Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 971
Filter
1.
Clin Epigenetics ; 16(1): 73, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840187

ABSTRACT

Silver-Russell syndrome (SRS) is a representative imprinting disorder characterized by pre- and postnatal growth failure. We encountered two Japanese SRS cases with a de novo pathogenic frameshift variant of HMGA2 (NM_003483.6:c.138_141delinsCT, p.(Lys46Asnfs*16)) and a de novo ~ 3.4 Mb microdeletion at 12q14.2-q15 involving HMGA2, respectively. Furthermore, we compared clinical features in previously reported patients with various genetic conditions leading to compromised IGF2 expression, i.e., HMGA2 aberrations, PLAG1 aberrations, IGF2 aberrations, and H19/IGF2:IG-DMR epimutations (hypomethylations). The results provide further support for HMGA2 being involved in the development of SRS and imply some characteristic features in patients with HMGA2 aberrations.


Subject(s)
HMGA2 Protein , Silver-Russell Syndrome , Humans , Silver-Russell Syndrome/genetics , HMGA2 Protein/genetics , Male , Female , Frameshift Mutation/genetics , Japan , Genomic Imprinting/genetics , Infant , Insulin-Like Growth Factor II/genetics , DNA Methylation/genetics , Chromosomes, Human, Pair 12/genetics
2.
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
3.
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
4.
World J Surg Oncol ; 22(1): 142, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38802807

ABSTRACT

BACKGROUND: The high mobility group A2 (HMGA2) gene is expressed extensively during early embryonic development but is inactivated in adulthood, and it is also reactivated in various benign and malignant tumors, including breast cancer. We first assessed the potential functional significance of the unstudied deletion polymorphism rs10573247 at the 3'UTR of HMGA2 on miRNA binding using bioinformatic tools, and subsequently, the association between this polymorphism and breast cancer susceptibility was investigated. MATERIALS AND METHODS: We applied the RNAhybrid tool to predict the functional effects of polymorphism rs10573247 located within the 3' UTR of the HMGA2 gene on miRNA binding. Then, following DNA extraction, 141 breast cancer patients and 123 healthy controls were genotyped for polymorphism rs10573247 using RFLP-PCR with the restriction enzyme Eam1104I. RESULTS: Our bioinformatic data have shown that polymorphism rs10573247 is located in the region that serves as a potential target site for eight miRNAs binding. Among them, miR-3125 exhibited decreased binding affinity for the allele delTT (MFE = -21.8) when compared to the allele TT (MFE = -23.9), but miR-4476 increased binding affinity for the allele delTT (MFE = -22.4) compared to the allele TT (MFE = -22.2). In addition, our results showed that the genotype TT/delTT (p = 0.005) and the genotype delTT/delTT (p = 0.029) were significantly associated with an increased risk of developing breast cancer compared to the genotype TT/TT using RFLP-PCR. DISCUSSION AND CONCLUSION: Our findings suggest that polymorphism rs10573247 may contribute to the risk of breast cancer through the functional effect of this polymorphism on miRNA binding.


Subject(s)
3' Untranslated Regions , Breast Neoplasms , Computational Biology , Genetic Predisposition to Disease , HMGA2 Protein , MicroRNAs , Humans , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Case-Control Studies , Computational Biology/methods , HMGA2 Protein/genetics , MicroRNAs/genetics , Middle Aged , 3' Untranslated Regions/genetics , Prognosis , Genotype , Adult , Polymorphism, Single Nucleotide , Biomarkers, Tumor/genetics , Sequence Deletion , Follow-Up Studies , Risk Factors , Polymorphism, Restriction Fragment Length
5.
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
6.
JCI Insight ; 9(6)2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38516887

ABSTRACT

Silver-Russell syndrome (SRS) is a heterogeneous disorder characterized by intrauterine and postnatal growth retardation. HMGA2 variants are a rare cause of SRS and its functional role in human linear growth is unclear. Patients with suspected SRS negative for 11p15LOM/mUPD7 underwent whole-exome and/or targeted-genome sequencing. Mutant HMGA2 protein expression and nuclear localization were assessed. Two Hmga2-knockin mouse models were generated. Five clinical SRS patients harbored HMGA2 variants with differing functional impacts: 2 stop-gain nonsense variants (c.49G>T, c.52C>T), c.166A>G missense variant, and 2 frameshift variants (c.144delC, c.145delA) leading to an identical, extended-length protein. Phenotypic features were highly variable. Nuclear localization was reduced/absent for all variants except c.166A>G. Homozygous knockin mice recapitulating the c.166A>G variant (Hmga2K56E) exhibited a growth-restricted phenotype. An Hmga2Ter76-knockin mouse model lacked detectable full-length Hmga2 protein, similarly to patient 3 and 5 variants. These mice were infertile, with a pygmy phenotype. We report a heterogeneous group of individuals with SRS harboring variants in HMGA2 and describe the first Hmga2 missense knockin mouse model (Hmga2K56E) to our knowledge causing a growth-restricted phenotype. In patients with clinical features of SRS but negative genetic screening, HMGA2 should be included in next-generation sequencing testing approaches.


Subject(s)
HMGA2 Protein , Silver-Russell Syndrome , Animals , Humans , Mice , Base Sequence , Growth Disorders/genetics , HMGA2 Protein/genetics , Phenotype , Silver-Russell Syndrome/genetics , Silver-Russell Syndrome/diagnosis
7.
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
8.
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
10.
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
11.
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
12.
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
13.
FASEB J ; 37(12): e23288, 2023 12.
Article in English | MEDLINE | ID: mdl-37997502

ABSTRACT

Thyroid carcinoma (THCA) is the most common malignancy in the endocrine system. Long intergenic non-coding RNA 2454 (LINC02454) exhibits an HMGA2-like expression pattern, but their relationship and roles in THCA are largely unknown. The present purpose was to delineate the roles of LINC02454 in THCA progression and its molecular mechanisms. We collected THCA tissues from patients and monitored patient survival. THCA cell colony formation, migration, and invasion were evaluated. Metastasis was evaluated by examining EMT markers through Western blotting. Gene interaction was determined with ChIP, RIP, RNA pull-down, and luciferase activity assays. A mouse model of a subcutaneous tumor was used to determine the activity of LINC02454 knockdown in vivo. We found that LINC02454 was highly expressed in THCA, and its upregulation was associated with poor survival. The knockdown of LINC02454 repressed colony formation, migration, and invasion. Moreover, loss of LINC02454 inhibited tumor growth and metastasis in mice. HMGA2 promoted LINC02454 transcription via binding to the LINC02454 promoter, and silencing of HMGA2 suppressed malignant behaviors through downregulation of LINC02454. HMGA2 was a novel functional target of LINC02454 in THCA cells, and knockdown of LINC02454-mediated anti-tumor effects was reversed by HMGA2 overexpression. Mechanically, LINC02454 promoted CREB1 phosphorylation and nuclear translocation, and CREB1 was subsequently bound to the HMGA2 promoter to facilitate its expression. LINC02454 cis-regulates HMGA2 transcription via facilitating CREB1 phosphorylation and nuclear translocation, and, in turn, HMGA2 promotes LINC02454 expression, thus accelerating thyroid carcinoma progression. Our results support therapeutic targets of LINC02454 and HMGA2 for THCA.


Subject(s)
MicroRNAs , Thyroid Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , HMGA2 Protein/genetics , MicroRNAs/genetics , Thyroid Neoplasms/metabolism , Transcriptional Activation , Up-Regulation
14.
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
15.
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
16.
J Vet Intern Med ; 37(3): 1186-1192, 2023.
Article in English | MEDLINE | ID: mdl-37148171

ABSTRACT

BACKGROUND: The HMGA2:c.83G>A variant was identified in Welsh ponies having pleiotropic effects on height and insulin concentration. OBJECTIVE: Determine whether the HMGA2:c.83G>A variant is associated with decreased height and higher basal insulin concentrations across pony breeds. ANIMALS: Two hundred thirty-six ponies across 6 breeds. METHODS: Cross-sectional study. Ponies were genotyped for the HMGA2:c.83G>A variant and phenotyped for height and basal insulin concentrations. Stepwise regression was performed for model analysis using a linear regression model for height and mixed linear model for insulin with farm as a random effect. Coefficient of determination, pairwise comparison of the estimated marginal means and partial correlation coefficients (parcor) were calculated to assess the relationship between HMGA2 genotype and height or insulin. RESULTS: Breed and genotype accounted for 90.5% of the variation in height across breeds, and genotype explained 21% to 44% of the variation within breeds. Breed, genotype, cresty neck score, sex, age, and farm accounted for 45.5% of the variation in insulin, with genotype accounting for 7.1%. The HMGA2 A allele frequency was 62% and correlated with both height (parcor = -0.39; P < .001) and insulin (parcor = 0.22; P = .02). Pairwise comparisons found A/A ponies were >10 cm shorter than other genotypes. Compared with G/G individuals, A/A and G/A individuals had 4.3 µIU/mL (95% confidence interval [CI]: 1.8-10.5) and 2.7 µIU/mL (95% CI: 1.4-5.3) higher basal insulin concentrations, respectively. CONCLUSIONS AND CLINICAL IMPORTANCE: These data demonstrate the pleiotropic effects of the HMGA2:c.83G>A variant and its role in identifying ponies at increased risk for insulin dysregulation.


Subject(s)
HMGA2 Protein , Horse Diseases , Insulin Resistance , Animals , Cross-Sectional Studies , Genotype , Horse Diseases/genetics , Horses , Insulin , Insulin Resistance/physiology , Phenotype , HMGA2 Protein/genetics
17.
J Mol Cell Biol ; 15(3)2023 08 03.
Article in English | MEDLINE | ID: mdl-36945110

ABSTRACT

Subretinal fibrosis is a major cause of the poor visual prognosis for patients with neovascular age-related macular degeneration (nAMD). Myofibroblasts originated from retinal pigment epithelial (RPE) cells through epithelial-mesenchymal transition (EMT) contribute to the fibrosis formation. N6-Methyladenosine (m6A) modification has been implicated in the EMT process and multiple fibrotic diseases. The role of m6A modification in EMT-related subretinal fibrosis has not yet been elucidated. In this study, we found that during subretinal fibrosis in the mouse model of laser-induced choroidal neovascularization, METTL3 was upregulated in RPE cells. Through m6A epitranscriptomic microarray and further verification, high-mobility group AT-hook 2 (HMGA2) was identified as the key downstream target of METTL3, subsequently activating potent EMT-inducing transcription factor SNAIL. Finally, by subretinal injections of adeno-associated virus vectors, we confirmed that METTL3 deficiency in RPE cells could efficiently attenuate subretinal fibrosis in vivo. In conclusion, our present research identified an epigenetic mechanism of METTL3-m6A-HMGA2 in subretinal fibrosis and EMT of RPE cells, providing a novel therapeutic target for subretinal fibrosis secondary to nAMD.


Subject(s)
Epithelial-Mesenchymal Transition , Methyltransferases , Animals , Humans , Mice , Epithelial-Mesenchymal Transition/genetics , Fibrosis , Methyltransferases/genetics , RNA, Messenger/genetics , Transcription Factors , HMGA2 Protein
18.
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
19.
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
20.
Int J Mol Sci ; 24(4)2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36835656

ABSTRACT

The non-histone chromatin binding protein High Mobility Group AT-hook protein 2 (HMGA2) has important functions in chromatin remodeling, and genome maintenance and protection. Expression of HMGA2 is highest in embryonic stem cells, declines during cell differentiation and cell aging, but it is re-expressed in some cancers, where high HMGA2 expression frequently coincides with a poor prognosis. The nuclear functions of HMGA2 cannot be explained by binding to chromatin alone but involve complex interactions with other proteins that are incompletely understood. The present study used biotin proximity labeling, followed by proteomic analysis, to identify the nuclear interaction partners of HMGA2. We tested two different biotin ligase HMGA2 constructs (BioID2 and miniTurbo) with similar results, and identified known and new HMGA2 interaction partners, with functionalities mainly in chromatin biology. These HMGA2 biotin ligase fusion constructs offer exciting new possibilities for interactome discovery research, enabling the monitoring of nuclear HMGA2 interactomes during drug treatments.


Subject(s)
Biotin , HMGA2 Protein , Proteomics , Cell Differentiation , Chromatin , Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...