1
|
Li M, Niu M, Fan X, Chen F, Cao H, Liu Q, Gan S, Yue P, Gao J. LncRNA MIR181A2HG inhibits keratinocytes proliferation through miR-223-3p/SOX6 axis. Aging (Albany NY) 2024; 16:9846-9858. [PMID: 38848163 PMCID: PMC11210253 DOI: 10.18632/aging.205902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/03/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Psoriasis is a complex and recurrent chronic inflammatory skin disease, and the abnormal proliferation of keratinocytes plays a crucial role in the pathogenesis of psoriasis. Long non-coding RNAs (lncRNAs) play an indispensable role in regulating cellular functions. This research aims to explore the potential impact of lncRNA MIR181A2HG on the regulation of keratinocyte proliferation. METHODS The expression level of MIR181A2HG and the mRNA level of KRT6, KRT16, and SOX6 were assessed using qRT-PCR. The viability and proliferation of keratinocytes were evaluated using CCK-8 and EdU assays. Cell cycle analysis was performed using flow cytometry. Dual-luciferase reporter assays were applied to test the interaction among MIR181A2HG/miR-223-3p/SOX6. Protein level was detected by Western blotting analysis. RESULTS The findings indicated that psoriasis lesions tissue exhibited lower levels of MIR181A2HG expression compared to normal tissue. The overexpression of MIR181A2HG resulted in the inhibition of HaCaT keratinocytes proliferation. The knockdown of MIR181A2HG promoted cell proliferation. The dual-luciferase reporter assay and rescue experiments provided evidence of the interaction among MIR181A2HG, SOX6, and miR-223-3p. CONCLUSIONS The lncRNA MIR181A2HG functions as a miR-223-3p sponge targeting SOX6 to regulate the proliferation of keratinocytes, which suggested that MIR181A2HG/miR-223-3p/SOX6 might be potential diagnostic and therapeutic targets for psoriasis.
Collapse
Affiliation(s)
- Mingzhao Li
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, P.R. China
- Key Laboratory of Biochemistry and Molecular Biology, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, P.R. China
| | - Mutian Niu
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, P.R. China
- Key Laboratory of Biochemistry and Molecular Biology, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, P.R. China
| | - Xiaomei Fan
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, P.R. China
- Key Laboratory of Biochemistry and Molecular Biology, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, P.R. China
| | - Fangru Chen
- Department of Dermatology, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, P.R. China
| | - Hui Cao
- Department of Dermatology, The Second Affiliated Hospital of Guilin Medical University, Guilin 541199, Guangxi, P.R. China
| | - Qingbo Liu
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, P.R. China
- Key Laboratory of Biochemistry and Molecular Biology, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, P.R. China
| | - Shaoqin Gan
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, P.R. China
- Key Laboratory of Biochemistry and Molecular Biology, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, P.R. China
| | - Pengpeng Yue
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, P.R. China
- Key Laboratory of Biochemistry and Molecular Biology, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, P.R. China
| | - Jintao Gao
- School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, Guangxi, P.R. China
- Key Laboratory of Biochemistry and Molecular Biology, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin 541199, Guangxi, P.R. China
| |
Collapse
|
2
|
Simbula M, Manchinu MF, Mingoia M, Pala M, Asunis I, Caria CA, Perseu L, Shah M, Crossley M, Moi P, Ristaldi MS. miR-365-3p mediates BCL11A and SOX6 erythroid-specific coregulation: A new player in HbF activation. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102025. [PMID: 37744176 PMCID: PMC10514143 DOI: 10.1016/j.omtn.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 09/01/2023] [Indexed: 09/26/2023]
Abstract
Hemoglobin switching is a complex biological process not yet fully elucidated. The mechanism regulating the suppression of fetal hemoglobin (HbF) expression is of particular interest because of the positive impact of HbF on the course of diseases such as β-thalassemia and sickle cell disease, hereditary hemoglobin disorders that affect the health of countless individuals worldwide. Several transcription factors have been implicated in the control of HbF, of which BCL11A has emerged as a major player in HbF silencing. SOX6 has also been implicated in silencing HbF and is critical to the silencing of the mouse embryonic hemoglobins. BCL11A and SOX6 are co-expressed and physically interact in the erythroid compartment during differentiation. In this study, we observe that BCL11A knockout leads to post-transcriptional downregulation of SOX6 through activation of microRNA (miR)-365-3p. Downregulating SOX6 by transient ectopic expression of miR-365-3p or gene editing activates embryonic and fetal β-like globin gene expression in erythroid cells. The synchronized expression of BCL11A and SOX6 is crucial for hemoglobin switching. In this study, we identified a BCL11A/miR-365-3p/SOX6 evolutionarily conserved pathway, providing insights into the regulation of the embryonic and fetal globin genes suggesting new targets for treating β-hemoglobinopathies.
