1.Anemoside B4 regulates fatty acid metabolism reprogramming in mice with colitis-associated cancer.
Xin YANG ; Jing JIA ; Xin-Xu XIE ; Meng-Qiang WAN ; Yu-Lin FENG ; Ying-Ying LUO ; Hui OUYANG ; Jun YU
China Journal of Chinese Materia Medica 2023;48(9):2325-2333
		                        		
		                        			
		                        			The study aimed to investigate the effect of anemoside B4(B4) on fatty acid metabolism in mice with colitis-associated cancer(CAC). The CAC model was established by azoxymethane(AOM)/dextran sodium sulfate(DSS) in mice. Mice were randomly divided into a normal group, a model group, and low-, medium-, and high-dose anemoside B4 groups. After the experiment, the length of the mouse colon and the size of the tumor were measured, and the pathological alterations in the mouse colon were observed using hematoxylin-eosin(HE) staining. The slices of the colon tumor were obtained for spatial metabolome analysis to analyze the distribution of fatty acid metabolism-related substances in the tumor. The mRNA levels of SREBP-1, FAS, ACCα, SCD-1, PPARα, ACOX, UCP-2, and CPT-1 were determined by real-time quantitative PCR(RT-qPCR). The results revealed that the model group showed decreased body weight(P<0.05) and colon length(P<0.001), increased number of tumors, and increased pathological score(P<0.01). Spatial metabolome analysis revealed that the content of fatty acids and their derivatives, carnitine, and phospholipid in the colon tumor was increased. RT-qPCR results indicated that fatty acid de novo synthesis and β-oxidation-related genes, such as SREBP-1, FASN, ACCα, SCD-1, ACOX, UCP-2, and CPT-1 mRNA expression levels increased considerably(P<0.05, P<0.001). After anemoside B4 administration, the colon length increased(P<0.01), and the number of tumors decreased in the high-dose anemoside B4 group(P<0.05). Additionally, spatial metabolome analysis showed that anemoside B4 could decrease the content of fatty acids and their derivatives, carnitine, and phospholipids in colon tumors. Meanwhile, anemoside B4 could also down-regulate the expression of FASN, ACCα, SCD-1, PPARα, ACOX, UCP-2, and CPT-1 in the colon(P<0.05, P<0.01, P<0.001). The findings of this study show that anemoside B4 may inhibit CAC via regulating fatty acid metabolism reprogramming.
		                        		
		                        		
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Sterol Regulatory Element Binding Protein 1
		                        			;
		                        		
		                        			Colitis-Associated Neoplasms
		                        			;
		                        		
		                        			PPAR alpha/genetics*
		                        			;
		                        		
		                        			Colonic Neoplasms/genetics*
		                        			;
		                        		
		                        			Colon
		                        			;
		                        		
		                        			Azoxymethane
		                        			;
		                        		
		                        			RNA, Messenger
		                        			;
		                        		
		                        			Dextran Sulfate
		                        			;
		                        		
		                        			Colitis/drug therapy*
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Disease Models, Animal
		                        			
		                        		
		                        	
2.Mechanism of miR-26a-5p/cAMP response element binding protein 1 molecular axis regulating osteogenic differentiation of adipose-derived mesenchymal stem cells.
Sanfu LIN ; Shoubo CHEN ; Kaibin FANG ; Jinnan SHI ; Wenhua WU ; Wenhuai WANG
Chinese Journal of Reparative and Reconstructive Surgery 2023;37(5):615-621
		                        		
		                        			OBJECTIVE:
		                        			To investigate the regulatory effects of miR-26a-5p on the osteogenic differentiation of adipose-derived mesenchymal stem cells (ADSCs) by regulating cAMP response element binding protein 1 (CREB1).
		                        		
