Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 480
Filter
1.
Genes Chromosomes Cancer ; 63(5): e23244, 2024 May.
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
2.
J Cancer Res Clin Oncol ; 150(5): 259, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753081

ABSTRACT

High mobility group AT-hook 2 (HMGA2) is a member of the non-histone chromosomal high mobility group (HMG) protein family, which participate in embryonic development and other biological processes. HMGA2 overexpression is associated with breast cancer (BC) cell growth, proliferation, metastasis, and drug resistance. Furthermore, HMGA2 expression is positively associated with poor prognosis of patients with BC, and inhibiting HMGA2 signaling can stimulate BC cell progression and metastasis. In this review, we focus on HMGA2 expression changes in BC tissues and multiple BC cell lines. Wnt/ß-catenin, STAT3, CNN6, and TRAIL-R2 proteins are upstream mediators of HMGA2 that can induce BC invasion and metastasis. Moreover, microRNAs (miRNAs) can suppress BC cell growth, invasion, and metastasis by inhibiting HMGA2 expression. Furthermore, long noncoding RNAs (LncRNAs) and circular RNAs (CircRNAs) mainly regulate HMGA2 mRNA and protein expression levels by sponging miRNAs, thereby promoting BC development. Additionally, certain small molecule inhibitors can suppress BC drug resistance by reducing HMGA2 expression. Finally, we summarize findings demonstrating that HMGA2 siRNA and HMGA2 siRNA-loaded nanoliposomes can suppress BC progression and metastasis.


Subject(s)
Breast Neoplasms , HMGA2 Protein , Humans , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Female , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Drug Resistance, Neoplasm/genetics
3.
Sci Rep ; 14(1): 9616, 2024 04 26.
Article in English | MEDLINE | ID: mdl-38671227

ABSTRACT

In this study, we aimed to study the role of TCONS_00006091 in the pathogenesis of oral squamous cellular carcinoma (OSCC) transformed from oral lichen planus (OLP). This study recruited 108 OSCC patients which transformed from OLP as the OSCC group and 102 OLP patients with no sign of OSCC as the Control group. ROC curves were plotted to measure the diagnostic values of TCONS_00006091, miR-153, miR-370 and let-7g, and the changes in gene expressions were measured by RT-qPCR. Sequence analysis and luciferase assays were performed to analyze the molecular relationships among these genes. Cell proliferation and apoptosis were observed via MTT and FCM. TCONS_00006091 exhibited a better diagnosis value for OSCC transformed from OLP. OSCC group showed increased TCONS_00006091 expression and decreased expressions of miR-153, miR-370 and let-7g. The levels of SNAI1, IRS and HMGA2 was all significantly increased in OSCC patients. And TCONS_00006091 was found to sponge miR-153, miR-370 and let-7g, while these miRNAs were respectively found to targe SNAI1, IRS and HMGA2. The elevated TCONS_00006091 suppressed the expressions of miR-153, miR-370 and let-7g, leading to the increased expression of SNAI1, IRS and HMGA2. Also, promoted cell proliferation and suppressed apoptosis were observed upon the over-expression of TCONS_00006091. This study demonstrated that the expressions of miR-153, miR-370 and let-7g were down-regulated by the highly expressed TCONS_00006091 in OSCC patients, which accordingly up-regulated the expressions of SNAI1, IRS and HMGA2, resulting in the promoted cell proliferation and suppressed cell apoptosis.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Cell Proliferation , Gene Expression Regulation, Neoplastic , HMGA2 Protein , MicroRNAs , Mouth Neoplasms , Snail Family Transcription Factors , Humans , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Proliferation/genetics , Female , Male , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Apoptosis/genetics , Middle Aged , Up-Regulation , Cell Line, Tumor , Lichen Planus, Oral/genetics , Lichen Planus, Oral/metabolism , Lichen Planus, Oral/pathology
4.
J Diabetes Investig ; 15(6): 711-724, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38483136

