Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 550
Filter
1.
Cells ; 13(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38891067

ABSTRACT

Rapid information processing in the central nervous system requires the myelination of axons by oligodendrocytes. The transcription factor Sox2 and its close relative Sox3 redundantly regulate the development of myelin-forming oligodendrocytes, but little is known about the underlying molecular mechanisms. Here, we characterized the expression profile of cultured oligodendroglial cells during early differentiation and identified Bcas1, Enpp6, Zfp488 and Nkx2.2 as major downregulated genes upon Sox2 and Sox3 deletion. An analysis of mice with oligodendrocyte-specific deletion of Sox2 and Sox3 validated all four genes as downstream targets in vivo. Additional functional assays identified regulatory regions in the vicinity of each gene that are responsive to and bind both Sox proteins. Bcas1, Enpp6, Zfp488 and Nkx2.2 therefore likely represent direct target genes and major effectors of Sox2 and Sox3. Considering the preferential expression and role of these genes in premyelinating oligodendrocytes, our findings suggest that Sox2 and Sox3 impact oligodendroglial development at the premyelinating stage with Bcas1, Enpp6, Zfp488 and Nkx2.2 as their major effectors.


Subject(s)
Cell Differentiation , Homeobox Protein Nkx-2.2 , Oligodendroglia , SOXB1 Transcription Factors , Transcription Factors , Animals , Mice , Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Oligodendroglia/metabolism , Oligodendroglia/cytology , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
2.
Diagn Pathol ; 19(1): 76, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851744

ABSTRACT

BACKGROUND: CIC-rearranged sarcomas (CRS) represent a new entity of undifferentiated small round cell sarcoma belonging to the Ewing-like sarcomas family. CRS are the most common type. Fusion partners for the CIC gene include DUX4, FOXO4, and the recently recognizedNUTM1. Rare cases of CIC::NUTM1 sarcoma in pediatric patients have recently been reported in brain, kidney, bone, and soft tissues. However, such cases have not been identified in the soft tissues of the limbs. CASE PRESENTATION: We reported a case of CIC::NUTM1 sarcoma located in the right upper limb of an 18-year-old man. The tumor displayed morphologic features typical of CIC::DUX4 sarcomas, with small- to medium-sized round cells, a lobular pattern, focal spindling, myxoid stroma, and patchy necrosis. The tumor diffusely expressed NUTM1, was positive for WT1cter at weak to moderate intensity, and was focally positive for CD99, while it was negative for keratins, EMA, P40, MyoD1, myogenin, NKX2.2, BCOR, and pan-TRK. Fluorescence in situ hybridization analyses revealed cleavage of the CIC and NUTM1 genes. CONCLUSION: CIC::NUTM1 sarcomas represent a novel molecular variant of CRS with a preference for the central nervous system and younger pediatric persons. Its morphology and phenotype may be mistaken for NUT carcinomas, and the behavior is more progressive than other forms of CRS. For this rare and newly discovered gene fusion variant, it is necessary to integrate molecular and immunohistochemical findings with morphologic features in the diagnosis of undifferentiated neoplasms.


Subject(s)
Repressor Proteins , Soft Tissue Neoplasms , Humans , Male , Adolescent , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/pathology , Repressor Proteins/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Oncogene Proteins, Fusion/genetics , Sarcoma/genetics , Sarcoma/pathology , Sarcoma/diagnosis , Upper Extremity/pathology , Gene Rearrangement , Homeobox Protein Nkx-2.2 , In Situ Hybridization, Fluorescence , Transcription Factors , Homeodomain Proteins
3.
Pathol Res Pract ; 259: 155389, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38850845

ABSTRACT

A female in her 60's presented with a left-sided breast mass. A core needle biopsy specimen showed diffuse proliferation of a round cell tumor, which was positive for vimentin, NKX2.2, BCOR, and focal CD99 on immunohistochemistry (IHC). No fusion genes of the Ewing family sarcomas were detected. With a tentative diagnosis of primary breast sarcoma (PBS), total mastectomy was performed after chemotherapy. The resected tissues showed proliferation of round or spindle-shaped tumor cells with a high nuclear-to-cytoplasmic ratio, exhibiting solid and fascicular arrangements but no epithelial component or organoid pattern. While IHC indicated no particular histological diagnosis, genomic examination revealed gene alterations in MED12 p.G44D, MLL2 (KMT2D) p.T1496fs*27, and EGFR variant III (vIII). Moreover, a retrospective IHC study showed overexpression of EGFRvIII. A malignant phyllodes tumor (PT) with extensive sarcomatous overgrowth was indicated as an integrative diagnosis. This is a rare case of a malignant PT harboring EGFRvIII. The present case provides an importance of accurate diagnosis and genomic analysis of rare breast tumors, as malignant PT and PBS are different in its treatment strategy and prognosis.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms , ErbB Receptors , Immunohistochemistry , Mutation , Phyllodes Tumor , Humans , Female , Phyllodes Tumor/genetics , Phyllodes Tumor/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Middle Aged , ErbB Receptors/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Homeobox Protein Nkx-2.2 , DNA-Binding Proteins/genetics , Homeodomain Proteins , Nuclear Proteins , Mediator Complex , Transcription Factors , Neoplasm Proteins
4.
Obesity (Silver Spring) ; 32(7): 1315-1328, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38798028