		                        			METHODS:
		                        			The adipose tissues of four 3-4 weeks old female C57BL/6 mice were collected and the cells were isolated and cultured by digestion separation method. After morphological observation and identification by flow cytometry, the 3rd-generation cells were subjected to osteogenic differentiation induction. At 0, 3, 7, and 14 days after osteogenic differentiation induction, the calcium deposition was observed by alizarin red staining, ALP activity was detected, miR- 26a-5p and CREB1 mRNA expressions were examined by real-time fluorescence quantitative PCR, and CREB1 protein and its phosphorylation (phospho-CREB1, p-CREB1) level were measured by Western blot. After the binding sites between miR-26a-5p and CREB1 was predicted by the starBase database, HEK-293T cells were used to conduct a dual-luciferase reporter gene experiment to verify the targeting relationship (represented as luciferase activity after 48 hours of culture). Finally, miR-26a-p inhibitor (experimental group) and the corresponding negative control (control group) were transfected into ADSCs. Alizarin red staining, ALP activity, real-time fluorescent quantitative PCR (miR-26a-5p) and Western blot [CREB1, p-CREB1, Runt-related transcription factor 2 (RUNX2), and osteocalcin (OCN)] were performed at 7 and 14 days after osteogenic induction culture.
		                        		
		                        			RESULTS:
		                        			The cultured cells were identified as ADSCs. With the prolongation of osteogenic induction culture, the number of calcified nodules and ALP activity significantly increased ( P<0.05). The relative expression of miR-26a-5p in the cells gradually decreased, while the relative expressions of CREB1 mRNA and protein, as well as the relative expression of p-CREB1 protein were increased. The differences were significant between 7, 14 days and 0 day ( P<0.05). There was no significant difference in p-CREB1/CREB1 between different time points ( P>0.05). The starBase database predicted that miR-26a-5p and CREB1 had targeted binding sequences, and the dual-luciferase reporter gene experiment revealed that overexpression of miR-26a-5p significantly suppressed CREB1 wild-type luciferase activity ( P<0.05). After 7 and 14 days of osteogenic induction, compared with the control group, the number of calcified nodules, ALP activity, and relative expressions of CREB1, p-CREB1, OCN, and RUNX2 proteins in the experimental group significantly increased ( P<0.05). There was no significant difference in p-CREB1/CREB1 between the two groups ( P>0.05).
		                        		
		                        			CONCLUSION
		                        			Knocking down miR-26a-5p promoted the osteogenic differentiation of ADSCs by up-regulating CREB1 and its phosphorylation.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			Cells, Cultured
		                        			;
		                        		
		                        			Core Binding Factor Alpha 1 Subunit/metabolism*
		                        			;
		                        		
		                        			Cyclic AMP Response Element-Binding Protein/metabolism*
		                        			;
		                        		
		                        			Mesenchymal Stem Cells
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			MicroRNAs/metabolism*
		                        			;
		                        		
		                        			Osteocalcin/metabolism*
		                        			;
		                        		
		                        			Osteogenesis/genetics*
		                        			;
		                        		
		                        			RNA, Messenger/genetics*
		                        			
		                        		
		                        	
3.Genetic analysis of a child with early onset neurodevelopmental disorder with involuntary movement and a literature review.
Wenjing HU ; Hongjun FANG ; Jingwen TANG ; Zhen ZHOU ; Liwen WU
Chinese Journal of Medical Genetics 2023;40(4):385-389
		                        		
		                        			OBJECTIVE:
		                        			To explore the clinical phenotype and genetic basis of a child with early onset neurodevelopmental disorder with involuntary movement (NEDIM).
		                        		
		                        			METHODS:
		                        			A child who presented at Department of Neurology of Hunan Children's Hospital on October 8, 2020 was selected as the study subject. Clinical data of the child were collected. Genomic DNA was extracted from peripheral blood samples of the child and his parents. Whole exome sequencing (WES) was carried out for the child. Candidate variant was verified by Sanger sequencing and bioinformatic analysis. Relevant literature was searched from the CNKI, PubMed and Google Scholar databases to summarize the clinical phenotypes and genetic variants of the patients.
		                        		
		                        			RESULTS:
		                        			This child was a 3-year-and-3-month boy with involuntary trembling of limbs and motor and language delay. WES revealed that the child has harbored a c.626G>A (p.Arg209His) variant of the GNAO1 gene. Sanger sequencing confirmed that neither of his parents has carried the same variant. The variant had been reported in HGMD and ClinVar databases, but not in the dbSNP, ExAC and 1000 Genomes databases. Prediction with SIFT, PolyPhen-2, and Mutation Taster online software suggested that the variant may be deleterious to the protein function. By UniProt database analysis, the encode amino acid is highly conserved among various species. Prediction with Modeller and PyMOL software indicated that the variant may affect the function of GαO protein. Based on the guideline of the American College of Medical Genetics and Genomics (ACMG), the variant was rated as pathogenic.
		                        		