ABSTRACT

BACKGROUND: MiRNA let7d-5p has been recently reported to be abnormally expressed in diabetes-associated atherosclerosis (AS). However, it still remains unknown how let7d-5p contributes to the process of atherosclerosis. METHODS: Twenty fresh tissues and a total of 28 wax block specimens from carotid endarterectomy procedures were obtained from the Luoyang Central Hospital affiliated to Zhengzhou University. The expression of let7d-5p was assessed using quantitative RT-PCR (qRT-PCR). A series of in vitro experiments was used to determine the roles of let7d-5p knockdown and overexpression in vascular smooth muscle cells (VSMCs). RESULTS: We discovered that the carotid plaques from diabetic patients had lower expression levels of miR let7d-5p. In VSMCs, the expression of miRNA let7d-5p was significantly lower in high glucose conditions compared with low glucose situations. The proliferation and migration of VSMCs were also inhibited by the overexpression of let7d-5p, whereas the opposite was true when let7d-5p was inhibited, according to gain and loss of function studies. Mechanically, let7d-5p might activate the GSK3ß/ß-catenin signaling pathway via binding to the high mobility group AT-Hook 2 (HMGA2) mRNA in VSMCs. Additionally, GLP-1RA liraglutide may prevent the migration and proliferation of VSMCs by raising let7d-5p levels. CONCLUSIONS: High glucose stimulated the proliferation and migration of VSMCs by regulating the let7d-5p/HMGA2/GSK3ß/ß-catenin pathway, and liraglutide may slow atherosclerosis by increasing the levels of miR let7d-5p.


Subject(s)
Atherosclerosis , Cell Proliferation , Glucose , MicroRNAs , Muscle, Smooth, Vascular , MicroRNAs/genetics , Humans , Atherosclerosis/metabolism , Atherosclerosis/genetics , Glucose/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Cell Movement , Male , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Myocytes, Smooth Muscle/metabolism , Middle Aged , Cells, Cultured , Female , beta Catenin/metabolism , beta Catenin/genetics , Signal Transduction
6.
Stem Cell Res Ther ; 15(1): 54, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38414051

ABSTRACT

BACKGROUND: Unlike in lower vertebrates, Müller glia (MG) in adult mammalian retinas lack the ability to reprogram into neurons after retinal injury or degeneration and exhibit reactive gliosis instead. Whether a transition in MG cell fate from gliosis to reprogramming would help preserve photoreceptors is still under exploration. METHODS: A mouse model of retinitis pigmentosa (RP) was established using MG cell lineage tracing mice by intraperitoneal injection of sodium iodate (SI). The critical time point for the fate determination of MG gliosis was determined through immunohistochemical staining methods. Then, bulk-RNA and single-cell RNA seq techniques were used to elucidate the changes in RNA transcription of the retina and MG at that time point, and new genes that may determine the fate transition of MG were screened. Finally, the selected gene was specifically overexpressed in MG cells through adeno-associated viruses (AAV) in the mouse RP model. Bulk-RNA seq technique, immunohistochemical staining methods, and visual function testing were used to elucidate and validate the mechanism of new genes function on MG cell fate transition and retinal function. RESULTS: Here, we found the critical time point for MG gliosis fate determination was 3 days post SI injection. Hmga2 was screened out as a candidate regulator for the cell fate transition of MG. After retinal injury caused by SI, the Hmga2 protein is temporarily and lowly expressed in MG cells. Overexpression of Hmga2 in MG down-regulated glial cell related genes and up-regulated photoreceptor related genes. Besides, overexpressing Hmga2 exclusively to MG reduced MG gliosis, made MG obtain cone's marker, and retained visual function in mice with acute retinal injury. CONCLUSION: Our results suggested the unique reprogramming properties of Hmga2 in regulating the fate transition of MG and neuroprotective effects on the retina with acute injury. This work uncovers the reprogramming ability of epigenetic factors in MG.


Subject(s)
Ependymoglial Cells , Retinitis Pigmentosa , Animals , Mice , Ependymoglial Cells/metabolism , Gliosis/metabolism , HMGA2 Protein/metabolism , Retina/metabolism , Retinitis Pigmentosa/metabolism , Disease Models, Animal , RNA/metabolism , Neuroglia/metabolism , Mammals
7.
Arab J Gastroenterol ; 25(2): 135-142, 2024 May.
Article in English | MEDLINE | ID: mdl-38378354