ABSTRACT

OBJECTIVE: This study aimed to investigate the role of Nkx1-2, a transcription factor with the NK homeobox domain, in the regulation of fat production. METHODS: Gene expression was analyzed using quantitative real-time polymerase chain reaction or transcriptome sequencing. CRISPR/Cas9 technology was employed to generate nkx1.2 knockout zebrafish and nkx1.2-deleted 3T3-L1 cells. Lipid droplet production in zebrafish larvae was visually quantified using Nile red staining, whereas lipid droplets in 3T3-L1 cells were stained with Oil red O. The binding of Nkx1-2 to the promoter was verified through an electrophoretic mobility shift assay experiment. RESULTS: Nkx1-2 plays crucial roles in the regulation of fat production in zebrafish. Knockout of nkx1.2 in zebrafish leads to weight loss, accompanied by significantly reduced lipid droplet production and decreased visceral and liver fat content. Furthermore, genes related to lipid biosynthesis are significantly downregulated. In 3T3-L1 preadipocytes, Nkx1-2 induces differentiation into mature adipocytes by binding to the cebpa promoter, thereby activating its transcription. Additionally, the expression of nkx1.2 is regulated by the p38 MAPK, JNK, or Smad2/3 signaling pathways in 3T3-L1 cells. CONCLUSIONS: Our findings suggest that Nkx1-2 functions as a positive regulator of fat production, playing a critical role in adipocyte differentiation and lipid biosynthesis.


Subject(s)
3T3-L1 Cells , Homeodomain Proteins , Transcription Factors , Zebrafish Proteins , Zebrafish , Animals , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Adipocytes/metabolism , Adipogenesis/genetics , Cell Differentiation , CRISPR-Cas Systems , Signal Transduction , Lipid Droplets/metabolism , Promoter Regions, Genetic , Lipid Metabolism/genetics , Larva/metabolism , Homeobox Protein Nkx-2.2
5.
J Cancer Res Clin Oncol ; 150(5): 267, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38769118

ABSTRACT

BACKGROUND: Ewing's sarcoma (ES) is an aggressive cancer of bone and soft tissue, most of which tend to occur in the bone. Extraosseous Ewing's sarcoma (EES) of the cervix is extremely rare. CASE PRESENTATION: In the present work, we reported a 39-year-old cervical EES patient with a 2.5*2.1*1.8 cm tumor mass. According to previous literatures, our case is the smallest tumor found in primary cervical ES ever. The patient initially came to our hospital due to vaginal bleeding, and then the gynecological examination found a neoplasm between the cervical canal and partially in the external cervical orifice. The diagnosis of EES was confirmed below: Hematoxylin & Eosin staining (H&E) revealed small round blue malignant cells in biopsy specimens. Immunohistochemistry (IHC) showed the positive staining for CD99, NKX2.2, and FLI1. Disruption of EWSR1 gene was found by fluorescence in situ hybridization (FISH), and the EWSR1-FLI1 gene fusion was determined by next-generation sequencing (NGS). The patient received laparoscopic wide hysterectomy, bilateral adnexectomy, pelvic lymphadenectomy, and postoperative adjuvant chemotherapy and remained disease free with regular follow-up for 1 year. CONCLUSIONS: Through a systematic review of previously reported cervical ES and this case, we highlighted the importance of FISH and NGS for the accuracy of ESS diagnosis, which could assist on the optimal treatment strategy. However, due to the rarity of the disease, there is no standard treatment schemes. Investigation on molecular pathological diagnosis and standardization of treatment regimens for cervical ES are critical to patients' prognosis.