		                        			CONCLUSION
		                        			The GNAO1 gene c.626G>A (p.Arg209His) variant probably underlay the NEDIM in this child. Above finding has expanded the phenotypic spectrum of GNAO1 gene c.626G>A (p.Arg209His) variant and provided a reference for clinical diagnosis and genetic counseling.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Computational Biology
		                        			;
		                        		
		                        			Genetic Counseling
		                        			;
		                        		
		                        			Genomics
		                        			;
		                        		
		                        			Mutation
		                        			;
		                        		
		                        			Neurodevelopmental Disorders/genetics*
		                        			;
		                        		
		                        			Dyskinesias
		                        			;
		                        		
		                        			GTP-Binding Protein alpha Subunits, Gi-Go
		                        			
		                        		
		                        	
4.Association of insulin signaling pathway -related gene polymorphisms and gene -gene interactions with MAFLD in obese children.
Xiang XIAO ; Junxia YAN ; Ning'an XU ; Rutong KANG ; Jiayou LUO ; Yan ZHONG
Journal of Central South University(Medical Sciences) 2023;48(4):516-525
		                        		
		                        			OBJECTIVES:
		                        			Insulin signaling pathway plays an important role in metabolic associated fatty liver disease (MAFLD), however, the association between polymorphisms of genes related to insulin signaling pathway and MAFLD remains unclear. This study aims to investigate the association between insulin signaling pathway-related gene polymorphisms and gene-gene interactions with MAFLD susceptibility in obese children so as to provide scientific basis for further study of genetic mechanism.
		                        		
		                        			METHODS:
		                        			A total of 502 obese children with MAFLD who admitted to Hunan Provincial Children's Hospital from September 2019 to October 2021, were recruited as a case group, and 421 obese children with non-MAFLD admitted during the same period were recruited as a control group. Socio-demographic information, preterm birth history, eating habits, and exercise status of the subjects were collected by inquiry survey, and anthropometric information was collected by physical measurement. At the same time, 2 mL of venous blood was collected to extract DNA, and the polymorphism of insulin signaling pathway-related genes (5 representative candidate genes, 12 variants) was detected. Multivariate Logistic regression analysis was used to investigate the association between insulin signaling pathway-related gene polymorphisms and MAFLD in obese children.
		                        		
		                        			RESULTS:
		                        			After adjusting for confounder factors, INS rs3842748 was significantly associated with the risk of MAFLD in obese children in allele, heterozygous, and dominant models [OR and 95% CI 1.749 (1.053 to 2.905), 1.909 (1.115 to 3.267), 1.862 (1.098 to 3.157), all P<0.05]; INS rs3842752 was significantly associated with the risk of MAFLD in obese children in heterozygous and dominant models [OR and 95% CI 1.736 (1.028 to 2.932), 1.700 (1.015 to 2.846), all P<0.05]. NR1H3 rs3758674 was significantly correlated with the risk of MAFLD in obese children in allele model [OR and 95% CI 0.716 (0.514 to 0.997), P<0.05]. SREBP-1c rs2297508 was significantly associated with the risk of MAFLD in obese children in allele and dominant models [OR and 95% CI 0.772 (0.602 to 0.991) and 0.743 (0.557 to 0.991), all P<0.05]. SREBP-1c rs8066560 was significantly associated with the risk of MAFLD in obese children in allele, heterozygous, and dominant models [OR and 95% CI 0.759 (0.589 to 0.980), 0.733 (0.541 to 0.992), 0.727 (0.543 to 0.974), all P<0.05]. NR1H3 rs3758674 mutant C and SREBP-1c rs2297508 mutant G had interaction in the development of MAFLD in obese children [OR and 95% CI 0.407 (0.173 to 0.954), P<0.05].
		                        		
		                        			CONCLUSIONS
		                        			The INS, NR1H3, and SREBP-1c gene polymorphisms in the insulin signaling pathway are associated with the susceptibility of MAFLD in obese children, but the functions and mechanisms of these genes need to be further studied.
		                        		