ABSTRACT

BACKGROUND AND STUDY AIMS: The high mobility group A2 (HMGA2), a nonhistone nuclear binding protein, modulates transcription by altering the chromatin architecture of the target gene DNA in its specific AT-hooks region. HMGA2 overexpression has been observed in embryonic tissue and many malignant neoplasms. This study sought to verify whether HMGA2 plays a role in the biological functions of gastric cancer cells, such as cell proliferation, invasiveness, migration, and stem cell acquisition, and to provide some ideas for further research on the metastatic mechanism of gastric cancer. PATIENTS AND METHODS: HMGA2's effects on the proliferation, invasiveness, and migration capabilities of gastric cancer cells were individually detected by BrdU, Transwell, and wound healing assays. Western blotting and immunofluorescence were used to evaluate whether HMGA2 could promote the acquisition of gastric cancer cells. Biostatistical analyses were performed using SPSS 17.0 for Windows. RESULTS: HMGA2 expression levels in gastric cancer cell lines were significantly higher than those in human immortalized gastric epithelial cell lines (p < 0.01). Gastric cancer cell proliferation was inhibited when HMGA2 was overexpressed (p < 0.05). The invasiveness and migration capabilities of gastric cancer cells with HMGA2 overexpression were enhanced more than those of the corresponding control groups (p < 0.05). HMGA2 overexpression promotes the stemness acquisition of stem cells from gastric cancer cells. CONCLUSIONS: This study verified that the HMGA2 structural transcription factor promotes invasiveness, migration, and acquisition of gastric cancer cells. Furthermore, our findings provide significant insight for further research on the metastatic mechanism of gastric cancer.


Subject(s)
Cell Movement , Cell Proliferation , HMGA2 Protein , Neoplasm Invasiveness , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Cell Movement/genetics , Cell Line, Tumor , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
8.
J Orthop Res ; 42(6): 1244-1253, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38151824

ABSTRACT

Postmenopausal osteoporosis (PMOP) affects hundreds of millions of elderly women worldwide. The imbalance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption is the key factor in the progression of PMOP. Recently, exosomal circular RNAs have been considered as critical regulators in physiological and pathological progress. However, their roles in PMOP still require further exploration. Herein, we identified that the expression of exosomal circFAM63B significantly increased in PMOP patients and is closely related to bone density. We further demonstrated that circFAM63B inhibits osteogenic differentiation of bone marrow stromal cells and bone formation in ovariectomy mice by using a combination of in vitro and in vivo experiment strategies. Mechanistically, circFAM63B promotes HMGA2 expression by inhibiting miR-578, thereby suppressing bone repair. Our study proved that exosomal circFAM63B suppresses the bone regeneration of PMOP by regulating the miR-578/HMGA2 axis, which may provide new insights into the pathogenesis and development of PMOP. Knocking down exosomal circFAM63B could be regarded as a new strategy for the treatment of PMOP.


Subject(s)
Bone Regeneration , Exosomes , HMGA2 Protein , MicroRNAs , Osteoporosis, Postmenopausal , RNA, Circular , MicroRNAs/metabolism , MicroRNAs/genetics , Osteoporosis, Postmenopausal/metabolism , Osteoporosis, Postmenopausal/genetics , Female , HMGA2 Protein/metabolism , HMGA2 Protein/genetics , Animals , Humans , RNA, Circular/genetics , RNA, Circular/metabolism , Exosomes/metabolism , Mice , Middle Aged , Osteogenesis , Aged , Mice, Inbred C57BL
9.
Aging (Albany NY) ; 15(24): 14930-14944, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38112616

ABSTRACT

OBJECTIVE: Papillary thyroid cancer (PTC) is one of the most serious diseases of the endocrine system. In view of the limited therapeutic effects of current medical methods, this study starts from the molecular level and looks for potential treatments. The interaction between HAGLROS/miR-206/HMGA2 was studied using multi-omics methods, which provided new ideas and methods for future treatments. METHOD: Microarray analysis and R language were used for differential analysis to screening experimental targets of lncRNA, miRNA, and mRNA. qRT-PCR was used to detect RNA expression in tissues and cells. Double luciferase reporter assays analyzed and validated binding relationships between different RNAs. Colony formation, flow cytometry, and transwell assays were used to measure the effect of them on cell proliferation, apoptosis, and migration. RESULT: Microarray analysis identified lncRNAs, miRNAs, and mRNAs differentially expressed in PTC and normal cells, and selected lncRNA HAGLROS, miR-206, and mRNA HMGA2 as study subjects. LncRNA HAGLROS and mRNA HMGA2 were highly expressed in PTC cells while miR-206 was lowly expressed in PTC cells. LncRNA HAGLROS/HMGA2 can inhibit apoptosis of PTC cells, promote proliferation and migration, and miR-206 promotes the above process. HAGLROS and HMGA2 were negatively correlated with miR-206. shHAGLROS promoted miR-206 expression, inhibited HMGA2 expression and repressed PTC tumor growth in mice. CONCLUSIONS: HAGLROS promotes the growth of PTC by competitively binding to miR-206 to promote HMGA2 expression.