Collapse
Affiliation(s)
- Michela Simbula
- Istituto Di Ricerca Genetica e Biomedica del Consiglio Nazionale Delle Ricerche (IRGB-CNR), 09042 Monserrato, Italy
| | - Maria Francesca Manchinu
- Istituto Di Ricerca Genetica e Biomedica del Consiglio Nazionale Delle Ricerche (IRGB-CNR), 09042 Monserrato, Italy
| | - Maura Mingoia
- Istituto Di Ricerca Genetica e Biomedica del Consiglio Nazionale Delle Ricerche (IRGB-CNR), 09042 Monserrato, Italy
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi di Cagliari, 09121 Cagliari, Italy
| | - Mauro Pala
- Istituto Di Ricerca Genetica e Biomedica del Consiglio Nazionale Delle Ricerche (IRGB-CNR), 09042 Monserrato, Italy
| | - Isadora Asunis
- Istituto Di Ricerca Genetica e Biomedica del Consiglio Nazionale Delle Ricerche (IRGB-CNR), 09042 Monserrato, Italy
| | - Cristian Antonio Caria
- Istituto Di Ricerca Genetica e Biomedica del Consiglio Nazionale Delle Ricerche (IRGB-CNR), 09042 Monserrato, Italy
| | - Lucia Perseu
- Istituto Di Ricerca Genetica e Biomedica del Consiglio Nazionale Delle Ricerche (IRGB-CNR), 09042 Monserrato, Italy
| | - Manan Shah
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Merlin Crossley
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Paolo Moi
- Istituto Di Ricerca Genetica e Biomedica del Consiglio Nazionale Delle Ricerche (IRGB-CNR), 09042 Monserrato, Italy
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi di Cagliari, 09121 Cagliari, Italy
| | - Maria Serafina Ristaldi
- Istituto Di Ricerca Genetica e Biomedica del Consiglio Nazionale Delle Ricerche (IRGB-CNR), 09042 Monserrato, Italy
| |
Collapse
|
3
|
Diawara M, Martin LJ. Regulatory mechanisms of SoxD transcription factors and their influences on male fertility. Reprod Biol 2023; 23:100823. [PMID: 37979495 DOI: 10.1016/j.repbio.2023.100823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/20/2023]
Abstract
Members of the SRY-related box (SOX) subfamily D (SoxD) of transcription factors are well conserved among vertebrate species and play important roles in different stages of male reproductive development. In mammals, the SoxD subfamily contains three members: SOX5, SOX6 and SOX13. Here, we describe their implications in testicular development and spermatogenesis, contributing to fertility. We also cover the mechanisms of action of SoxD transcription factors in gene regulation throughout male development. The specificity of activation of target genes by SoxD members depends, in part, on their post-translational modifications and interactions with other partners. Sperm production in adult males requires the coordination in the regulation of gene expression by different members of the SoxD subfamily of transcription factors in the testis. Specifically, the regulation of genes promoting adequate spermatogenesis by SoxD members is discussed in comparison between species.
Collapse
Affiliation(s)
- Mariama Diawara
- Biology Department, Université de Moncton, Moncton, New Brunswick E1A 3E9, Canada
| | - Luc J Martin
- Biology Department, Université de Moncton, Moncton, New Brunswick E1A 3E9, Canada.
| |
Collapse
|
4
|
miRNA signatures in diabetic retinopathy and nephropathy: delineating underlying mechanisms. J Physiol Biochem 2022; 78:19-37. [DOI: 10.1007/s13105-021-00867-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
|
5
|
Chen N, Wu D, Li H, Liu Y, Yang H. MiR-17-3p inhibits osteoblast differentiation by downregulating Sox6 expression. FEBS Open Bio 2020; 10:2499-2506. [PMID: 32946669 PMCID: PMC7609786 DOI: 10.1002/2211-5463.12979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/15/2020] [Accepted: 09/10/2020] [Indexed: 11/18/2022] Open
Abstract
Osteoporosis and osteoarthritis are orthopedic disorders that affect millions of elderly people worldwide; stimulation of bone formation is a potential therapeutic strategy for the treatment of these conditions. As the only bone‐forming cells, osteoblasts play a key role in bone reconstruction. The microRNA miR‐17‐3p is downregulated during osteogenic differentiation of human bone marrow mesenchymal stem cells, but its precise role in this process is unknown. Here, we investigated the role of miR‐17‐3p in osteoblast differentiation. An in vitro model of osteogenesis was established by treating MC3T3‐E1 murine preosteoblast cells with bone morphogenetic protein 2 (BMP2). The expression of miR‐17‐3p in BMP2‐induced MC3T3‐E1 cells was detected by reverse transcription‐quantitative PCR, and its effects on cells transfected with miR‐17‐3p mimic or inhibitor were evaluated by Alizarin Red staining, alkaline phosphatase (ALP) activity assay, and by detection of osteoblast markers including the ALP, collagen type I α1 chain, and osteopontin genes. Bioinformatics analysis was carried out to identify putative target genes of miR‐17‐3p, and the luciferase reporter assay was used for functional validation. Rescue experiments were performed to determine whether SRY‐box transcription factor 6 (Sox6) plays a role in the regulation of osteoblast differentiation by miR‐17‐3p. We report that miR‐17‐3p was downregulated upon BMP2‐induced osteoblast differentiation in MC3T3‐E1 cells, and this was accompanied by decreased differentiation and mineralization, ALP activity, and expression of osteogenesis‐related genes. Sox6 was confirmed to be a target gene of miR‐17‐3p in osteoblasts, and the inhibitory effect of miR‐17‐3p on osteoblast differentiation was observed to occur via Sox6. These results suggest the existence of a novel mechanism underlying miRNA‐mediated regulation of osteogenesis, which has potential implications for the treatment of orthopedic disorders.
Collapse
Affiliation(s)
- Nan Chen
- Department of Orthopedics, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Di Wu
- Department of Orthopedics, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hua Li
- Department of Orthopedics, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yi Liu
- Department of Orthopedics, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hao Yang
- Department of Orthopedics, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
6
|
Jiang ZH, Tang YZ, Song HN, Yang M, Li B, Ni CL. miRNA‑342 suppresses renal interstitial fibrosis in diabetic nephropathy by targeting SOX6. Int J Mol Med 2019; 45:45-52. [PMID: 31746345 PMCID: PMC6889927 DOI: 10.3892/ijmm.2019.4388] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022] Open
Abstract
Diabetic kidney disease (DKD) is one of the major microvascular complications in patients with type 1 and/or type 2 diabetes, the first cause of end-stage renal disease (ESRD) in several countries and regions. However, the pathogenesis of DKD and the mechanisms through which it leads to ESRD remain unknown. Thus, in this study, we aimed to elucidate some of these mechanisms. The expression of microRNA (miRNA or miR)-342-3p and SRY-box 6 (SOX6) in the renal tissues of mice with DKD and mouse renal mesangial cells (MCs) was determined by RT-qPCR and western blot analysis. The diabetic kidney environment was established using high-glucose medium. SOX6 was verified as a target gene of miR-342-3p by dual-luciferase activity assay. In addition, western blot analysis was employed to determine the changes in the levels of several biomarkers of fibrosis [transforming growth factor (TGF)-β1, fibronectin (FN), collagen IV (referred to as C-IV) and phosphatase and tensin homolog (PTEN)]. Compared with THE control mice, the expression of miR-342-3p in the kidney tissues of mice with DKD was down-regulated, whereas that of SOX6 was upregulated. The same phenomenon was observed in the MCs cultured in high-glucose medium. Subsequently, miR-342-3p inhibited SOX6 expression, promoted cell proliferation and inhibited the apoptosis of MCs. Moreover, the overexpression of miR-342-3p suppressed high glucose-induced renal interstitial fibrosis. In addition, it was found that miR-342-3p inhibited SOX6 expression by binding to the 3′-UTR of SOX6. On the whole, the findings of this study demonstrate that miR-342-3p suppresses the progression of DKD by inducing the degradation of SOX6. Thus, the miR-342-3p/SOX6 axis may serve as a novel therapeutic target in the treatment of DKD.
Collapse
Affiliation(s)
- Zhen-Huan Jiang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien‑I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300134, P.R. China
| | - Yun-Zhao Tang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien‑I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300134, P.R. China
| | - Hong-Na Song
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien‑I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300134, P.R. China
| | - Min Yang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien‑I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300134, P.R. China
| | - Bin Li
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien‑I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300134, P.R. China
| | - Chang-Lin Ni
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien‑I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300134, P.R. China
| |
Collapse
|
7
|
Ma J, Lwigale P. Transformation of the Transcriptomic Profile of Mouse Periocular Mesenchyme During Formation of the Embryonic Cornea. Invest Ophthalmol Vis Sci 2019; 60:661-676. [PMID: 30786278 PMCID: PMC6383728 DOI: 10.1167/iovs.18-26018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose Defects in neural crest development are a major contributing factor in corneal dysgenesis, but little is known about the genetic landscape during corneal development. The purpose of this study was to provide a detailed transcriptome profile and evaluate changes in gene expression during mouse corneal development. Methods RNA sequencing was used to uncover the transcriptomic profile of periocular mesenchyme (pNC) isolated at embryonic day (E) 10.5 and corneas isolated at E14.5 and E16.5. The spatiotemporal expression of several differentially expressed genes was validated by in situ hybridization. Results Analysis of the whole-transcriptome profile between pNC and embryonic corneas identified 3815 unique differentially expressed genes. Pathway analysis revealed an enrichment of differentially expressed genes involved in signal transduction (retinoic acid, transforming growth factor-β, and Wnt pathways) and transcriptional regulation. Conclusions Our analyses, for the first time, identify a large number of differentially expressed genes during progressive stages of mouse corneal development. Our data provide a comprehensive transcriptomic profile of the developing cornea. Combined, these data serve as a valuable resource for the identification of novel regulatory networks crucial for the advancement of studies in congenital defects, stem cell therapy, bioengineering, and adult corneal diseases.
Collapse
Affiliation(s)
- Justin Ma
- BioSciences Department, Rice University, Houston, Texas, United States
| | - Peter Lwigale
- BioSciences Department, Rice University, Houston, Texas, United States
| |
Collapse
|
8
|
MicroRNA-135a-5p promotes neuronal differentiation of pluripotent embryonal carcinoma cells by repressing Sox6/CD44 pathway. Biochem Biophys Res Commun 2018; 509:603-610. [PMID: 30606481 DOI: 10.1016/j.bbrc.2018.12.162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 12/25/2018] [Indexed: 12/13/2022]
Abstract
MicroRNA-135a-5p has been reported to play a potential role in the generation of new neurons. However, the underlying targets of miR-135a-5p in regulating neuronal differentiation have been poorly understood. Our study recently has uncovered that Sox6 and CD44 genes were significantly downregulated during neuronal differentiation of P19 cells, a multipotent cell type. We then found that Sox6 directly bound to the promoter of CD44. Importantly, we identified Sox6 as a direct target of miR-135a-5p. Additionally, we demonstrated that miR-135a-5p is crucial for the neuronal differentiation of P19 cells. More significantly, we found that Sox6 overexpression could overturn miR-135a-5p-mediated neuronal differentiation and dendrite development. In conclusion, these findings indicated that miR-135a-5p/Sox6/CD44 axis provides an important molecular target mechanism for neurodifferentiation.
Collapse
|
9
|
Roumaud P, Haché J, Martin LJ. Expression profiles of Sox transcription factors within the postnatal rodent testes. Mol Cell Biochem 2018; 447:175-187. [PMID: 29383560 DOI: 10.1007/s11010-018-3302-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 01/25/2018] [Indexed: 12/22/2022]
Abstract
SRY-related box (Sox) transcription factors are conserved among vertebrate species. These proteins regulate multiple processes including sex determination and testis differentiation of the male embryo. Members of the Sox family have been identified in pre- and postnatal testis and are known to play an important role in sex determination (Sry, Sox9), male gonadal development, and fertility (Sox4, Sox8, Sox30). However, their expression profiles per cell types remain elusive. The objectives of this research were to characterize the expression profiles of Sox family members within adult testes using publically available datasets and to determine whether these findings are consistent with literature as well as immunofluorescence and in situ hybridization results. We have found that Sox4, Sox8, Sox9, and Sox12 are highly expressed in Sertoli cells, whereas Sox5, Sox6, and Sox30 were typically expressed in spermatocytes and spermatids. Spermatogonia were characterized by the expressions of Sox3, Sox4, Sox12, Sox13, and Sox18. Hence, these results suggest that Sox transcription factors may play different roles according to cell types of the adult mammalian testis.
Collapse
Affiliation(s)
- Pauline Roumaud
- Biology Department, Université de Moncton, 18, avenue Antonine Maillet, Moncton, NB, E1A 3E9, Canada
| | - Josée Haché
- Biology Department, Université de Moncton, 18, avenue Antonine Maillet, Moncton, NB, E1A 3E9, Canada
| | - Luc J Martin
- Biology Department, Université de Moncton, 18, avenue Antonine Maillet, Moncton, NB, E1A 3E9, Canada.
| |
Collapse
|