		                        		
		                        		
		                        			Child
		                        			;
		                        		
		                        			Infant, Newborn
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Pediatric Obesity/genetics*
		                        			;
		                        		
		                        			Sterol Regulatory Element Binding Protein 1
		                        			;
		                        		
		                        			Premature Birth
		                        			;
		                        		
		                        			Non-alcoholic Fatty Liver Disease
		                        			;
		                        		
		                        			Signal Transduction/genetics*
		                        			;
		                        		
		                        			Insulins
		                        			
		                        		
		                        	
5.Establishment of leukemia cell model with inducible AML1-ETO expression and its effect on fatty acid metabolism in leukemia cells.
Wan Qing XIE ; Xue YANG ; Run Xia GU ; Zheng TIAN ; Hai Yan XING ; Ke Jing TANG ; Qing RAO ; Shao Wei QIU ; Min WANG ; Jian Xiang WANG
Chinese Journal of Hematology 2023;44(5):366-372
		                        		
		                        			
		                        			Objective: To investigate the effect of the AML1-ETO (AE) fusion gene on the biological function of U937 leukemia cells by establishing a leukemia cell model that induces AE fusion gene expression. Methods: The doxycycline (Dox) -dependent expression of the AE fusion gene in the U937 cell line (U937-AE) were established using a lentivirus vector system. The Cell Counting Kit 8 methods, including the PI and sidanilide induction, were used to detect cell proliferation, cell cycle-induced differentiation assays, respectively. The effect of the AE fusion gene on the biological function of U937-AE cells was preliminarily explored using transcriptome sequencing and metabonomic sequencing. Results: ①The Dox-dependent Tet-on regulatory system was successfully constructed to regulate the stable AE fusion gene expression in U937-AE cells. ②Cell proliferation slowed down and the cell proliferation rate with AE expression (3.47±0.07) was lower than AE non-expression (3.86 ± 0.05) after inducing the AE fusion gene expression for 24 h (P<0.05). The proportion of cells in the G(0)/G(1) phase in the cell cycle increased, with AE expression [ (63.45±3.10) %) ] was higher than AE non-expression [ (41.36± 9.56) %] (P<0.05). The proportion of cells expressing CD13 and CD14 decreased with the expression of AE. The AE negative group is significantly higher than the AE positive group (P<0.05). ③The enrichment analysis of the transcriptome sequencing gene set revealed significantly enriched quiescence, nuclear factor kappa-light-chain-enhancer of activated B cells, interferon-α/γ, and other inflammatory response and immune regulation signals after AE expression. ④Disorder of fatty acid metabolism of U937-AE cells occurred under the influence of AE. The concentration of the medium and short-chain fatty acid acylcarnitine metabolites decreased in cells with AE expressing, propionyl L-carnitine, wherein those with AE expression (0.46±0.13) were lower than those with AE non-expression (1.00±0.27) (P<0.05). The metabolite concentration of some long-chain fatty acid acylcarnitine increased in cells with AE expressing tetradecanoyl carnitine, wherein those with AE expression (1.26±0.01) were higher than those with AE non-expression (1.00±0.05) (P<0.05) . Conclusion: This study successfully established a leukemia cell model that can induce AE expression. The AE expression blocked the cell cycle and inhibited cell differentiation. The gene sets related to the inflammatory reactions was significantly enriched in U937-AE cells that express AE, and fatty acid metabolism was disordered.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			U937 Cells
		                        			;
		                        		
		                        			RUNX1 Translocation Partner 1 Protein
		                        			;
		                        		
		                        			Leukemia/genetics*
		                        			;
		                        		
		                        			Core Binding Factor Alpha 2 Subunit/genetics*
		                        			;
		                        		
		                        			Oncogene Proteins, Fusion/genetics*
		                        			;
		                        		
		                        			Leukemia, Myeloid, Acute/genetics*
		                        			
		                        		
		                        	
8.Expression of TUBB4B in mouse primary spermatocyte GC-2 cells and its regulatory effect on NF-κB and MAPK signaling pathway.
Tongjia LIU ; Wanlun WANG ; Ting ZHANG ; Shuang LIU ; Yanchao BIAN ; Chuanling ZHANG ; Rui XIAO
Journal of Southern Medical University 2023;43(6):1002-1009
		                        		
		                        			OBJECTIVE:
		                        			To explore the interaction between Tubulin beta 4B class IVb (TUBB4B) and Agtpbp1/cytosolic carboxypeptidase- like1 (CCP1) in mouse primary spermatocytes (GC-2 cells) and the role of TUBB4B in regulating the development of GC-2 cells.
		                        		