Subject(s)
Sarcoma, Ewing , Uterine Cervical Neoplasms , Humans , Female , Sarcoma, Ewing/pathology , Sarcoma, Ewing/genetics , Sarcoma, Ewing/diagnosis , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/surgery , Adult , Oncogene Proteins, Fusion/genetics , Homeobox Protein Nkx-2.2 , RNA-Binding Protein EWS/genetics , In Situ Hybridization, Fluorescence , Transcription Factors/genetics , Proto-Oncogene Protein c-fli-1/genetics , Nuclear Proteins , Homeodomain Proteins
6.
Ann Diagn Pathol ; 72: 152321, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38759563

ABSTRACT

Retroperitoneal Ewing sarcomas (RES) are very rare and mostly described in case reports. The purpose of this study was to retrospectively analyze the clinicopathology, molecular characteristics, biological behavior, and therapeutic information of 13 cases of primary RES with immunohistochemical staining, fluorescence in situ hybridization, RT-PCR and NGS sequencing detection techniques. The thirteen patients included eight males and five females with a mean age of 34 years. Morphologically, the tumors were comprised of small round or epithelial-like cells with vacuolated cytoplasm (6/13,46 %) arranged in diffuse, nested (8/13,62 %) and perivascular (7/13,54 %) patterns. Unusual morphologic patterns, such as meningioma-like swirling structures and sieve-like structures were relatively novel findings. Immunohistochemical studies showed CD99 (12/13; 92 %), CD56 (11/13; 85 %), NKX2.2 (9/13; 69 %), PAX7 (10/11;91 %) and CD117(6/9;67 %) to be positive.12 cases (92 %) demonstrated EWSR1 rearrangement and 3 cases displayed EWSR1::FLI1 fusion by FISH. ERCC4 splice-site variant, a novel pathogenic variant, was discovered for the first time via RNA sequencing. With a median follow-up duration of 14 months (6 to 79 months), 8/13 (62 %) patients died, while 5/13(38 %) survived. Three cases recurred, and five patients developed metastasis to the liver (2 cases), lung (2 cases) and bone (1 case). RES is an aggressive, high-grade tumor, prone to multiple recurrences and metastases, with distinctive morphologic, immunohistochemical, and molecular genetic features. ERCC4 splicing mutation, which is a novel pathogenic variant discovered for the first time, with possible significance for understanding the disease, as well as the development of targeted drugs.


Subject(s)
Homeobox Protein Nkx-2.2 , RNA-Binding Protein EWS , Retroperitoneal Neoplasms , Sarcoma, Ewing , Humans , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/metabolism , Male , Female , Adult , Retroperitoneal Neoplasms/genetics , Retroperitoneal Neoplasms/pathology , Retroperitoneal Neoplasms/diagnosis , Retrospective Studies , Middle Aged , Young Adult , Adolescent , RNA-Binding Protein EWS/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , In Situ Hybridization, Fluorescence/methods , Gene Rearrangement , Immunohistochemistry/methods , Transcription Factors/genetics , Transcription Factors/metabolism , Oncogene Proteins, Fusion/genetics , Child , Nuclear Proteins , Homeodomain Proteins , Zebrafish Proteins
7.
BMC Genomics ; 25(1): 427, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689254

ABSTRACT

BACKGROUND: Current approaches to profile the single-cell transcriptomics of human pancreatic endocrine cells almost exclusively rely on freshly isolated islets. However, human islets are limited in availability. Furthermore, the extensive processing steps during islet isolation and subsequent single cell dissolution might alter gene expressions. In this work, we report the development of a single-nucleus RNA sequencing (snRNA-seq) approach with targeted islet cell enrichment for endocrine-population focused transcriptomic profiling using frozen archival pancreatic tissues without islet isolation. RESULTS: We cross-compared five nuclei isolation protocols and selected the citric acid method as the best strategy to isolate nuclei with high RNA integrity and low cytoplasmic contamination from frozen archival human pancreata. We innovated fluorescence-activated nuclei sorting based on the positive signal of NKX2-2 antibody to enrich nuclei of the endocrine population from the entire nuclei pool of the pancreas. Our sample preparation procedure generated high-quality single-nucleus gene-expression libraries while preserving the endocrine population diversity. In comparison with single-cell RNA sequencing (scRNA-seq) library generated with live cells from freshly isolated human islets, the snRNA-seq library displayed comparable endocrine cellular composition and cell type signature gene expression. However, between these two types of libraries, differential enrichments of transcripts belonging to different functional classes could be observed. CONCLUSIONS: Our work fills a technological gap and helps to unleash frozen archival pancreatic tissues for molecular profiling targeting the endocrine population. This study opens doors to retrospective mappings of endocrine cell dynamics in pancreatic tissues of complex histopathology. We expect that our protocol is applicable to enrich nuclei for transcriptomics studies from various populations in different types of frozen archival tissues.


Subject(s)
Cell Nucleus , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Islets of Langerhans , Nuclear Proteins , Sequence Analysis, RNA , Single-Cell Analysis , Transcription Factors , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Single-Cell Analysis/methods , Sequence Analysis, RNA/methods , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Profiling/methods , Pancreas/metabolism , Pancreas/cytology , Transcriptome
8.
Glia ; 72(7): 1304-1318, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38546197

ABSTRACT

Oligodendrocyte differentiation and myelination in the central nervous system are controlled and coordinated by a complex gene regulatory network that contains several transcription factors, including Zfp488 and Nkx2.2. Despite the proven role in oligodendrocyte differentiation little is known about the exact mode of Zfp488 and Nkx2.2 action, including their target genes. Here, we used overexpression of Zfp488 and Nkx2.2 in differentiating CG4 cells to identify aspects of the oligodendroglial expression profile that depend on these transcription factors. Although both transcription factors are primarily described as repressors, the detected changes argue for an additional function as activators. Among the genes activated by both Zfp488 and Nkx2.2 was the G protein-coupled receptor Gpr37 that is important during myelination. In agreement with a positive effect on Gpr37 expression, downregulation of the G protein-coupled receptor was observed in Zfp488- and in Nkx2.2-deficient oligodendrocytes in the mouse. We also identified several potential regulatory regions of the Gpr37 gene. Although Zfp488 and Nkx2.2 both bind to one of the regulatory regions downstream of the Gpr37 gene in vivo, none of the regulatory regions was activated by either transcription factor alone. Increased activation by Zfp488 or Nkx2.2 was only observed in the presence of Sox10, a transcription factor continuously present in oligodendroglial cells. Our results argue that both Zfp488 and Nkx2.2 also act as transcriptional activators during oligodendrocyte differentiation and cooperate with Sox10 to allow the expression of Gpr37 as a modulator of the myelination process.


Subject(s)
Cell Differentiation , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Oligodendroglia , Receptors, G-Protein-Coupled , SOXE Transcription Factors , Transcription Factors , Animals , Female , Male , Mice , Cell Differentiation/physiology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , SOXE Transcription Factors/metabolism , SOXE Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
9.
Genes Dev ; 37(11-12): 451-453, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37399332

ABSTRACT

The homeodomain transcription factor (TF) Nkx2.2 governs crucial cell fate decisions in several developing organs, including the central nervous system (CNS), pancreas, and intestine. How Nkx2.2 regulates unique targets in these different systems to impact their individual transcriptional programs remains unclear. In this issue of Genes & Development Abarinov and colleagues (pp. 490-504) generated and analyzed mice in which the Nkx2.2 SD is mutated and found that the SD is required for normal pancreatic islet differentiation but dispensable for most aspects of neuronal differentiation.


Subject(s)
Homeodomain Proteins , Islets of Langerhans , Mice , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeobox Protein Nkx-2.2 , Zebrafish Proteins/genetics , Islets of Langerhans/metabolism , Cell Differentiation/genetics , Neurons/metabolism , Gene Expression Regulation, Developmental
10.
Genes Dev ; 37(11-12): 490-504, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37364986

ABSTRACT

The consolidation of unambiguous cell fate commitment relies on the ability of transcription factors (TFs) to exert tissue-specific regulation of complex genetic networks. However, the mechanisms by which TFs establish such precise control over gene expression have remained elusive-especially in instances in which a single TF operates in two or more discrete cellular systems. In this study, we demonstrate that ß cell-specific functions of NKX2.2 are driven by the highly conserved NK2-specific domain (SD). Mutation of the endogenous NKX2.2 SD prevents the developmental progression of ß cell precursors into mature, insulin-expressing ß cells, resulting in overt neonatal diabetes. Within the adult ß cell, the SD stimulates ß cell performance through the activation and repression of a subset of NKX2.2-regulated transcripts critical for ß cell function. These irregularities in ß cell gene expression may be mediated via SD-contingent interactions with components of chromatin remodelers and the nuclear pore complex. However, in stark contrast to these pancreatic phenotypes, the SD is entirely dispensable for the development of NKX2.2-dependent cell types within the CNS. Together, these results reveal a previously undetermined mechanism through which NKX2.2 directs disparate transcriptional programs in the pancreas versus neuroepithelium.


Subject(s)
Homeodomain Proteins , Insulin-Secreting Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeobox Protein Nkx-2.2 , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Differentiation , Zebrafish Proteins/genetics
11.
Diagn Cytopathol ; 51(6): E189-E194, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36999306

ABSTRACT

Immunohistochemistry plays a vital role in the diagnosis of small round cell tumors. CD99 immunonegativity is one of the features, which helps in distinguishing neuroblastoma from other small round cell tumors. NKX2.2 is a specific marker of Ewing sarcoma, which is a differential for poorly differentiated neuroblastoma. Here, we present a case of metastatic neuroblastoma showing immunoreactivity for both CD99 and NKX2.2 on cytology of the metastatic site causing diagnostic dilemma. Biopsy study of the adrenal lesion revealed presence of differentiating cells and neuropil, highlighting the importance of evaluation of the primary site and limitation of cytology.


Subject(s)
Neuroblastoma , Sarcoma, Ewing , Sarcoma , Humans , Transcription Factors , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Nuclear Proteins , Sarcoma, Ewing/pathology , Sarcoma/diagnosis , Neuroblastoma/diagnosis , Diagnosis, Differential , Biomarkers, Tumor , 12E7 Antigen
12.
Sci Rep ; 13(1): 2008, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36737643

ABSTRACT

V3 spinal interneurons are a key element of the spinal circuits, which control motor function. However, to date, there are no effective ways of deriving a pure V3 population from human pluripotent stem cells. Here, we report a method for differentiation and isolation of spinal V3 interneurons, combining extrinsic factor-mediated differentiation and magnetic activated cell sorting. We found that differentiation of V3 progenitors can be enhanced with a higher concentration of Sonic Hedgehog agonist, as well as culturing cells in 3D format. To enable V3 progenitor purification from mixed differentiation cultures, we developed a transgene reporter, with a part of the regulatory region of V3-specific gene Nkx2-2 driving the expression of a membrane marker CD14. We found that in human cells, NKX2-2 initially exhibited co-labelling with motor neuron progenitor marker, but V3 specificity emerged as the differentiation culture progressed. At these later differentiation timepoints, we were able to enrich V3 progenitors labelled with CD14 to ~ 95% purity, and mature them to postmitotic V3 interneurons. This purification tool for V3 interneurons will be useful for in vitro disease modeling, studies of normal human neural development and potential cell therapies for disorders of the spinal cord.


Subject(s)
Human Embryonic Stem Cells , Humans , Cell Differentiation , Hedgehog Proteins/metabolism , Interneurons/metabolism , Motor Neurons/metabolism , Spinal Cord/metabolism , Homeobox Protein Nkx-2.2/genetics
13.
Indian J Pathol Microbiol ; 66(1): 58-62, 2023.
Article in English | MEDLINE | ID: mdl-36656211

ABSTRACT

Context: Ewing sarcoma (ES) are malignant small round cell tumors (MSRCT) characterized by rearrangements of EWSR1 gene. Although gold standard for diagnosis is detection of specific fusion genes by molecular testing, these ancillary tests are costly and only available in limited number of settings. There is a persuasive evidence for reliability of NKX2.2 immunohistochemistry (IHC) as a surrogate marker for EWSR1 gene rearrangement in ES. Aims: The aim of this study is to correlate the NKX2.2 immuno-expression with genetically confirmed ES cases and also to assess the reliability and accuracy of NKX2.2 along with combined positivity of NXX2.2 and CD99 in diagnosing ES and differentiating it from other relevant histological mimics. Settings and Design: The present study is a retrospective study conducted over a period of 6-year duration in a tertiary cancer care center. Methods and Material: We evaluated NKX2.2 immunoexpression in 35 genetically confirmed cases of ES and also in pertaining differential entities (n = 58) of ES including rhabdomyosarcoma (n = 20), lymphoblastic lymphoma (n = 14), Wilms tumor (n = 10), poorly differentiated synovial sarcoma (n = 4), small-cell osteosarcoma (n = 4), neuroblastoma (n = 5), and mesenchymal chondrosarcoma (n = 1). CD99 was performed in the category of MSRCTs showing NKX2.2 positivity to evaluate combined specificity for the diagnosis of ES. Results: Of the 35 genetically confirmed cases of ES, 29 cases (83%) showed NKX2.2-positive expression (83% sensitivity). Compared to ES, NKX2.2 was positive in only 05% cases (3/58 cases) of non-ES MSRCT. Only two of five cases of neuroblastomas and one case of mesenchymal chondrosarcoma showed NKX2.2 positivity. CD99 positivity was seen in 100% of ES and in the single case of mesenchymal chondrosarcoma. All five cases (100%) of neuroblastoma were negative for CD99. Conclusions: The presented study, which is the first from an Indian oncology center, showed NKX2.2 IHC is quite reliable in diagnosis of ES in the right clinicopathological context. With remarkable sensitivity and specificity of NKX2.2 IHC for diagnosis of ES, we propose that combined positivity of CD99 and NKX2.2 IHC can obviate or minimize the need of EWSR1 gene rearrangement molecular testing for diagnosis of ES.


Subject(s)
Chondrosarcoma, Mesenchymal , Neuroblastoma , Neuroectodermal Tumors, Primitive, Peripheral , Sarcoma, Ewing , Sarcoma , Humans , 12E7 Antigen/metabolism , Biomarkers, Tumor/genetics , Immunohistochemistry , Reproducibility of Results , Retrospective Studies , RNA-Binding Protein EWS , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Homeobox Protein Nkx-2.2
14.
Development ; 149(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35815619

ABSTRACT

The midbrain reticular formation (MRF) is a mosaic of diverse GABAergic and glutamatergic neurons that have been associated with a variety of functions, including sleep regulation. However, the molecular characteristics and development of MRF neurons are poorly understood. As the transcription factor, Gata2 is required for the development of all GABAergic neurons derived from the embryonic mouse midbrain, we hypothesized that the genes expressed downstream of Gata2 could contribute to the diversification of GABAergic neuron subtypes in this brain region. Here, we show that Gata2 is required for the expression of several GABAergic lineage-specific transcription factors, including Nkx2-2 and Skor2, which are co-expressed in a restricted group of post-mitotic GABAergic precursors in the MRF. Both Gata2 and Nkx2-2 function is required for Skor2 expression in GABAergic precursors. In the adult mouse and rat midbrain, Nkx2-2-and Skor2-expressing GABAergic neurons locate at the boundary of the ventrolateral periaqueductal gray and the MRF, an area containing REM-off neurons regulating REM sleep. In addition to the characteristic localization, Skor2+ cells increase their activity upon REM-sleep inhibition, send projections to the dorsolateral pons, a region associated with sleep control, and are responsive to orexins, consistent with the known properties of midbrain REM-off neurons.


Subject(s)
GABAergic Neurons , Sleep, REM , Animals , GABAergic Neurons/metabolism , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Homeobox Protein Nkx-2.2/metabolism , Mesencephalon , Mice , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Rats , Sleep/physiology , Sleep, REM/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Virchows Arch ; 480(4): 909-917, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34985580

ABSTRACT

Ewing sarcoma (ES) is an aggressive neoplasm with variable morphology. It has no specific immunoprofile or molecular signature. Neither CD99, NKX2.2 nor PAX7 immunoreactivity alone is completely specific, although diagnostic specificity improves when combined. The purpose of the present study was to investigate the immunohistochemical (IHC) expression of PAX7 in a large series of genetically confirmed ES. Existing results for CD99 and NKX2.2 immunoexpression, morphological findings and molecular studies (fusion gene subtypes) were retrieved from a previous study. Survival analyses were performed in cases with available clinical follow-up. PAX7 was positive in 95.5% of ES with diffuse staining (> 50%) in all positive cases and moderate or strong intensity for most cases. Nineteen ES displayed both PAX7 and CD99 immunoreactivity but lacked NKX2.2 immunoexpression. No relationships could be found between PAX7 expression and the histological types or ES gene fusion subtypes. Univariant/multivariate analysis showed that lack of PAX7 and/or NKX2.2 immunoexpression constitute independent poor prognostic factors for progression free survival (PFS) and overall survival (OS). In conclusion, IHC for CD99, NKX2.2, and PAX7 may be useful in daily practice for ES diagnosis, particularly in hospitals lacking facilities for molecular studies. In addition, the combination of strong CD99 membranous positivity and nuclear PAX7 and NKX2.2 immunoreactivity seems to be very reliable for ES diagnosis when supported by a corroborating histomorphologic and clinical picture. Although PAX7 is not entirely specific for ES, it seems to have a more extensive and strong nuclear immunoreactivity than NKX2.2 expression, even in tumors with decalcification artifact. Considering the prognostically significant data herein reported, we strongly recommend validation in prospective ES series that include localized and disseminated tumors.


Subject(s)
Sarcoma, Ewing , 12E7 Antigen , Biomarkers, Tumor/analysis , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/analysis , Humans , Immunohistochemistry , Nuclear Proteins , PAX7 Transcription Factor , Prognosis , Prospective Studies , Sarcoma, Ewing/genetics , Transcription Factors/analysis
16.
Skeletal Radiol ; 51(1): 209-217, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34110448

ABSTRACT

Adamantinoma-like Ewing sarcoma is uncommonly reported in the skeletal sites, including small bones of the feet.A 15-year-old girl presented with pain and swelling in her left foot, leading to difficulty in walking for 8 months. Plain radiograph revealed an ill-defined, lytic-sclerotic lesion without significant periosteal reaction in her left calcaneus. Magnetic resonance imaging (MRI) revealed an expansile lesion involving the anterior calcaneus, which was hypointense on T1 and heterogeneously hyperintense on T2-weighted sequences, infiltrating the adjacent bones and soft tissues. On imaging, the differential diagnoses considered were a giant cell tumor and other primary bone tumors.Histopathological examination revealed a tumor composed of small round cells, with interspersed keratin pearls. Immunohistochemically, the tumor cells were positive for CD99/MIC2, pan-cytokeratin (AE1/AE3), p40, p63, NKX2.2, and synaptophysin. Diagnosis of adamantinoma-like Ewing sarcoma was offered on the initial biopsy. Furthermore, the tumor cells revealed EWSR1 gene rearrangement by fluorescence in situ hybridization, confirming this diagnosis. The patient underwent neoadjuvant chemotherapy, had a poor response, and finally underwent below-knee amputation.This constitutes a rare case of adamantinoma-like Ewing sarcoma in the calcaneus. Ewing sarcoma may be considered as a differential diagnosis for intraosseous lytic-sclerotic lesions, even without significant periosteal reaction, at unusual sites, such as the bones of the foot. Awareness of this entity and application of ancillary techniques is recommended for its exact diagnosis and in differentiating this rare variant from its diagnostic mimics. This case also indicates a poor chemotherapy response in this unusual variant of Ewing sarcoma, occurring in the calcaneus.


Subject(s)
Adamantinoma , Calcaneus , Carcinoma, Squamous Cell , Sarcoma, Ewing , Adamantinoma/diagnostic imaging , Adamantinoma/genetics , Adolescent , Biomarkers, Tumor/genetics , Calcaneus/diagnostic imaging , Cell Differentiation , Female , Gene Rearrangement , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , In Situ Hybridization, Fluorescence , Nuclear Proteins , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/diagnostic imaging , Sarcoma, Ewing/genetics , Transcription Factors
17.
Nat Med ; 27(11): 1928-1940, 2021 11.
Article in English | MEDLINE | ID: mdl-34663987

ABSTRACT

Genes involved in distinct diabetes types suggest shared disease mechanisms. Here we show that One Cut Homeobox 1 (ONECUT1) mutations cause monogenic recessive syndromic diabetes in two unrelated patients, characterized by intrauterine growth retardation, pancreas hypoplasia and gallbladder agenesis/hypoplasia, and early-onset diabetes in heterozygous relatives. Heterozygous carriers of rare coding variants of ONECUT1 define a distinctive subgroup of diabetic patients with early-onset, nonautoimmune diabetes, who respond well to diabetes treatment. In addition, common regulatory ONECUT1 variants are associated with multifactorial type 2 diabetes. Directed differentiation of human pluripotent stem cells revealed that loss of ONECUT1 impairs pancreatic progenitor formation and a subsequent endocrine program. Loss of ONECUT1 altered transcription factor binding and enhancer activity and NKX2.2/NKX6.1 expression in pancreatic progenitor cells. Collectively, we demonstrate that ONECUT1 controls a transcriptional and epigenetic machinery regulating endocrine development, involved in a spectrum of diabetes, encompassing monogenic (recessive and dominant) as well as multifactorial inheritance. Our findings highlight the broad contribution of ONECUT1 in diabetes pathogenesis, marking an important step toward precision diabetes medicine.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Hepatocyte Nuclear Factor 6/genetics , Pancreas/embryology , Cell Differentiation/genetics , Congenital Abnormalities/genetics , Fetal Growth Retardation/genetics , Gallbladder/abnormalities , Homeobox Protein Nkx-2.2/biosynthesis , Homeodomain Proteins/biosynthesis , Humans , Infant , Infant, Newborn , Male , Multifactorial Inheritance/genetics , Organogenesis/genetics , Pancreas/abnormalities , Pancreatic Diseases/congenital , Pancreatic Diseases/genetics , Pluripotent Stem Cells/cytology , Transcription, Genetic/genetics
18.
Acta Diabetol ; 58(12): 1689-1700, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34426871

ABSTRACT

AIMS: Neonatal diabetes mellitus (NDM) is a rare disease where diabetes presents during the first six months of life. There are two types of this disorder: permanent neonatal diabetes (PNDM) and transient neonatal diabetes mellitus (TNDM). PNDM occurs due to mutations in genes involved in either beta-cell survival, insulin regulation, and secretion. This study aims to define the genetic aetiology and clinical phenotypes of PNDM in a large Egyptian cohort from a single centre. METHODS: Patients with PNDM who were diagnosed, treated, or referred for follow-up between January 2002 and January 2021 were identified and clinically phenotyped. All patients were tested for mutations in EIF2AK3, KCNJ11, ABCC8, INS, FOXP3, GATA4, GATA6, GCK, GLIS3, HNF1B, IER3IP1, PDX1, PTF1A, NEUROD1, NEUROG3, NKX2-2, RFX6, SLC2A2, SLC19A2, STAT3, WFS1, ZFP57 using targeted next-generation sequencing (NGS) panel. INSR gene mutation was tested in one patient who showed clinical features of insulin resistance. RESULTS: Twenty-nine patients from twenty-six families were diagnosed with PNDM. Pathogenic variants were identified in 17/29 patients (59%). EIF2AK3, INS, and KATP channel mutations were the commonest causes with frequency of 17%, 17%, and 14%, respectively. Patients with ABBC8 and KCNJ11 mutations were successfully shifted to sulfonylureas (SU). Paired data of glycosylated haemoglobin before and after SU transfer showed improved glycaemic control; 9.6% versus 7.1%, P = 0.041. CONCLUSIONS: PNDM is a heterogenous disease with variable genotypes and clinical phenotypes among Egyptian patients. EIF2AK3, INS, ABCC8, and KCNJ11 mutations were the commonest causes of PNDM in the study cohort. All patients with KATP channel mutations were effectively treated with glyburide, reflecting the fact that genetic testing for patients with NDM is not only important for diagnosis but also for treatment plan and prognosis.


Subject(s)
Diabetes Mellitus , Diabetes Mellitus/epidemiology , Diabetes Mellitus/genetics , Genetic Testing , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Infant , Insulin/genetics , Membrane Transport Proteins , Mutation , Nuclear Proteins , Phenotype , Transcription Factors
19.
Dev Biol ; 477: 232-240, 2021 09.
Article in English | MEDLINE | ID: mdl-34097879

ABSTRACT

In mammals, multiple cell-signaling pathways and transcription factors regulate development of the embryonic taste system and turnover of taste cells in the adult stage. Using single-cell RNA-Seq of mouse taste cells, we found that the homeobox-containing transcription factor Nkx2-2, a target of the Sonic Hedgehog pathway and a key regulator of the development and regeneration of multiple cell types in the body, is highly expressed in type III taste cells but not in type II or taste stem cells. Using in situ hybridization and immunostaining, we confirmed that Nkx2-2 is expressed specifically in type III taste cells in the endoderm-derived circumvallate and foliate taste papillae but not in the ectoderm-derived fungiform papillae. Lineage tracing revealed that Nkx2-2-expressing cells differentiate into type III, but not type II or type I cells in circumvallate and foliate papillae. Neonatal Nkx2-2-knockout mice did not express key type III taste cell marker genes, while the expression of type II and type I taste cell marker genes were unaffected in these mice. Our findings indicate that Nkx2-2-expressing cells are committed to the type III lineage and that Nkx2-2 may be critical for the development of type III taste cells in the posterior tongue, thus illustrating a key difference in the mechanism of type III cell lineage specification between ectoderm- and endoderm-derived taste fields.


Subject(s)
Cell Lineage/physiology , Homeodomain Proteins/physiology , Taste Buds/embryology , Zebrafish Proteins/physiology , Animals , Animals, Newborn , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/physiology , Cell Count , Cell Lineage/genetics , Female , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/biosynthesis , Male , Mice , RNA-Seq , Taste Buds/cytology , Taste Buds/metabolism , Zebrafish Proteins/biosynthesis
20.
Cells ; 10(6)2021 05 24.
Article in English | MEDLINE | ID: mdl-34073779

ABSTRACT

Ewing's sarcoma (ES) is caused by a chromosomal translocation leading to the formation of the fused EWSFLI1 gene, which codes for an aberrant transcription factor EWSFLI1. The transcriptional targets of EWSFLI1 have been viewed as promising and novel drug targets in the treatment of ES. One such target is six transmembrane epithelial antigen of the prostate 1 (STEAP1), a transmembrane protein that is upregulated by EWSFLI1 in ES. STEAP1 is a hallmark of tumor invasiveness and an indicator of tumor responsiveness to therapy. EWSFLI1 binds to the STEAP1 promoter region, but the mechanism of action by which it upregulates STEAP1 expression in ES is not entirely understood. Upon analysis of the STEAP1 promoter, we predicted two binding sites for NKX2.2, another crucial transcription factor involved in ES pathogenesis. We confirmed the interaction of NKX2.2 with the STEAP1 promoter using chromatin immunoprecipitation (ChIP) analysis. We used single-molecule RNA imaging, biochemical, and genetic studies to identify the novel role of NKX2.2 in regulating STEAP1 expression in ES. Our results show that NKX2.2 is a co-regulator of STEAP1 expression and functions by interacting with the STEAP1 promoter at sites proximal to the reported EWSFLI1 sites. The co-operative interaction of NKX2.2 with EWSFLI1 in regulating STEAP1 holds potential as a new target for therapeutic interventions for ES.


Subject(s)
Antigens, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/physiology , Neoplasm Invasiveness/pathology , Oxidoreductases/genetics , Sarcoma, Ewing/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Neoplasm Invasiveness/genetics , Nuclear Proteins , Oncogene Proteins, Fusion/genetics , Oxidoreductases/metabolism , Promoter Regions, Genetic/genetics , Sarcoma, Ewing/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...