Subject(s)
HMGA2 Protein , MicroRNAs , RNA, Long Noncoding , Thyroid Cancer, Papillary , Thyroid Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , HMGA2 Protein/genetics , HMGA2 Protein/metabolism
10.
FEBS Lett ; 597(15): 1977-1988, 2023 08.
Article in English | MEDLINE | ID: mdl-37259564

ABSTRACT

The architectural chromatin factor high-mobility group AT-hook 2 (HMGA2) is causally involved in several human malignancies and pathologies. HMGA2 is not expressed in most normal adult somatic cells, which renders the protein an attractive drug target. An established cell-based compound library screen identified the fibroblast growth factor receptor (FGFR) inhibitor PD173074 as an antagonist of HMGA2-mediated transcriptional reporter gene activation. We determined that PD173074 binds the C-terminus of HMGA2 and interferes with functional coordination of the three AT-hook DNA-binding domains mediated by the C-terminus. The HMGA2-antagonistic effect of PD173074 on transcriptional activation may therefore result from an induced altered DNA-binding mode of HMGA2. PD173074 as a novel HMGA2-specific antagonist could trigger the development of derivates with enhanced attributes and clinical potential.


Subject(s)
Neoplasms , Receptors, Fibroblast Growth Factor , Adult , Humans , Transcriptional Activation , Chromatin , DNA/metabolism , HMGA2 Protein/genetics , HMGA2 Protein/metabolism
11.
Hum Cell ; 36(5): 1741-1754, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37368192

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common primary liver cancer worldwide with high mortality rate, and the N6-methyladenosine (m6A) epigenetic modifications have been reported to be closely associated with the pathogenesis of HCC, but the detailed molecular mechanisms by which m6A regulates HCC progression have not been fully delineated. In this study, we evidenced that the m6A methyltransferase-like 3 (METTL3)-mediated m6A modification contributed to HCC aggressiveness through modulating a novel circ_KIAA1429/miR-133a-3p/HMGA2 axis. Specifically, circ_KIAA1429 was aberrantly overexpressed in HCC tissues and cells, and the expression levels of circ_KIAA1429 was positively regulated by METTL3 in HCC cells in a m6A-dependent manner. Then, functional experiments confirmed that deletion of both circ_KIAA1429 and METTL3 suppressed HCC cell proliferation, migration and cell mitosis in vitro and in vivo, and conversely, circ_KIAA1429 overexpression had opposite effects to accelerate HCC development. Furthermore, the downstream mechanisms by which circ_KIAA1429 regulated HCC progression were uncovered, and we validated that silencing of circ_KIAA1429 restrained the malignant phenotypes in HCC cells through modulating the miR-133a-3p/high mobility group AT-hook 2 (HMGA2) axis. To summarize, our study firstly investigated the involvement of a novel METTL3/m6A/circ_KIAA1429/miR-133a-3p/HMGA2 axis in regulating HCC development, which provided novel indicators for HCC diagnosis, therapy and prognosis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , RNA, Circular , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/pathology , Methyltransferases/genetics , Methyltransferases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , HMGA2 Protein/metabolism
12.
Pharmacol Res ; 190: 106732, 2023 04.
Article in English | MEDLINE | ID: mdl-36931542

ABSTRACT

High mobility group A 2 (HMGA2) is a protein that modulates the structure of chromatin in the nucleus. Importantly, aberrant expression of HMGA2 occurs during carcinogenesis, and this protein is an upstream mediator of cancer hallmarks including evasion of apoptosis, proliferation, invasion, metastasis, and therapy resistance. HMGA2 targets critical signaling pathways such as Wnt/ß-catenin and mTOR in cancer cells. Therefore, suppression of HMGA2 function notably decreases cancer progression and improves outcome in patients. As HMGA2 is mainly oncogenic, targeting expression by non-coding RNAs (ncRNAs) is crucial to take into consideration since it affects HMGA2 function. MicroRNAs (miRNAs) belong to ncRNAs and are master regulators of vital cell processes, which affect all aspects of cancer hallmarks. Long ncRNAs (lncRNAs) and circular RNAs (circRNAs), other members of ncRNAs, are upstream mediators of miRNAs. The current review intends to discuss the importance of the miRNA/HMGA2 axis in modulation of various types of cancer, and mentions lncRNAs and circRNAs, which regulate this axis as upstream mediators. Finally, we discuss the effect of miRNAs and HMGA2 interactions on the response of cancer cells to therapy. Regarding the critical role of HMGA2 in regulation of critical signaling pathways in cancer cells, and considering the confirmed interaction between HMGA2 and one of the master regulators of cancer, miRNAs, targeting miRNA/HMGA2 axis in cancer therapy is promising and this could be the subject of future clinical trial experiments.


Subject(s)
MicroRNAs , Neoplasms , RNA, Long Noncoding , Humans , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , RNA, Circular/genetics , RNA, Long Noncoding/metabolism , RNA, Untranslated/genetics , HMGA2 Protein/metabolism
13.
Gynecol Endocrinol ; 39(1): 2172155, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36809792

ABSTRACT

OBJECTIVE: Granular cells (GCs) are involved in polycystic ovarian syndrome (PCOS) progression. MicroRNA (miR)-23a downregulation is linked to PCOS development. Therefore, this research explored the influences of miR-23a-3p on GC proliferation and apoptosis in PCOS. METHODS: Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were conducted to examine miR-23a-3p and HMGA2 expression in GCs of patients with PCOS. Then, miR-23a-3p and/or HMGA2 expression was altered in GCs (KGN and SVOG), after which miR-23a-3p, HMGA2, Wnt2, and ß-catenin expression, GC viability, and GC apoptosis were measured by RT-qPCR and western blotting, MTT assay, and flow cytometry, respectively. A dual-luciferase reporter gene assay was utilized to assess the targeting relationship between miR-23a-3p and HMGA2. Finally, GC viability and apoptosis were tested after the combined treatment of miR-23a-3p mimic and pcDNA3.1-HMGA2. RESULTS: miR-23a-3p was poorly expressed but HMGA2 was overexpressed in GCs of patients with PCOS. Mechanistically, HMGA2 was negatively targeted by miR-23a-3p in GCs. Furthermore, miR-23a-3p inhibition or HMGA2 upregulation elevated viability and reduced apoptosis of KGN and SVOG cells, along with increased Wnt2 and ß-catenin expression. In KNG cells, HMGA2 overexpression abrogated the impacts of miR-23a-3p overexpression on GC viability and apoptosis. CONCLUSIONS: Collectively, miR-23a-3p decreased HMGA2 expression to block the Wnt/ß-catenin pathway, thereby depressing viability and facilitating apoptosis of GCs.


Subject(s)
HMGA2 Protein , MicroRNAs , Polycystic Ovary Syndrome , Female , Humans , Apoptosis , beta Catenin/genetics , Cell Proliferation , MicroRNAs/genetics , MicroRNAs/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , HMGA2 Protein/metabolism
14.
Genes Chromosomes Cancer ; 62(1): 27-38, 2023 01.
Article in English | MEDLINE | ID: mdl-35822448

ABSTRACT

Uterine leiomyomas, or fibroids, are very common smooth muscle tumors that arise from the myometrium. They can be divided into distinct molecular subtypes. We have previously shown that 3'RNA-sequencing is highly effective in classifying archival formalin-fixed paraffin-embedded (FFPE) leiomyomas according to the underlying mutation. In this study, we performed 3'RNA-sequencing with 111 FFPE leiomyomas previously classified as negative for driver alterations in mediator complex subunit 12 (MED12), high mobility group AT-hook 2 (HMGA2), and fumarate hydratase (FH) by Sanger sequencing and immunohistochemistry. This revealed 43 tumors that displayed expression features typically seen in HMGA2-positive tumors, including overexpression of PLAG1. We explored 12 such leiomyomas by whole-genome sequencing to identify their underlying genomic drivers and to evaluate the feasibility of detecting chromosomal driver alterations from FFPE material. Four tumors with significant HMGA2 overexpression at the protein-level served as controls. We identified chromosomal rearrangements targeting either HMGA2, HMGA1, or PLAG1 in all 16 tumors, demonstrating that it is possible to detect chromosomal driver alterations in archival leiomyoma specimens as old as 18 years. Furthermore, two tumors displayed biallelic loss of DEPDC5 and one tumor harbored a COL4A5-COL4A6 deletion. These observations suggest that instead of only HMGA2-positive leiomyomas, a distinct leiomyoma subtype is characterized by rearrangements targeting either HMGA2, HMGA1, or PLAG1. The results indicate that the frequency of HMGA2-positive leiomyomas may be higher than estimated in previous studies where immunohistochemistry has been used. This study also demonstrates the feasibility of detecting chromosomal driver alterations from archival FFPE material.


Subject(s)
Leiomyoma , Uterine Neoplasms , Female , Humans , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , HMGA1a Protein/genetics , Leiomyoma/genetics , Leiomyoma/pathology , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Fumarate Hydratase/genetics , Chromosome Aberrations , Mutation , Transcription Factors/genetics , RNA , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
15.
Reprod Biol Endocrinol ; 20(1): 146, 2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36180890

ABSTRACT

BACKGROUND: Tamoxifen (TAM) is a frequently-used treatment for breast cancer (BC). But the TAM resistance seriously affects the patient therapeutic effect. Previous research indicated that circular RNAs (circRNAs) might participate in the regulatory processes of BC. Here, we discovered the parts of circular RNA tripartite motif-containing 28 (circTRIM28) in BC. METHODS: CircTRIM28, microRNA-409-3p (miR-409-3p), and high mobility group AT-hook 2 (HMGA2) levels were perceived by qRT-PCR and western blot. Moreover, the biological functions of the cells were examined. Furthermore, dual-luciferase report was employed to reconnoiter the targeted relationship between miR-409-3p and circTRIM28 or HMGA2. RESULTS: CircTRIM28 and HMGA2 were augmented, and the miR-409-3p was repressed in BC. Silencing circTRIM28 enhanced tamoxifen sensitivity and cell apoptosis, whereas hampered cell development in BC cells. In mechanism, circTRIM28 could sponge miR-409-3p to increase HMGA2. In addition, silencing circTRIM28 impeded tumor growth. CONCLUSION: CircTRIM28 facilitated the BC via miR-409-3p/HMGA2.


Subject(s)
Breast Neoplasms , MicroRNAs , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , HMGA2 Protein/metabolism , Humans , MicroRNAs/genetics , RNA, Circular/genetics , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
16.
Mediators Inflamm ; 2022: 1376959, 2022.
Article in English | MEDLINE | ID: mdl-35966335

ABSTRACT

Cerebral infarction is a common disease with a higher disability and fatality rates. The incidence rates of cerebral infarction or cerebral ischemic stroke gradually increase with aging and cerebrovascular disease progression. This study is aimed at evaluating the effects of HMGA2 on cerebral infarction-induced brain tissue damage and its underlying mechanisms. Adult Sprague Dawley rats were pretreated with sh-HMGA2 before cerebral infarction operation. The effect of HMGA2 on the arrangement, distribution, and morphological structure of neurons and the cell apoptosis ratio in brain tissue were detected via hematoxylin and eosin staining, brain-water content, TTC staining, and TUNEL staining. The results from ELISA assay, qPCR, and western blot indicated that downregulation of HMGA2 mitigated inflammatory stress via regulating the expression of TLR4/NF-κB. In addition, results showed that suppressed HMGA2 attenuated the neurological dysfunction of brain injury rats and markedly reduced infarct volume. HMGA2 might be able to alleviate the damage associated with cerebral infarction-induced inflammatory response and cell apoptosis. Moreover, downregulation of HMGA2 had a protective effect on the brain damage derived from cerebral infarction by mediating the TLR4/NF-κB pathway. In conclusion, the current study demonstrated that downregulation of HMGB2 decreased the infarct size, inflammatory responses, and apoptosis in cerebral injury and further had neuroprotective effects against cerebral infarction-induced brain damage. Finally, these results indicated that the downregulation of the TLR4/NF-κB pathway after ischemia by HMGB2 inhibition is a potential mechanism of the neuroprotective effect of cerebral injury.


Subject(s)
Brain Injuries , Brain Ischemia , HMGA2 Protein/metabolism , Neuroprotective Agents , Stroke , Animals , Brain Ischemia/metabolism , Cerebral Infarction , HMGB2 Protein/metabolism , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptor 4/metabolism
17.
Histopathology ; 81(5): 587-599, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35961656

ABSTRACT

HMGA2 overexpression is found in 10-15% of leiomyomas (LM). HMGA2 overexpression is common in variants of hydropic, intravenous and lipo-LM. Cellular or highly cellular LM (CLM) is a LM variant with a less well-defined molecular nature. In this study, we identified and examined 52 hypercellular LM with sclerotic collagen, herein defined as cellular leiomyoma with sclerosis (CLM-S). CLM-S shows large tumour size (average 12.2 cm) and characteristic histology of tumour cells, arranged in cellular fascicles, sheets and trabeculae with abundant dense, pink sclerotic extracellular matrix in bands and nodules and increased vascularity. Tumour cells are uniform with small, round-oval nuclei and scant, pale-eosinophilic to vacuolated cytoplasm reminiscent of pericytes. The differential diagnosis of CLM-S includes conventional CLM, endometrial stromal tumours and perivascular epithelioid cell tumour. Immunohistochemical profile [HMGA2, fumarate hydratase, smooth muscle markers, Melan A and HMB-45] and molecular alterations [by HMGA2 mRNA reverse transcription-polymerase chain reaction (RT-PCR), HMGA2 fluorescence in-situ hybridisation and MED12 sequencing] were analysed in comparison to matched myometrium and CLM controls. Remarkably, 96% (50 of 52) of CLM-S demonstrated diffuse positive immunoreactivity for HMGA2 and up to an 80-fold increase in HMGA2 mRNA, determined by RT-PCR. FISH analysis with break-part probes at intron 3 and the 5' UTR detected HMGA2 rearrangements in 47% (18 of 38) of CLM-S. All CLM-S retained expression of fumarate hydratase. No MED12 mutations were found in any CLM-S. Our findings show that CLM-S has unique and characteristic histomorphology probably driven by HMGA2 overexpression.


Subject(s)
Leiomyoma , Uterine Neoplasms , 5' Untranslated Regions , Female , Fumarate Hydratase/genetics , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Humans , Leiomyoma/genetics , Leiomyoma/pathology , MART-1 Antigen/genetics , RNA, Messenger , Sclerosis , Uterine Neoplasms/pathology
18.
BMC Biol ; 20(1): 171, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35918713

ABSTRACT

BACKGROUND: The high-mobility group Hmga family of proteins are non-histone chromatin-interacting proteins which have been associated with a number of nuclear functions, including heterochromatin formation, replication, recombination, DNA repair, transcription, and formation of enhanceosomes. Due to its role based on dynamic interaction with chromatin, Hmga2 has a pathogenic role in diverse tumors and has been mainly studied in a cancer context; however, whether Hmga2 has similar physiological functions in normal cells remains less explored. Hmga2 was additionally shown to be required during the exit of embryonic stem cells (ESCs) from the ground state of pluripotency, to allow their transition into epiblast-like cells (EpiLCs), and here, we use that system to gain further understanding of normal Hmga2 function. RESULTS: We demonstrated that Hmga2 KO pluripotent stem cells fail to develop into EpiLCs. By using this experimental system, we studied the chromatin changes that take place upon the induction of EpiLCs and we observed that the loss of Hmga2 affects the histone mark H3K27me3, whose levels are higher in Hmga2 KO cells. Accordingly, a sustained expression of polycomb repressive complex 2 (PRC2), responsible for H3K27me3 deposition, was observed in KO cells. However, gene expression differences between differentiating wt vs Hmga2 KO cells did not show any significant enrichments of PRC2 targets. Similarly, endogenous Hmga2 association to chromatin in epiblast stem cells did not show any clear relationships with gene expression modification observed in Hmga2 KO. Hmga2 ChIP-seq confirmed that this protein preferentially binds to the chromatin regions associated with nuclear lamina. Starting from this observation, we demonstrated that nuclear lamina underwent severe alterations when Hmga2 KO or KD cells were induced to exit from the naïve state and this phenomenon is accompanied by a mislocalization of the heterochromatin mark H3K9me3 within the nucleus. As nuclear lamina (NL) is involved in the organization of 3D chromatin structure, we explored the possible effects of Hmga2 loss on this phenomenon. The analysis of Hi-C data in wt and Hmga2 KO cells allowed us to observe that inter-TAD (topologically associated domains) interactions in Hmga2 KO cells are different from those observed in wt cells. These differences clearly show a peculiar compartmentalization of inter-TAD interactions in chromatin regions associated or not to nuclear lamina. CONCLUSIONS: Overall, our results indicate that Hmga2 interacts with heterochromatic lamin-associated domains, and highlight a role for Hmga2 in the crosstalk between chromatin and nuclear lamina, affecting the establishment of inter-TAD interactions.


Subject(s)
Nuclear Envelope , Pluripotent Stem Cells , Chromatin/genetics , Chromatin/metabolism , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Heterochromatin/metabolism , Histones/genetics , Nuclear Envelope/metabolism , Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics
19.
J Biol Chem ; 298(7): 102098, 2022 07.
Article in English | MEDLINE | ID: mdl-35679898

ABSTRACT

Sarcopenia is an aging-associated attenuation of muscular volume and strength and is the major cause of frailty and falls in elderly individuals. The number of individuals with sarcopenia is rapidly increasing worldwide; however, little is known about the underlying mechanisms of the disease. Sarcopenia often copresents with obesity, and some patients with sarcopenia exhibit accumulation of peri-organ or intra-organ adipose tissue as ectopic fat deposition, including atrophied skeletal muscle. In this study, we showed that transplantation of the perimuscular adipose tissue (PMAT) to the hindlimb thigh muscles of young mice decreased the number of integrin α7/CD29-double positive muscular stem/progenitor cells and that the reaction was mediated by PMAT-derived exosomes. We also found that the inhibition of cell proliferation was induced by Let-7d-3p miRNA that targets HMGA2, which is an important transcription factor for stem cell self-renewal, in muscular stem/progenitor cells and the composite molecular reaction in aged adipocytes. Reduction of Let-7 miRNA repressor Lin28 A/B and activation of nuclear factor-kappa B signaling can lead to the accumulation of Let-7d-3p in the exosomes of aged PMAT. These findings suggest a novel crosstalk between adipose tissue and skeletal muscle in the development of aging-associated muscular atrophy and indicate that adipose tissue-derived miRNAs may play a key role in sarcopenia.


Subject(s)
Adipose Tissue/metabolism , Exosomes , HMGA2 Protein/metabolism , MicroRNAs/metabolism , Sarcopenia , Animals , Cell Proliferation , Exosomes/genetics , Mice , MicroRNAs/genetics , Sarcopenia/genetics , Transcription Factors/metabolism
20.
Exp Cell Res ; 418(1): 113271, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35764101

ABSTRACT

Cancer stem cells (CSCs) are a tumorigenic cell subpopulation, which contributes to treatment resistance, tumor recurrence, and metastasis. This study aimed to investigate the role and underlying molecular targets of high mobility group AT-hook 2 (HMGA2) in the progression and CSCs regulation of head and neck squamous cell carcinoma (HNSCC). HMGA2 mRNA and protein expression levels were examined in HNSCC specimens and cells by qRT-PCR, Western blot, and immunohistochemistry. The roles of HMGA2 were validated via loss-of-function and exogenous overexpression experiments in vitro and in vivo, and CSCs properties were assessed by tumorsphere formation assay. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays provided further insight into the molecular mechanisms by which HMGA2 regulates stemness. HMGA2 was abnormally overexpressed in HNSCC, and it promoted the expression of the CSCs markers including SOX2, CD133, CD44, ALDH1A1, and Bmi1. HMGA2 was correlated with stemness, malignant progression, and reduced survival in HNSCC. Luciferase reporter assay indicated that Snai2 was a direct downstream target gene of HMGA2. Mechanistically, ChIP-qPCR assay showed that HMGA2 was recruited to three binding sites on the Snai2 promoter, directly facilitating the transcription of Snai2 in HNSCC. Snai2 overexpression reversed the inhibitory effect of HMGA2 interference on the proliferation, invasion, and metastasis of HNSCC and CSC marker expression in vitro and in vivo. HMGA2 promoted the malignant progression of HNSCC and acquired CSCs properties through direct regulation of Snai2, thereby suggesting that targeting the HMGA2-Snai2 axis might be a promising therapeutic strategy for HNSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HMGA2 Protein/metabolism , Head and Neck Neoplasms/pathology , Humans , Neoplasm Recurrence, Local/genetics , Neoplastic Stem Cells/metabolism , Snail Family Transcription Factors/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...