		                        			METHODS:
		                        			Lentiviral vectors were used to infect GC-2 cells to construct TUBB4B knockdown and negative control (NC-KD) cells. The stable cell lines with TUBB4B overexpression (Tubb4b-OE) and the negative control (NC-OE) cells were screened using purinomycin. RT-qPCR and Western blotting were used to verify successful cell modeling and explore the relationship between TUBB4B and CCP1 expressions in GC-2 cells. The effects of TUBB4B silencing and overexpression on the proliferation and cell cycle of GC-2 cells were evaluated using CCK8 assay and flow cytometry. The signaling pathway proteins showing significant changes in response to TUBB4B silencing or overexpression were identified using Western blotting and immunofluorescence assay and then labeled for verification at the cellular level.
		                        		
		                        			RESULTS:
		                        			Both TUBB4B silencing and overexpression in GC-2 cells caused consistent changes in the mRNA and protein expressions of CCP1 (P < 0.05). Similarly, TUBB4B expression also showed consistent changes at the mRNA and protein after CCP1 knockdown and restoration (P < 0.05). TUBB4B knockdown and overexpression had no significant effect on proliferation rate or cell cycle of GC-2 cells, but caused significant changes in the key proteins of the nuclear factor kappa-B (NF-κB) signaling pathway (p65 and p-p65) and the mitogen-activated protein kinase (MAPK) signaling pathway (ErK1/2 and p-Erk1/2) (P < 0.05); CCP1 knockdown induced significant changes in PolyE expression in GC-2 cells (P < 0.05).
		                        		
		                        			CONCLUSIONS
		                        			TUBB4B and CCP1 interact via a mutual positive regulation mechanism in GC-2 cells. CCP-1 can deglutamize TUBB4B, and the latter is involved in the regulation of NF-κB and MAPK signaling pathways in primary spermatocytes.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			GTP-Binding Proteins/metabolism*
		                        			;
		                        		
		                        			Mitogen-Activated Protein Kinases/metabolism*
		                        			;
		                        		
		                        			NF-kappa B/metabolism*
		                        			;
		                        		
		                        			RNA, Messenger
		                        			;
		                        		
		                        			Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism*
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			Spermatocytes
		                        			;
		                        		
		                        			Tubulin/genetics*
		                        			
		                        		
		                        	
9.Clinical feature difference between juvenile amyotrophic lateral sclerosis with SPTLC1 and FUS mutations.
Peishan WANG ; Qiao WEI ; Hongfu LI ; Zhi-Ying WU
Chinese Medical Journal 2023;136(2):176-183
		                        		
		                        			BACKGROUND:
		                        			Juvenile amyotrophic lateral sclerosis (JALS) is an uncommon form of amyotrophic lateral sclerosis whose age at onset (AAO) is defined as prior to 25 years. FUS mutations are the most common cause of JALS. SPTLC1 was recently identified as a disease-causative gene for JALS, which has rarely been reported in Asian populations. Little is known regarding the difference in clinical features between JALS patients carrying FUS and SPTLC1 mutations. This study aimed to screen mutations in JALS patients and to compare the clinical features between JALS patients with FUS and SPTLC1 mutations.
		                        		
		                        			METHODS:
		                        			Sixteen JALS patients were enrolled, including three newly recruited patients between July 2015 and August 2018 from the Second Affiliated Hospital, Zhejiang University School of Medicine. Mutations were screened by whole-exome sequencing. In addition, clinical features such as AAO, onset site and disease duration were extracted and compared between JALS patients carrying FUS and SPTLC1 mutations through a literature review.
		                        		
		                        			RESULTS:
		                        			A novel and de novo SPTLC1 mutation (c.58G>A, p.A20T) was identified in a sporadic patient. Among 16 JALS patients, 7/16 carried FUS mutations and 5/16 carried respective SPTLC1 , SETX , NEFH , DCTN1 , and TARDBP mutations. Compared with FUS mutation patients, those with SPTLC1 mutations had an earlier AAO (7.9 ± 4.6 years vs. 18.1 ± 3.9 years, P  < 0.01), much longer disease duration (512.0 [416.7-607.3] months vs. 33.4 [21.6-45.1] months, P  < 0.01), and no onset of bulbar.
		                        		
		                        			CONCLUSION
		                        			Our findings expand the genetic and phenotypic spectrum of JALS and help to better understand the genotype-phenotype correlation of JALS.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Amyotrophic Lateral Sclerosis/genetics*
		                        			;
		                        		
		                        			DNA Helicases/genetics*
		                        			;
		                        		
		                        			Genetic Association Studies
		                        			;
		                        		
		                        			Multifunctional Enzymes/genetics*
		                        			;
		                        		
		                        			Mutation/genetics*
		                        			;
		                        		
		                        			RNA Helicases/genetics*
		                        			;
		                        		
		                        			RNA-Binding Protein FUS/genetics*
		                        			;
		                        		
		                        			Serine C-Palmitoyltransferase/genetics*
		                        			;
		                        		
		                        			Child, Preschool
		                        			;
		                        		
		                        			Child
		                        			;
		                        		
		                        			Adolescent
		                        			;
		                        		
		                        			Young Adult
		                        			
		                        		
		                        	
10.EWSR1-SMAD3 positive fibroblastic tumor: a clinicopathological analysis.
Hai Yan SU ; Lu ZHAO ; Gang JI ; Qian Lan YAO ; Qian Ming BAI ; Xiao Yan ZHOU ; Jian WANG
Chinese Journal of Pathology 2023;52(1):19-24
		                        		
		                        			
		                        			Objective: To investigate the clinicopathological features, immunophenotypes and molecular genetics of EWSR1-SMAD3 positive fibroblastic tumor (ESFT) with an emphasis on differential diagnosis. Methods: The clinicopathological data, immunohistochemical profiles and molecular profiles of 3 ESFT cases diagnosed at the Department of Pathology, Fudan University Shanghai Cancer Center from 2018 to 2021were analyzed. The related literature was also reviewed. Results: There were two males and one female. The patients were 24, 12 and 36 years old, respectively. All three tumors occurred in the subcutis of the foot with the disease duration of 6 months to 2 years. The tumors were presented with a slowly growing mass or nodule, accompanied with pain in 1 patient. The tumors ranged in size from 0.1 to 1.6 cm (mean, 1.0 cm). Microscopically, the tumors were located in the subcutaneous tissue with a nodular or plexiform growth pattern. They were composed of cellular fascicles of bland spindle cells with elongated nuclei and fine chromatin. One of the tumors infiltrated into adjacent adipose tissue. There was no nuclear atypia or mitotic activities. All three tumors showed prominent stromal hyalinization with zonal pattern present in one case. Focal punctate calcification was noted in two cases. The immunohistochemical studies showed that tumor cells were diffusely positive for ERG and negative for CD31 and CD34, with Ki-67 index less than 2%. Fluorescence in situ hybridization on the two tested cases identified EWSR1 gene rearrangement. The next generation sequencing analysis demonstrated EWSR1-SMAD3 fusion in all three cases. During the follow up, one patient developed local recurrence 24 months after the surgery. Conclusions: ESFT is a benign fibroblastic neoplasm and has a predilection for the foot, characterized by ERG immunoreactivity and EWSR1-SMAD3 fusion. Local recurrence might occur when incompletely excised. Familiarity with its clinicopathological features is helpful in distinguishing it from other spindle cell neoplasms that tend to occur at acral sites.
		                        		
		                        		
		                        		
		                        			Adult
		                        			;
		                        		
		                        			Child
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Biomarkers, Tumor/analysis*
		                        			;
		                        		
		                        			China
		                        			;
		                        		
		                        			In Situ Hybridization, Fluorescence
		                        			;
		                        		
		                        			Neoplasms, Fibrous Tissue/pathology*
		                        			;
		                        		
		                        			RNA-Binding Protein EWS/genetics*
		                        			;
		                        		
		                        			Smad3 Protein/genetics*
		                        			;
		                        		
		                        			Soft Tissue Neoplasms/surgery*
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail