene fusion. The tumors involved 24 and 37-year-old women. Both tumors presented as palpable vulvar masses that were circumscribed, measuring 2.8 and 5.1 cm in greatest dimension. Histologically, they were composed of epithelioid to spindle-shaped cells with minimal cytologic atypia and prominent myxoid matrix. Rare mitotic figures were present (1-3 mitotic figures per 10 high-power field (HPF)), and no areas of tumor necrosis were identified. By immunohistochemistry, the neoplastic cells strongly expressed smooth muscle actin, calponin, and desmin, confirming smooth muscle origin. Next-generation sequencing identified identical MEF2D::NCOA2 gene fusions. These two cases demonstrate that at least a subset of myxoid epithelioid smooth muscle tumors of the vulva represent a distinct entity characterized by a novel MEF2D::NCOA2 gene fusion. Importantly, recognition of the distinct morphologic and genetic features of these tumors is key to understanding the biological potential of these rare tumors.


Subject(s)
Smooth Muscle Tumor , Adult , Female , Humans , Young Adult , Biomarkers, Tumor/genetics , Gene Fusion , MEF2 Transcription Factors/genetics , Nuclear Receptor Coactivator 2/genetics , Smooth Muscle Tumor/pathology , Vulva/pathology
13.
Int J Gynecol Pathol ; 43(1): 47-55, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37043646

ABSTRACT

MEIS1::NCOA1/2 fusion sarcomas are a recently described novel entity arising in a variety of locations with a predilection for the genitourinary tract and gynecologic organs. Despite multiple locoregional recurrences, these tumors are thought to behave in a low-grade malignant manner. Here we report a uterine MEIS1::NCOA2 fusion sarcoma with lung metastasis. The patient was a 47-yr-old woman with a history of abnormal uterine bleeding who was found to have a myometrial mass confirmed by pathology to be uterine sarcoma. The tumor was predominantly composed of monotonous spindle cells with scant cytoplasm, crowded nuclei, and brisk mitotic activity, growing in a fascicular and streaming pattern. The morphologic and immunophenotypic features were nonspecific and a diagnosis of high-grade uterine sarcoma with a differential of leiomyosarcoma versus high-grade endometrial stromal sarcoma was rendered. At the 27-mo follow-up, the patient was found to have a lung metastasis consisting of a monotonous round cell sarcoma. A retrospective RNA-based and DNA-based next-generation sequencing of the primary uterine sarcoma revealed a MEIS1::NCOA2 gene fusion, a c.94G>C/p.D32H mutation in exon 3 of CTNNB1 gene, HMGA2 , and CDK4 gene amplification, and an intermediate/marginal level of MDM2 gene amplification. Polymerase chain reaction-based molecular analysis further demonstrated that the MEIS1::NCOA2 gene fusion and CTNNB1 somatic mutation were also present in the lung metastasis. This case represents the first case of such gynecologic sarcoma with distant (lung) metastasis, and the second metastatic case among all reported MEIS1::NCOA1/2 fusion sarcomas, highlighting the malignant metastatic potential of this emerging entity. Our case also indicates that HMGA2/CDK4/MDM2 region amplification and CTNNB1 somatic mutation might be recurrent genetic events in this rare sarcoma subtype.


Subject(s)
Endometrial Neoplasms , Lung Neoplasms , Sarcoma, Endometrial Stromal , Sarcoma , Uterine Neoplasms , Humans , Female , Retrospective Studies , Neoplasm Recurrence, Local , Sarcoma/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Sarcoma, Endometrial Stromal/pathology , Endometrial Neoplasms/pathology , Lung Neoplasms/genetics , Biomarkers, Tumor/analysis , Nuclear Receptor Coactivator 2/genetics
14.
Cytopathology ; 35(1): 149-152, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37688410

ABSTRACT

Mesenchymal chondrosarcoma (MC) is a rare but extremely aggressive type of chondrosarcoma distinguished by the presence of both primitive mesenchymal cells and fully developed chondroid tissue. The identification of a biphasic morphology in pleural effusion, along with detection of the HEY1::NCOA2 fusion using next-generation sequencing, serve as vital indicators for an accurate diagnosis.


Subject(s)
Bone Neoplasms , Chondrosarcoma, Mesenchymal , Pleural Effusion , Humans , Chondrosarcoma, Mesenchymal/diagnosis , Chondrosarcoma, Mesenchymal/genetics , Chondrosarcoma, Mesenchymal/metabolism , Immunohistochemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins , High-Throughput Nucleotide Sequencing , Nuclear Receptor Coactivator 2/metabolism
15.
FASEB J ; 37(12): e23313, 2023 12.
Article in English | MEDLINE | ID: mdl-37962238

ABSTRACT

Although we have shown that steroid receptor coactivator-2 (SRC-2), a member of the p160/SRC family of transcriptional coregulators, is essential for decidualization of both human and murine endometrial stromal cells, SRC-2's role in the earlier stages of the implantation process have not been adequately addressed. Using a conditional SRC-2 knockout mouse (SRC-2d/d ) in timed natural pregnancy studies, we show that endometrial SRC-2 is required for embryo attachment and adherence to the luminal epithelium. Implantation failure is associated with the persistent expression of Mucin 1 and E-cadherin on the apical surface and basolateral adherens junctions of the SRC-2d/d luminal epithelium, respectively. These findings indicate that the SRC-2d/d luminal epithelium fails to exhibit a plasma membrane transformation (PMT) state known to be required for the development of uterine receptivity. Transcriptomics demonstrated that the expression of genes involved in steroid hormone control of uterine receptivity were significantly disrupted in the SRC-2d/d endometrium as well as genes that control epithelial tight junctional biology and the emergence of the epithelial mesenchymal transition state, with the latter sharing similar biological properties with PMT. Collectively, these findings uncover a new role for endometrial SRC-2 in the induction of the luminal epithelial PMT state, which is a prerequisite for the development of uterine receptivity and early pregnancy establishment.


Subject(s)
Embryo Implantation , Uterus , Animals , Female , Humans , Mice , Pregnancy , Embryo Implantation/genetics , Endometrium/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Mice, Knockout , Nuclear Receptor Coactivator 2/genetics , Uterus/metabolism
16.
Cancer Invest ; 41(8): 704-712, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37668330

ABSTRACT

Spindle cell/sclerosing rhabdomyosarcoma (SSRMS) is a clinicopathologically and molecularly heterogeneous disease. Gene fusions have been identified in intraosseous SSRMS, consisting predominantly of EWSR1/FUS::TFCP2 and MEIS1::NCOA2. The former often follow an aggressive clinical course; there is limited clinical follow-up available for the latter. We report here a new case of the very rare intraosseous SSRMS with MEIS1::NCOA2 gene fusion and include the detailed treatment course and 52 months of clinical follow-up. SSRMS with MEIS1::NCOA2 gene fusion appears biologically distinct from other intraosseous SSRMS, following a course characterized by local recurrence with rare reports of metastasis to date.


Subject(s)
Rhabdomyosarcoma , Transcription Factors , Adult , Humans , Child , Follow-Up Studies , Transcription Factors/genetics , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/therapy , Rhabdomyosarcoma/pathology , Nuclear Receptor Coactivator 2/genetics , DNA-Binding Proteins/genetics
17.
Cancer Immunol Res ; 11(10): 1414-1431, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37540802

ABSTRACT

Nuclear receptor coactivator 2 (Ncoa2) is a member of the Ncoa family of coactivators, and we previously showed that Ncoa2 regulates the differentiation of induced regulatory T cells. However, it remains unknown if Ncoa2 plays a role in CD8+ T-cell function. Here, we show that Ncoa2 promotes CD8+ T cell-mediated immune responses against tumors by stimulating T-cell activation via upregulating PGC-1α expression to enhance mitochondrial function. Mice deficient in Ncoa2 in T cells (Ncoa2fl/fl/CD4Cre) displayed defective immune responses against implanted MC38 tumors, which associated with significantly reduced tumor-infiltrating CD8+ T cells and decreased IFNγ production. Consistently, CD8+ T cells from Ncoa2fl/fl/CD4Cre mice failed to reject tumors after adoptive transfer into Rag1-/- mice. Further, in response to TCR stimulation, Ncoa2fl/fl/CD4Cre CD8+ T cells failed to increase mitochondrial mass, showed impaired oxidative phosphorylation, and had lower expression of PGC-1α, a master regulator of mitochondrial biogenesis and function. Mechanically, T-cell activation-induced phosphorylation of CREB triggered the recruitment of Ncoa2 to bind to enhancers, thus, stimulating PGC-1α expression. Forced expression of PGC-1α in Ncoa2fl/fl/CD4Cre CD8+ T cells restored mitochondrial function, T-cell activation, IFNγ production, and antitumor immunity. This work informs the development of Ncoa2-based therapies that modulate CD8+ T cell-mediated antitumor immune responses.


Subject(s)
Mitochondria , Neoplasms , Animals , Mice , CD8-Positive T-Lymphocytes/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Nuclear Receptor Coactivator 2/metabolism , Up-Regulation
18.
Proc Natl Acad Sci U S A ; 120(18): e2221352120, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37094160

ABSTRACT

T cell activation stimulates substantially increased protein synthesis activity to accumulate sufficient biomass for cell proliferation. The protein synthesis is fueled by the amino acids transported from the environment. Steroid nuclear receptor coactivator 2 (SRC2) is a member of a family of transcription coactivators. Here, we show that SRC2 recruited by c-Myc enhances CD4+ T cell activation to stimulate immune responses via upregulation of amino acid transporter Slc7a5. Mice deficient of SRC2 in T cells (SRC2fl/fl/CD4Cre) are resistant to the induction of experimental autoimmune encephalomyelitis (EAE) and susceptible to Citrobacter rodentium (C. rodentium) infection. Adoptive transfer of naive CD4+ T cells from SRC2fl/fl/CD4Cre mice fails to elicit EAE and colitis in Rag1/ recipients. Further, CD4+ T cells from SRC2fl/fl/CD4Cre mice display defective T cell proliferation, cytokine production, and differentiation both in vitro and in vivo. Mechanically, SRC2 functions as a coactivator to work together with c-Myc to stimulate the expression of amino acid transporter Slc7a5 required for T cell activation. Slc7a5 fails to be up-regulated in CD4+ T cells from SRC2fl/fl/CD4Cre mice, and forced expression of Slc7a5 rescues proliferation, cytokine production, and the ability of SRC2fl/fl/CD4Cre CD4+ T cells to induce EAE. Therefore, SRC2 is essential for CD4+ T cell activation and, thus, a potential drug target for controlling CD4+ T cell-mediated autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , T-Lymphocytes , Animals , Mice , CD4-Positive T-Lymphocytes , Cytokines/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Coactivator 2/metabolism , Up-Regulation
19.
Int J Gynecol Pathol ; 42(5): 460-465, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-36811835

ABSTRACT

MEIS1-NCOA1/2 fusions are recently described gene rearrangements found in rare sarcomas, mainly involving the genitourinary and gynecologic tracts, with 3 cases reported in the uterine corpus. Although local recurrence was very common, no death has been reported, and some investigators consider these sarcomas low grade. Amplification of genes located at the 12q13-15 locus, especially MDM2 , is the hallmark genetic abnormality in well-differentiated and dedifferentiated liposarcoma of the soft tissue. Some uterine tumors have also been reported to harbor MDM2 amplification, including a proportion of Müllerian adenosarcomas, BCOR fusion-positive high-grade endometrial stromal sarcoma, BCORL1 -altered high-grade endometrial stromal sarcoma, rare JAZF1 fusion-positive low-grade endometrial stromal sarcoma, rare undifferentiated uterine sarcoma, and a single case of MEIS1-NCOA2 fusion sarcoma. Here, we report a case of high-grade MEIS1-NCOA2 fusion uterine sarcoma which also harbored amplification of multiple 12q13-15 genes, including MDM2 , CDK4 , MDM4 , and FRS2 , that exhibited aggressive clinical course leading to patient's death within 2 yr of the initial diagnosis. To the best of our knowledge, this is the first documented case of fatal MEIS1-NCOA2 fusion uterine sarcoma, and the second case of MEIS1-NCOA2 fusion uterine sarcoma that also harbors MDM2 amplification.


Subject(s)
Endometrial Neoplasms , Sarcoma, Endometrial Stromal , Sarcoma , Humans , Female , Uterus , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Nuclear Receptor Coactivator 2 , Proto-Oncogene Proteins/genetics , Cell Cycle Proteins
20.
Cell Rep ; 42(1): 112013, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36656711

ABSTRACT

Clinical sequencing efforts are rapidly identifying sarcoma gene fusions that lack functional validation. An example is the fusion of transcriptional coactivators, VGLL2-NCOA2, found in infantile rhabdomyosarcoma. To delineate VGLL2-NCOA2 tumorigenic mechanisms and identify therapeutic vulnerabilities, we implement a cross-species comparative oncology approach with zebrafish, mouse allograft, and patient samples. We find that VGLL2-NCOA2 is sufficient to generate mesenchymal tumors that display features of immature skeletal muscle and recapitulate the human disease. A subset of VGLL2-NCOA2 zebrafish tumors transcriptionally cluster with embryonic somitogenesis and identify VGLL2-NCOA2 developmental programs, including a RAS family GTPase, ARF6. In VGLL2-NCOA2 zebrafish, mouse, and patient tumors, ARF6 is highly expressed. ARF6 knockout suppresses VGLL2-NCOA2 oncogenic activity in cell culture, and, more broadly, ARF6 is overexpressed in adult and pediatric sarcomas. Our data indicate that VGLL2-NCOA2 is an oncogene that leverages developmental programs for tumorigenesis and that reactivation or persistence of ARF6 could represent a therapeutic opportunity.


Subject(s)
Rhabdomyosarcoma , Sarcoma , Child , Adult , Humans , Animals , Mice , Zebrafish/metabolism , Transcription Factors/genetics , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Gene Fusion , Nuclear Receptor Coactivator 2/genetics , Muscle Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL