1
|
Liu Y, Li Z, Chen X, Cui X, Gao Z, Jiang R. INSTINCT: Multi-sample integration of spatial chromatin accessibility sequencing data via stochastic domain translation. Nat Commun 2025; 16:1247. [PMID: 39893190 PMCID: PMC11787322 DOI: 10.1038/s41467-025-56535-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
Recent advances in spatial epigenomic techniques have given rise to spatial assay for transposase-accessible chromatin using sequencing (spATAC-seq) data, enabling the characterization of epigenomic heterogeneity and spatial information simultaneously. Integrative analysis of multiple spATAC-seq samples, for which no method has been developed, allows for effective identification and elimination of unwanted non-biological factors within the data, enabling comprehensive exploration of tissue structures and providing a holistic epigenomic landscape, thereby facilitating the discovery of biological implications and the study of regulatory processes. In this article, we present INSTINCT, a method for multi-sample INtegration of Spatial chromaTIN accessibility sequencing data via stochastiC domain Translation. INSTINCT can efficiently handle the high dimensionality of spATAC-seq data and eliminate the complex noise and batch effects of samples through a stochastic domain translation procedure. We demonstrate the superiority and robustness of INSTINCT in integrating spATAC-seq data across multiple simulated scenarios and real datasets. Additionally, we highlight the advantages of INSTINCT in spatial domain identification, visualization, spot-type annotation, and various downstream analyses, including motif enrichment analysis, expression enrichment analysis, and partitioned heritability analysis.
Collapse
Affiliation(s)
- Yuyao Liu
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Zhen Li
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Xiaoyang Chen
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Xuejian Cui
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Zijing Gao
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Rui Jiang
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
2
|
Huang J, Yang H, Chai S, Lin Y, Zhang Z, Huang H, Wan L. Identification of miRNAs related to osteoporosis by high-throughput sequencing. Front Pharmacol 2024; 15:1451695. [PMID: 39175544 PMCID: PMC11338934 DOI: 10.3389/fphar.2024.1451695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
Background Osteoporosis is a major health issue. MicroRNAs (miRNAs) play multiple roles in regulating cell growth and development. High-throughput sequencing technology is widely used nowadays. Objective To screen for and validate miRNAs associated with osteoporosis. Method Bone specimens from patients with (n = 3) and without (n = 3) osteoporosis were collected. High-throughput sequencing was used to screen for miRNAs that were then analyzed using volcano maps, Wayne maps, gene ontology (GO) analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Confirmation of the miRNAs was done using qRT-PCR. Results The analysis of sequencing showed that there were 12 miRNAs that were down-regulated and five miRNAs that were upregulated in osteoporosis. GO and KEGG identified these miRNAs as being associated with bone metabolism. The qRT-PCR results showed that miR-140-5p, miR-127-3p, miR-199b-5p, miR-181a-5p, miR-181d-5p, and miR-542-3p exhibited a decrease of 2.27-, 3.00-, 3.48-, 2.67-, 2.41-, and 1.98-fold (all P < 0.05) in osteoporosis compared to controls. Conversely, miR-486-3p and miR-486-5p demonstrated an increase of 2.17- and 3.89-fold (P < 0.05) (all P < 0.05). Conclusion This study utilized high-throughput sequencing to detect miRNAs that were expressed differently in individuals with osteoporosis. In osteoporosis, six miRNAs (miR-140-5p, miR-127-3p, miR-199b-5p, miR-181a-5p, miR-181d-5p, and miR-542) were found to have decreased expression, whereas two miRNAs (miR-486-3p and miR-486-5p) were found to have increased expression. The initial manifestation of various miRNAs might serve as predictive indicators and potentially anticipate the progression of osteoporosis.
Collapse
Affiliation(s)
- Jiachun Huang
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Haolin Yang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shuang Chai
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yanping Lin
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhihai Zhang
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hongxing Huang
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lei Wan
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Wang W, Ji L, Jing X, Zhao P, Xia Q. MicroRNA let-7 targets BmCDK1 to regulate cell proliferation and endomitosis of silk gland in the silkworm, Bombyx mori. INSECT SCIENCE 2024; 31:1026-1040. [PMID: 38053466 DOI: 10.1111/1744-7917.13302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/17/2023] [Accepted: 10/19/2023] [Indexed: 12/07/2023]
Abstract
MicroRNAs play critical roles in multiple developmental processes in insects. Our previous study showed that CRISPR/Cas9-mediated knock down of the microRNA let-7 in silkworms increased the size of larvae and silk glands, thereby improving the silk production capacity. In this study, we elucidate the molecular mechanism underlying of let-7 regulates growth. Identification of differentially expressed genes in response to let-7 knock down revealed enrichment of pathways associated with cell proliferation and DNA replication. let-7 dysregulation affected the cell cycle and proliferation of the Bombyx mori cell line BmN. Dual-luciferase and target site mutation assays showed that BmCDK1 is a direct target gene of let-7, with only 1 binding site on its 3'-untranslated region. RNA interference of BmCDK1 inhibited cell proliferation, but this effect was counteracted by co-transfection with let-7 antagomir. Moreover, let-7 knock down induced BmCDK1 expression and promoted cell proliferation in multiple tissues, and further induced endomitosis in the silk gland in vivo. Knock down of BmCDK1 resulted in abnormal formation of a new epidermis, and larval development was arrested at the 2nd or 3rd molt stage. Taken together, our results demonstrated that BmCDK1 is a novel target of let-7 in cell fate determination, possessing potential for improving silk yield in silkworm.
Collapse
Affiliation(s)
- Wei Wang
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
- Engineering Laboratory of Sericultural and Functional Genome and Biotechnology, Development and Reform Commission, Chongqing, China
| | - Linshengzhe Ji
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Xinyuan Jing
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Ping Zhao
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
- Engineering Laboratory of Sericultural and Functional Genome and Biotechnology, Development and Reform Commission, Chongqing, China
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
- Engineering Laboratory of Sericultural and Functional Genome and Biotechnology, Development and Reform Commission, Chongqing, China
| |
Collapse
|
4
|
Szala D, Kopańska M, Trojniak J, Jabłoński J, Hanf-Osetek D, Snela S, Zawlik I. The Role of MicroRNAs in the Pathophysiology of Osteoarthritis. Int J Mol Sci 2024; 25:6352. [PMID: 38928059 PMCID: PMC11204066 DOI: 10.3390/ijms25126352] [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: 05/05/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Worldwide, osteoarthritis (OA) is the most common cause of joint pain in older people. Many factors contribute to osteoarthritis' development and progression, including secondary osteoarthritis' underlying causes. It is important to note that osteoarthritis affects all four tissues: cartilage, bone, joint capsule, and articular apparatus. An increasingly prominent area of research in osteoarthritis regulation is microRNAs (miRNAs), a small, single-stranded RNA molecule that controls gene expression in eukaryotes. We aimed to assess and summarize current knowledge about the mechanisms of the action of miRNAs and their clinical significance. Osteoarthritis (OA) is affected by the interaction between miRNAs and inflammatory processes, as well as cartilage metabolism. MiRNAs also influence cartilage cell apoptosis, contributing to the degradation of the cartilage in OA. Studies have shown that miRNAs may have both an inhibitory and promoting effect on osteoporosis progression through their influence on molecular mechanisms. By identifying these regulators, targeted treatments for osteoarthritis may be developed. In addition, microRNA may also serve as a biomarker for osteoarthritis. By using these biomarkers, the disease could be detected faster, and early intervention can be instituted to prevent mobility loss and slow deterioration.
Collapse
Affiliation(s)
| | - Marta Kopańska
- Department of Pathophysiology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Julia Trojniak
- Student Research Club “Reh-Tech”, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
| | - Jarosław Jabłoński
- Faculty of Orthopaedic and Reumatology, Institute of Medical Sciences, Collegium Medicum, University of Rzeszow, 35-959 Rzeszow, Poland; (J.J.); (D.H.-O.); (S.S.)
- Orthopaedics and Traumatology Clinic, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| | - Dorota Hanf-Osetek
- Faculty of Orthopaedic and Reumatology, Institute of Medical Sciences, Collegium Medicum, University of Rzeszow, 35-959 Rzeszow, Poland; (J.J.); (D.H.-O.); (S.S.)
- Orthopaedics and Traumatology Clinic, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| | - Sławomir Snela
- Faculty of Orthopaedic and Reumatology, Institute of Medical Sciences, Collegium Medicum, University of Rzeszow, 35-959 Rzeszow, Poland; (J.J.); (D.H.-O.); (S.S.)
- Orthopaedics and Traumatology Clinic, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| | - Izabela Zawlik
- Department of General Genetics, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland;
| |
Collapse
|
5
|
Akyüz B, Sohel MMH, Konca Y, Arslan K, Gürbulak K, Abay M, Kaliber M, White SN, Cinar MU. Effects of Low and High Maternal Protein Intake on Fetal Skeletal Muscle miRNAome in Sheep. Animals (Basel) 2024; 14:1594. [PMID: 38891641 PMCID: PMC11171157 DOI: 10.3390/ani14111594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Prenatal maternal feeding plays an important role in fetal development and has the potential to induce long-lasting epigenetic modifications. MicroRNAs (miRNAs) are non-coding, single-stranded RNAs that serve as one epigenetic mechanism. Though miRNAs have crucial roles in fetal programming, growth, and development, there is limited data regarding the maternal diet and miRNA expression in sheep. Therefore, we analyzed high and low maternal dietary protein for miRNA expression in fetal longissimus dorsi. Pregnant ewes were fed an isoenergetic high-protein (HP, 160-270 g/day), low-protein (LP, 73-112 g/day), or standard-protein diet (SP, 119-198 g/day) during pregnancy. miRNA expression profiles were evaluated using the Affymetrix GeneChip miRNA 4.0 Array. Twelve up-regulated, differentially expressed miRNAs (DE miRNAs) were identified which are targeting 65 genes. The oar-3957-5p miRNA was highly up-regulated in the LP and SP compared to the HP. Previous transcriptome analysis identified that integrin and non-receptor protein tyrosine phosphatase genes targeted by miRNAs were detected in the current experiment. A total of 28 GO terms and 10 pathway-based gene sets were significantly (padj < 0.05) enriched in the target genes. Most genes targeted by the identified miRNAs are involved in immune and muscle disease pathways. Our study demonstrated that dietary protein intake during pregnancy affected fetal skeletal muscle epigenetics via miRNA expression.
Collapse
Affiliation(s)
- Bilal Akyüz
- Department of Genetics, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (B.A.); (M.M.H.S.); (K.A.)
| | - Md Mahmodul Hasan Sohel
- Department of Genetics, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (B.A.); (M.M.H.S.); (K.A.)
- Genome and Stem Cell Centre, Erciyes University, Kayseri 38039, Türkiye
| | - Yusuf Konca
- Department of Animal Science, Faculty of Agriculture, Erciyes University, Kayseri 38039, Türkiye; (Y.K.); (M.K.)
| | - Korhan Arslan
- Department of Genetics, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (B.A.); (M.M.H.S.); (K.A.)
| | - Kutlay Gürbulak
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (K.G.); (M.A.)
| | - Murat Abay
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye; (K.G.); (M.A.)
| | - Mahmut Kaliber
- Department of Animal Science, Faculty of Agriculture, Erciyes University, Kayseri 38039, Türkiye; (Y.K.); (M.K.)
| | - Stephen N. White
- Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA 99164, USA;
| | - Mehmet Ulas Cinar
- Department of Animal Science, Faculty of Agriculture, Erciyes University, Kayseri 38039, Türkiye; (Y.K.); (M.K.)
- Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA 99164, USA;
| |
Collapse
|
6
|
Yao Q, He T, Liao JY, Liao R, Wu X, Lin L, Xiao G. Noncoding RNAs in skeletal development and disorders. Biol Res 2024; 57:16. [PMID: 38644509 PMCID: PMC11034114 DOI: 10.1186/s40659-024-00497-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 04/09/2024] [Indexed: 04/23/2024] Open
Abstract
Protein-encoding genes only constitute less than 2% of total human genomic sequences, and 98% of genetic information was previously referred to as "junk DNA". Meanwhile, non-coding RNAs (ncRNAs) consist of approximately 60% of the transcriptional output of human cells. Thousands of ncRNAs have been identified in recent decades, and their essential roles in the regulation of gene expression in diverse cellular pathways associated with fundamental cell processes, including proliferation, differentiation, apoptosis, and metabolism, have been extensively investigated. Furthermore, the gene regulation networks they form modulate gene expression in normal development and under pathological conditions. In this review, we integrate current information about the classification, biogenesis, and function of ncRNAs and how these ncRNAs support skeletal development through their regulation of critical genes and signaling pathways in vivo. We also summarize the updated knowledge of ncRNAs involved in common skeletal diseases and disorders, including but not limited to osteoporosis, osteoarthritis, rheumatoid arthritis, scoliosis, and intervertebral disc degeneration, by highlighting their roles established from in vivo, in vitro, and ex vivo studies.
Collapse
Affiliation(s)
- Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jian-You Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Rongdong Liao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lijun Lin
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
7
|
Hu P, Zhang G, Ba H, Ren J, Li J, Wang Z, Li C. Reciprocal negative feedback between Prrx1 and miR-140-3p regulates rapid chondrogenesis in the regenerating antler. Cell Mol Biol Lett 2024; 29:56. [PMID: 38643083 PMCID: PMC11031908 DOI: 10.1186/s11658-024-00573-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/05/2024] [Indexed: 04/22/2024] Open
Abstract
During growth phase, antlers exhibit a very rapid rate of chondrogenesis. The antler is formed from its growth center reserve mesenchyme (RM) cells, which have been found to be the derivatives of paired related homeobox 1 (Prrx1)-positive periosteal cells. However, the underlying mechanism that drives rapid chondrogenesis is not known. Herein, the miRNA expression profiles and chromatin states of three tissue layers (RM, precartilage, and cartilage) at different stages of differentiation within the antler growth center were analyzed by RNA-sequencing and ATAC-sequencing. We found that miR-140-3p was the miRNA that exhibited the greatest degree of upregulation in the rapidly growing antler, increasing from the RM to the cartilage layer. We also showed that Prrx1 was a key upstream regulator of miR-140-3p, which firmly confirmed by Prrx1 CUT&Tag sequencing of RM cells. Through multiple approaches (three-dimensional chondrogenic culture and xenogeneic antler model), we demonstrated that Prrx1 and miR-140-3p functioned as reciprocal negative feedback in the antler growth center, and downregulating PRRX1/upregulating miR-140-3p promoted rapid chondrogenesis of RM cells and xenogeneic antler. Thus, we conclude that the reciprocal negative feedback between Prrx1 and miR-140-3p is essential for balancing mesenchymal proliferation and chondrogenic differentiation in the regenerating antler. We further propose that the mechanism underlying chondrogenesis in the regenerating antler would provide a reference for helping understand the regulation of human cartilage regeneration and repair.
Collapse
Affiliation(s)
- Pengfei Hu
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China.
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Guokun Zhang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
| | - Hengxing Ba
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
| | - Jing Ren
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
| | - Jiping Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
| | - Zhen Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China.
| |
Collapse
|
8
|
Lui JC. Growth disorders caused by variants in epigenetic regulators: progress and prospects. Front Endocrinol (Lausanne) 2024; 15:1327378. [PMID: 38370361 PMCID: PMC10870149 DOI: 10.3389/fendo.2024.1327378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Epigenetic modifications play an important role in regulation of transcription and gene expression. The molecular machinery governing epigenetic modifications, also known as epigenetic regulators, include non-coding RNA, chromatin remodelers, and enzymes or proteins responsible for binding, reading, writing and erasing DNA and histone modifications. Recent advancement in human genetics and high throughput sequencing technology have allowed the identification of causative variants, many of which are epigenetic regulators, for a wide variety of childhood growth disorders that include skeletal dysplasias, idiopathic short stature, and generalized overgrowth syndromes. In this review, we highlight the connection between epigenetic modifications, genetic variants in epigenetic regulators and childhood growth disorders being established over the past decade, discuss their insights into skeletal biology, and the potential of epidrugs as a new type of therapeutic intervention.
Collapse
Affiliation(s)
- Julian C. Lui
- Section on Growth and Development, National Institute of Child Health and Human Development, Bethesda, MD, United States
| |
Collapse
|
9
|
Wells LM, Roberts HC, Luyten FP, Roberts SJ. Identifying Fibroblast Growth Factor Receptor 3 as a Mediator of Periosteal Osteochondral Differentiation through the Construction of microRNA-Based Interaction Networks. BIOLOGY 2023; 12:1381. [PMID: 37997980 PMCID: PMC10669632 DOI: 10.3390/biology12111381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023]
Abstract
Human periosteum-derived progenitor cells (hPDCs) have the ability to differentiate towards both the chondrogenic and osteogenic lineages. This coordinated and complex osteochondrogenic differentiation process permits endochondral ossification and is essential in bone development and repair. We have previously shown that humanised cultures of hPDCs enhance their osteochondrogenic potentials in vitro and in vivo; however, the underlying mechanisms are largely unknown. This study aimed to identify novel regulators of hPDC osteochondrogenic differentiation through the construction of miRNA-mRNA regulatory networks derived from hPDCs cultured in human serum or foetal bovine serum as an alternative in silico strategy to serum characterisation. Sixteen differentially expressed miRNAs (DEMis) were identified in the humanised culture. In silico analysis of the DEMis with TargetScan allowed for the identification of 1503 potential miRNA target genes. Upon comparison with a paired RNAseq dataset, a 4.5% overlap was observed (122 genes). A protein-protein interaction network created with STRING interestingly identified FGFR3 as a key network node, which was further predicted using multiple pathway analyses. Functional analysis revealed that hPDCs with the activating mutation FGFR3N540K displayed increased expressions of chondrogenic gene markers when cultured under chondrogenic conditions in vitro and displayed enhanced endochondral bone formation in vivo. A further histological analysis uncovered known downstream mediators involved in FGFR3 signalling and endochondral ossification to be upregulated in hPDC FGFR3N540K-seeded implants. This combinational approach of miRNA-mRNA-protein network analysis with in vitro and in vivo characterisation has permitted the identification of FGFR3 as a novel mediator of hPDC biology. Furthermore, this miRNA-based workflow may also allow for the identification of drug targets, which may be of relevance in instances of delayed fracture repair.
Collapse
Affiliation(s)
- Leah M. Wells
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London NW1 0TU, UK;
| | - Helen C. Roberts
- Department of Natural Sciences, Middlesex University, London NW4 4BT, UK;
| | - Frank P. Luyten
- Skeletal Biology and Engineering Research Centre (SBE), KU Leuven, 3000 Leuven, Belgium;
| | - Scott J. Roberts
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London NW1 0TU, UK;
| |
Collapse
|
10
|
Dalle Carbonare L, Minoia A, Braggio M, Bertacco J, Piritore FC, Zouari S, Vareschi A, Elia R, Vedovi E, Scumà C, Carlucci M, Bhandary L, Mottes M, Romanelli MG, Valenti MT. Modulation of miR-146b Expression during Aging and the Impact of Physical Activity on Its Expression and Chondrogenic Progenitors. Int J Mol Sci 2023; 24:13163. [PMID: 37685971 PMCID: PMC10488278 DOI: 10.3390/ijms241713163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The finding of molecules associated with aging is important for the prevention of chronic degenerative diseases and for longevity strategies. MicroRNAs (miRNAs) are post-transcriptional regulators involved in many biological processes and miR-146b-5p has been shown to be involved in different degenerative diseases. However, miR-146b-5p modulation has not been evaluated in mesenchymal stem cells (MSCs) commitment or during aging. Therefore, the modulation of miR-146b-5p in the commitment and differentiation of mesenchymal cells as well as during maturation and aging in zebrafish model were analyzed. In addition, circulating miR-146b-5p was evaluated in human subjects at different age ranges. Thus, the role of physical activity in the modulation of miR-146b-5p was also investigated. To achieve these aims, RT (real-time)-PCR, Western blot, cell transfections, and three-dimensional (3D) culture techniques were applied. Our findings show that miR-146b-5p expression drives MSCs to adipogenic differentiation and increases during zebrafish maturation and aging. In addition, miR-146b-5p expression is higher in females compared to males and it is associated with the aging in humans. Interestingly, we also observed that the physical activity of walking downregulates circulating miR-146b-5p levels in human females and increases the number of chondroprogenitors. In conclusion, miR-146b-5p can be considered an age-related marker and can represent a useful marker for identifying strategies, such as physical activity, aimed at counteracting the degenerative processes of aging.
Collapse
Affiliation(s)
- Luca Dalle Carbonare
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Arianna Minoia
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Michele Braggio
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Jessica Bertacco
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| | - Francesca Cristiana Piritore
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| | - Sharazed Zouari
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Anna Vareschi
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Rossella Elia
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Ermes Vedovi
- Recovery and Functional Rehabilitation, Integrated University Hospital of Verona, 37100 Verona, Italy; (E.V.); (C.S.)
| | - Cristina Scumà
- Recovery and Functional Rehabilitation, Integrated University Hospital of Verona, 37100 Verona, Italy; (E.V.); (C.S.)
| | - Matilde Carlucci
- Health Directorate, Integrated University Hospital of Verona, 37100 Verona, Italy;
| | | | - Monica Mottes
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| | - Maria Grazia Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| | - Maria Teresa Valenti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| |
Collapse
|
11
|
Xiang M, Liu L, Wu T, Wei B, Liu H. RNA-binding proteins in degenerative joint diseases: A systematic review. Ageing Res Rev 2023; 86:101870. [PMID: 36746279 DOI: 10.1016/j.arr.2023.101870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/12/2023] [Accepted: 01/27/2023] [Indexed: 02/07/2023]
Abstract
RNA-binding proteins (RBPs), which are conserved proteins comprising multiple intermediate sequences, can interact with proteins, messenger RNA (mRNA) of coding genes, and non-coding RNAs to perform different biological functions, such as the regulation of mRNA stability, selective polyadenylation, and the management of non-coding microRNA (miRNA) synthesis to affect downstream targets. This article will highlight the functions of RBPs, in degenerative joint diseases (intervertebral disc degeneration [IVDD] and osteoarthritis [OA]). It will reviews the latest advancements on the regulatory mechanism of RBPs in degenerative joint diseases, in order to understand the pathophysiology, early diagnosis and treatment of OA and IVDD from a new perspective.
Collapse
Affiliation(s)
- Min Xiang
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Ling Liu
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Tingrui Wu
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Bo Wei
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| | - Huan Liu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
12
|
Shi Y, Shao X, Sun M, Ma J, Li B, Zou N, Li F. MiR-140 is involved in T-2 toxin-induced matrix degradation of articular cartilage. Toxicon 2023; 222:106987. [PMID: 36462649 DOI: 10.1016/j.toxicon.2022.106987] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
T-2 toxin is one of the most toxic mycotoxins contaminating various grains. It is considered an environmental risk factor for Kashin-Beck disease (KBD), an endemic degenerative osteochondrosis. Currently, the underlying molecular mechanisms of articular cartilage damage caused by T-2 toxin have not been elucidated. Studies have shown that miR-140 is essential for cartilage formation, and extracellular matrix (EMC) synthesis and degradation. The objective of this study was to investigate the mechanism of miR-140 involvement in T-2 toxin-induced articular cartilage damage. Two treatment groups, each containing wild-type mice and miR-140 knockout mice were administered with T-2 toxin (200 ng/g BW/day) or a normal diet for 1 month, 3 months, and 6 months. Results showed that T-2 toxin caused articular cartilage and growth plate damage in mice. The expression of miR-140 decreased in articular cartilage of wild-type mice treated with T-2 toxin, and miR-140 deficiency aggravated T-2 toxin-induced knee cartilage damage. T-2 toxin-caused the reduction of miR-140 expression was consistent with collagen type II (COL2A1), aggrecan (ACAN), and SRY-box containing gene 9 (SOX9) and opposite to matrix metalloproteinase 13 (MMP13), a disintegrin and metalloproteinase with thrombospondin motif 5 (ADAMTS-5), and v-ral simian leukemia viral oncogene homolog A (RALA). In addition, we collected finger joints cartilage and knee joints cartilage from KBD patients and controls for paraffin embedding and sectioning. Results found that the expression of miR-140 in the articular cartilage of the KBD group was lower than that of the control group. The expression of COL2A1, ACAN, and SOX9 decreased, whereas ADAMTS-5, MMP13, and RALA increased in the articular cartilage of the KBD group. These results revealed that miR-140 might be involved in T-2 toxin-induced degradation of the ECM of articular cartilage. Moreover, the occurrence of KBD might be related to the decreased expression of miR-140 in articular cartilage.
Collapse
Affiliation(s)
- Yaning Shi
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission of the People's Republic of China (23618504), Harbin Medical University, Harbin, 150081, China
| | - Xinhua Shao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission of the People's Republic of China (23618504), Harbin Medical University, Harbin, 150081, China
| | - Mengyi Sun
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jing Ma
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission of the People's Republic of China (23618504), Harbin Medical University, Harbin, 150081, China
| | - Bingsu Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission of the People's Republic of China (23618504), Harbin Medical University, Harbin, 150081, China
| | - Ning Zou
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission of the People's Republic of China (23618504), Harbin Medical University, Harbin, 150081, China.
| | - Fuyuan Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission of the People's Republic of China (23618504), Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
13
|
Guo R, Zhuang H, Chen X, Ben Y, Fan M, Wang Y, Zheng P. Tissue engineering in growth plate cartilage regeneration: Mechanisms to therapeutic strategies. J Tissue Eng 2023; 14:20417314231187956. [PMID: 37483459 PMCID: PMC10359656 DOI: 10.1177/20417314231187956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/29/2023] [Indexed: 07/25/2023] Open
Abstract
The repair of growth plate injuries is a highly complex process that involves precise spatiotemporal regulation of multiple cell types. While significant progress has been made in understanding the pathological mechanisms underlying growth plate injuries, effectively regulating this process to regenerate the injured growth plate cartilage remains a challenge. Tissue engineering technology has emerged as a promising therapeutic approach for achieving tissue regeneration through the use of functional biological materials, seed cells and biological factors, and it is now widely applied to the regeneration of bone and cartilage. However, due to the unique structure and function of growth plate cartilage, distinct strategies are required for effective regeneration. Thus, this review provides an overview of current research on the application of tissue engineering to promote growth plate regeneration. It aims to elucidates the underlying mechanisms by which tissue engineering promotes growth plate regeneration and to provide novel insights and therapeutic strategies for future research on the regeneration of growth plate.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Pengfei Zheng
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Crossland RE, Albiero A, Sanjurjo‐Rodríguez C, Reis M, Resteu A, Anderson AE, Dickinson AM, Pratt AG, Birch M, McCaskie AW, Jones E, Wang X. MicroRNA profiling of low concentration extracellular vesicle RNA utilizing NanoString nCounter technology. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e72. [PMID: 38938446 PMCID: PMC11080777 DOI: 10.1002/jex2.72] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/04/2022] [Accepted: 12/21/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EV) and the microRNAs that they contain are increasingly recognised as a rich source of informative biomarkers, reflecting pathological processes and fundamental biological pathways and responses. Their presence in biofluids makes them particularly attractive for biomarker identification. However, a frequent caveat in relation to clinical studies is low abundance of EV RNA content. In this study, we used NanoString nCounter technology to assess the microRNA profiles of n = 64 EV low concentration RNA samples (180-49125 pg), isolated from serum and cell culture media using precipitation reagent or sequential ultracentrifugation. Data was subjected to robust quality control parameters based on three levels of limit of detection stringency, and differential microRNA expression analysis was performed between biological subgroups. We report that RNA concentrations > 100 times lower than the current NanoString recommendations can be successfully profiled using nCounter microRNA assays, demonstrating acceptable output ranges for imaging parameters, binding density, positive/negative controls, ligation controls and normalisation quality control. Furthermore, despite low levels of input RNA, high-level differential expression analysis between biological subgroups identified microRNAs of biological relevance. Our results demonstrate that NanoString nCounter technology offers a sensitive approach for the detection and profiling of low abundance EV-derived microRNA, and may provide a solution for research studies that focus on limited sample material.
Collapse
Affiliation(s)
- Rachel E. Crossland
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Anna Albiero
- Division of Trauma and Orthopaedic Surgery, Department of SurgeryUniversity of Cambridge Addenbrooke's HospitalCambridgeUK
| | - Clara Sanjurjo‐Rodríguez
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
- Physiotherapy, Medicine and Biomedical Sciences department, University of A Coruña; University Hospital Complex from A Coruña (Sergas, CHUACInstitute of Biomedical Research of A Coruña (INIBIC)‐Centre of Advanced Scientific Researches (CICA)A CoruñaSpain
| | - Monica Reis
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
- Centre for Regenerative Medicine, Institute for Regeneration and RepairThe University of Edinburgh, Edinburgh BioQuarterEdinburghUK
| | - Anastasia Resteu
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Amy E. Anderson
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Anne M. Dickinson
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Arthur G. Pratt
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
- Musculoskeletal Services DirectorateNewcastle upon Tyne Hospitals NHS Foundation TrustUK
| | - Mark Birch
- Division of Trauma and Orthopaedic Surgery, Department of SurgeryUniversity of Cambridge Addenbrooke's HospitalCambridgeUK
| | - Andrew W. McCaskie
- Division of Trauma and Orthopaedic Surgery, Department of SurgeryUniversity of Cambridge Addenbrooke's HospitalCambridgeUK
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal MedicineUniversity of LeedsLeedsUK
| | - Xiao‐nong Wang
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
15
|
Application of Bioinformatics Tools for the Prediction of Helper MicroRNAs for Improvement of Oncolytic Virus Efficacy. Cell Microbiol 2022. [DOI: 10.1155/2022/5756131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Purpose. Oncolytic Reoviruses, as a self-limiting virus, can be used in cancer treatment, because they have the ability to replicate in tumor cells selectively and destroy them. Studies show that some immune response proteins may interfere with the virus life cycle. So, the main aim of this bioinformatic study is to check which microRNA is able to target some reovirus inhibitory proteins. Experimental Design. By use of online bioinformatics software, the microRNAs that could target inhibitory genes were selected. Then, other features like content ++ score and cell type were checked and finally the eligible microRNAs were determined. Results. After choosing 15 inhibitory proteins, analysis was performed and finally 37 microRNAs which could target inhibitory proteins in colorectal cell lines were selected. In the end, by investigation of web-based tools, just two microRNAs were finalized. Conclusions and Clinical Relevance. This bioinformatic study shows that microRNA-140 and microRNA-92a have the potential to target some inhibitory proteins which interfere with oncolytic Reovirus replication and it may help in the optimal use of this virus as a cancer treatment. Because selective reproduction of Reovirus in tumor cells, as a nonchemical therapy, can be a good way to overcome this disease with broad advantages.
Collapse
|
16
|
Vechetti IJ, Norrbom J, Alkner B, Hjalmarsson E, Palmcrantz A, Pontén E, Pingel J, von Walden F, Fernandez-Gonzalo R. Extracellular vesicle characteristics and microRNA content in cerebral palsy and typically developed individuals at rest and in response to aerobic exercise. Front Physiol 2022; 13:1072040. [PMID: 36620222 PMCID: PMC9811128 DOI: 10.3389/fphys.2022.1072040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
In this study, the properties of circulating extracellular vesicles (EVs) were examined in cerebral palsy (CP) and typically developed (TD) individuals at rest and after aerobic exercise, focusing on the size, concentration, and microRNA cargo of EVs. Nine adult individuals with CP performed a single exercise bout consisting of 45 min of Frame Running, and TD participants completed either 45 min of cycling (n = 10; TD EX) or were enrolled as controls with no exercise (n = 10; TD CON). Blood was drawn before and 30 min after exercise and analyzed for EV concentration, size, and microRNA content. The size of EVs was similar in CP vs. TD, and exercise had no effect. Individuals with CP had an overall lower concentration (∼25%, p < 0.05) of EVs. At baseline, let-7a, let-7b and let-7e were downregulated in individuals with CP compared to TD (p < 0.05), while miR-100 expression was higher, and miR-877 and miR-4433 lower in CP compared to TD after exercise (p < 0.05). Interestingly, miR-486 was upregulated ∼2-fold in the EVs of CP vs. TD both at baseline and after exercise. We then performed an in silico analysis of miR-486 targets and identified the satellite cell stemness factor Pax7 as a target of miR-486. C2C12 myoblasts were cultured with a miR-486 mimetic and RNA-sequencing was performed. Gene enrichment analysis revealed that several genes involved in sarcomerogenesis and extracellular matrix (ECM) were downregulated. Our data suggest that circulating miR-486 transported by EVs is elevated in individuals with CP and that miR-486 alters the transcriptome of myoblasts affecting both ECM- and sarcomerogenesis-related genes, providing a link to the skeletal muscle alterations observed in individuals with CP.
Collapse
Affiliation(s)
- Ivan J. Vechetti
- Department of Nutrition and Health Sciences, College of Education and Human Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Jessica Norrbom
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Björn Alkner
- Department of Orthopaedics, Eksjö, Region Jönköping County and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Emma Hjalmarsson
- Division of Pediatric Neurology, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Alexandra Palmcrantz
- Division of Pediatric Neurology, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Eva Pontén
- Division of Pediatric Neurology, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden,Department of Pediatric Orthopedic Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Jessica Pingel
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Ferdinand von Walden
- Division of Pediatric Neurology, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo Fernandez-Gonzalo
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden,Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden,*Correspondence: Rodrigo Fernandez-Gonzalo,
| |
Collapse
|
17
|
Characterization of microRNA Levels in Synovial Fluid from Knee Osteoarthritis and Anterior Cruciate Ligament Tears. Biomedicines 2022; 10:biomedicines10112909. [PMID: 36428476 PMCID: PMC9687202 DOI: 10.3390/biomedicines10112909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/07/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
This study investigated modifications of microRNA expression profiles in knee synovial fluid of patients with osteoarthritis (OA) and rupture of the anterior cruciate ligament (ACL). Twelve microRNAs (26a-5p, 27a-3p, let7a-5p, 140-5p, 146-5p, 155-5p, 16-5p,186-5p, 199a-3p, 210-3p, 205-5p, and 30b-5p) were measured by real-time quantitative polymerase chain reaction (RT-qPCR) in synovial fluids obtained from 30 patients with ACL tear and 18 patients with knee OA. These 12 miRNAs were chosen on the basis of their involvement in pathological processes of bone and cartilage. Our results show that miR-26a-5p, miR-186-5p, and miR-30b-5p were expressed in the majority of OA and ACL tear samples, whereas miR-199a-3p, miR-210-3p, and miR-205-5p were detectable only in a few samples. Interestingly, miR-140-5p was expressed in only one sample of thirty in the ACL tear group. miR-140-5p has been proposed to modulate two genes (BGN and COL5A1100) that are involved in ligamentous homeostasis; their altered expression could be linked with ACL rupture susceptibility. The expression of miR-30b-5p was higher in OA and chronic ACL groups compared to acute ACL samples. We provide evidence that specific miRNAs could be detected not only in synovial fluid of patients with OA, but also in post-traumatic ACL tears.
Collapse
|
18
|
Jouan Y, Bouchemla Z, Bardèche-Trystram B, Sana J, Andrique C, Ea HK, Richette P, Latourte A, Cohen-Solal M, Hay E. Lin28a induces SOX9 and chondrocyte reprogramming via HMGA2 and blunts cartilage loss in mice. SCIENCE ADVANCES 2022; 8:eabn3106. [PMID: 36026443 PMCID: PMC9417174 DOI: 10.1126/sciadv.abn3106] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
Articular cartilage has low regenerative capacity despite permanent stress. Irreversible cartilage lesions characterize osteoarthritis (OA); this is not followed by tissue repair. Lin28a, an RNA binding protein, is detected in damaged cartilage in humans and mice. We investigated the role of LIN28a in cartilage physiology and in osteoarthritis. Lin28a-inducible conditional cartilage deletion up-regulated Mmp13 in intact mice and exacerbated the cartilage destruction in OA mice. Lin28a-specific cartilage overexpression protected mice against cartilage breakdown, stimulated chondrocyte proliferation and the expression of Prg4 and Sox9, and down-regulated Mmp13. Lin28a overexpression inhibited Let-7b and Let-7c miRNA levels while RNA-sequencing analysis revealed five genes of transcriptional factors regulated by Let-7. Moreover, Lin28a overexpression up-regulated HMGA2 and activated SOX9 transcription, a factor required for chondrocyte reprogramming. HMGA2 siRNA knockdown inhibited the cartilage protective effect of Lin28a overexpression. This study provides insights into a new pathway including the Lin28a-Let7 axis, thus promoting chondrocyte anabolism in injured cartilage in mice.
Collapse
Affiliation(s)
- Yohan Jouan
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | - Zohra Bouchemla
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | | | - Joanna Sana
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | - Caroline Andrique
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | - Hang-Korng Ea
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
- Hôpital Lariboisière, APHP, Paris, France
| | - Pascal Richette
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
- Hôpital Lariboisière, APHP, Paris, France
| | - Augustin Latourte
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | - Martine Cohen-Solal
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
- Hôpital Lariboisière, APHP, Paris, France
| | - Eric Hay
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| |
Collapse
|
19
|
Celik N, Kim MH, Yeo M, Kamal F, Hayes DJ, Ozbolat IT. miRNA induced 3D bioprinted-heterotypic osteochondral interface. Biofabrication 2022; 14:10.1088/1758-5090/ac7fbb. [PMID: 35803212 PMCID: PMC9588307 DOI: 10.1088/1758-5090/ac7fbb] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022]
Abstract
The engineering of osteochondral interfaces remains a challenge. MicroRNAs (miRs) have emerged as significant tools to regulate the differentiation and proliferation of osteogenic and chondrogenic formation in the human musculoskeletal system. Here, we describe a novel approach to osteochondral reconstruction based on the three-dimensional (3D) bioprinting of miR-transfected adipose-derived stem cell (ADSC) spheroids to produce a heterotypic interface that addresses the intrinsic limitations of the traditional approach to inducing zonal differentiation via the use of diffusible cytokines. We evaluated the delivery of miR-148b for osteogenic differentiation and the codelivery of miR-140 and miR-21 for the chondrogenic differentiation of ADSC spheroids. Our results demonstrated that miR-transfected ADSC spheroids exhibited upregulated expression of osteogenic and chondrogenic differentiation related gene and protein markers, and enhanced mineralization and cell proliferation compared to spheroids differentiated using a commercially-available differentiation medium. Upon confirmation of the osteogenic and chondrogenic potential of miR-transfected ADSC spheroids, using aspiration-assisted bioprinting, these spheroids were 3D bioprinted into a dual-layer heterotypic osteochondral interface with a stratified arrangement of distinct osteogenic and chondrogenic zones. The proposed approach holds great promise for the biofabrication of stratified tissues, not only for the osteochondral interfaces presented in this work, but also for other composite tissues and tissue interfaces, such as, but not limited to, the bone-tendon-muscle interface and craniofacial tissues.
Collapse
Affiliation(s)
- Nazmiye Celik
- Department of Engineering Science and Mechanics, Penn State University, 212 Earth-Engineering Sciences Bldg., University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Myoung Hwan Kim
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
- Department of Biomedical Engineering, Penn State University, Chemical and Biomedical Engineering Bldg., University Park, PA 16802, USA
| | - Miji Yeo
- Department of Engineering Science and Mechanics, Penn State University, 212 Earth-Engineering Sciences Bldg., University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Fadia Kamal
- Center for Orthopedic Research and Translational Sciences, Department of Orthopedics and Re-Habilitation, Penn State University, Hershey, PA 17033, USA
| | - Daniel J. Hayes
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
- Department of Biomedical Engineering, Penn State University, Chemical and Biomedical Engineering Bldg., University Park, PA 16802, USA
- Materials Research Institute, Penn State University, University Park, PA 16802, USA
| | - Ibrahim T. Ozbolat
- Department of Engineering Science and Mechanics, Penn State University, 212 Earth-Engineering Sciences Bldg., University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
- Department of Biomedical Engineering, Penn State University, Chemical and Biomedical Engineering Bldg., University Park, PA 16802, USA
- Materials Research Institute, Penn State University, University Park, PA 16802, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
20
|
Young C, Caffrey M, Janton C, Kobayashi T. Reversing the miRNA -5p/-3p stoichiometry reveals physiological roles and targets of miR-140 miRNAs. RNA (NEW YORK, N.Y.) 2022; 28:854-864. [PMID: 35332065 PMCID: PMC9074898 DOI: 10.1261/rna.079013.121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/10/2022] [Indexed: 06/03/2023]
Abstract
The chondrocyte-specific miR-140 miRNAs are necessary for normal endochondral bone growth in mice. miR-140 deficiency causes dwarfism and craniofacial deformity. However, the physiologically important targets of miR-140 miRNAs are still unclear. The miR-140 gene (Mir140) encodes three chondrocyte-specific microRNAs, miR-140-5p, derived from the 5' strand of primary miR-140, and miR140-3p.1 and -3p.2, derived from the 3' strand of primary miR-140. miR-140-3p miRNAs are 10 times more abundant than miR-140-5p likely due to the nonpreferential loading of miR-140-5p to Argonaute proteins. To differentiate the role of miR-140-5p and -3p miRNAs in endochondral bone development, two distinct mouse models, miR140-C > T, in which the first nucleotide of miR-140-5p was altered from cytosine to uridine, and miR140-CG, where the first two nucleotides of miR-140-3p were changed to cytosine and guanine, were created. These changes are expected to alter Argonaute protein loading preference of -5p and -3p to increase -5p loading and decrease -3p loading without changing the function of miR140-5p. These models presented a mild delay in epiphyseal development with delayed chondrocyte maturation. Using RNA-sequencing analysis of the two models, direct targets of miR140-5p, including Wnt11, were identified. Disruption of the predicted miR140-5p binding site in the 3' untranslated region of Wnt11 was shown to increase Wnt11 mRNA expression and caused a modest acceleration of epiphyseal development. These results show that the relative abundance of miRNA-5p and -3p can be altered by changing the first nucleotide of miRNAs in vivo, and this method can be useful to identify physiologically important miRNA targets.
Collapse
Affiliation(s)
- Cameron Young
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Melissa Caffrey
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Christopher Janton
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Tatsuya Kobayashi
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
21
|
Georgieva VS, Bluhm B, Probst K, Zhu M, Heilig J, Niehoff A, Brachvogel B. Ablation of the miRNA cluster 24 in cartilage and osteoblasts impairs bone remodeling. Sci Rep 2022; 12:9116. [PMID: 35650319 PMCID: PMC9160244 DOI: 10.1038/s41598-022-13231-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/13/2022] [Indexed: 11/28/2022] Open
Abstract
MicroRNAs (miRNAs) post-transcriptionally regulate cartilage and bone development and function, however, only few miRNAs have been described to play a role for cartilage to bone transition in vivo. Previously, we showed that cartilage-specific deletion of the Mirc24 cluster in newborn male mice leads to impaired growth plate cartilage development due to increased RAF/MEK/ERK signaling and affects the stability of the cartilage extracellular matrix on account of decreased SOX6 and SOX9 and increased MMP13 levels. Here, we studied how Mirc24 cluster inactivation in cartilage and osteoblasts leads to an increased bone density associated with defects in collagen remodeling in trabecular bone. No changes in osteoblast distribution were observed, whereas the number of osteoclasts was reduced and TRAP activity in osteoclasts decreased. Surprisingly, an increased level of cluster-encoded miR-322 or miR-503 raises Rankl gene expression and inactivation of the cluster in chondrocytes reduces Rankl expression. These results suggest that the Mirc24 cluster regulates Rankl expression in chondrocytes at the chondro-osseous border, where the cluster is mainly expressed to modulate osteoclast formation, bone remodeling and bone integrity.
Collapse
Affiliation(s)
- Veronika S Georgieva
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Björn Bluhm
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Kristina Probst
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Mengjie Zhu
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Juliane Heilig
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, 50933, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), University of Cologne, 50931, Cologne, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, 50933, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), University of Cologne, 50931, Cologne, Germany
| | - Bent Brachvogel
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany. .,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
22
|
Wang X, Geng B, Wang H, Wang S, Zhao D, He J, Lu F, An J, Wang C, Xia Y. Fluid shear stress-induced down-regulation of microRNA-140-5p promotes osteoblast proliferation by targeting VEGFA via the ERK5 pathway. Connect Tissue Res 2022; 63:156-168. [PMID: 33588662 DOI: 10.1080/03008207.2021.1891228] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Fluid shear stress (FSS) plays a critical role in osteoblast proliferation. However, the role of miRNA in osteoblast proliferation induced by FSS and the possible molecular mechanisms remain to be defined. The aim of the present study was to investigate whether miR-140-5p regulates osteoblast proliferation under FSS and its molecular mechanism. MATERIALS AND METHODS miR-140-5p expression was measured by qRT-PCR. Western blot was used to measure the expressions of P-ERK1/2, ERK1/2, P-ERK5 and ERK5. The levels of VEGFA, PCNA, CDK4 and Cyclin D1 were identified through qRT-PCR and western blot, respectively. Cell proliferation was detected by CCK-8 assay and EdU labeling assay. Dual-luciferase reporter assay was used to validate the target of miR-140-5p. RESULTS miR-140-5p was significantly down-regulated when MC3T3-E1 cells were exposed to FSS. We then confirmed that up-regulation of miR-140-5p inhibited and down-regulation of miR-140-5p promoted osteoblast proliferation. In addition, FSS promotes osteoblast proliferation via down-regulating miR-140-5p. Luciferase reporter assay demonstrated that VEGFA is a direct target of miR-140-5p. Furthermore, transfection of mimic-140-5p inhibited the up-regulation of VEGFA protein level induced by FSS, suggesting that FSS regulates VEGFA protein expression via miR-140-5p. Further investigations demonstrated that VEGFA could promote osteoblast proliferation. Lastly, we demonstrated that miR-140-5p regulates osteoblast proliferation and ERK5 activation through VEGFA. CONCLUSIONS Our study demonstrates that FSS-induced the down-regulation of miR-140-5p promotes osteoblast proliferation through activing VEGFA/ERK5 signaling pathway. These findings may provide a novel mechanism of FSS-induced osteoblast proliferation and offer a new avenue to further investigate osteogenesis induced by mechanical loading.
Collapse
Affiliation(s)
- Xingwen Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Hong Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Shenghong Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Dacheng Zhao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Jinwen He
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Fan Lu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Jiangdong An
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Cuifang Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
23
|
Zhang D, Niu S, Ma Y, Chen H, Wen Y, Li M, Zhou B, Deng Y, Shi C, Pu G, Yang M, Wang X, Zou C, Chen Y, Ma L. Fenofibrate Improves Insulin Resistance and Hepatic Steatosis and Regulates the Let-7/SERCA2b Axis in High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease Mice. Front Pharmacol 2022; 12:770652. [PMID: 35126113 PMCID: PMC8807641 DOI: 10.3389/fphar.2021.770652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022] Open
Abstract
Fenofibrate is widely used in clinical therapy to effectively ameliorate the development of non-alcoholic fatty liver disease (NAFLD); however, its specific molecular mechanism of action remains largely unknown. MicroRNAs (miRNAs) are key mediators in regulating endoplasmic reticulum (ER) stress during NAFLD, and the deregulation of miRNAs has been demonstrated in NAFLD pathophysiology. The present study aimed to identify whether fenofibrate could influence miRNA expression in NAFLD and investigate the specific mechanism of action of fenofibrate in lipid metabolism disorder-associated diseases. We found that fenofibrate alleviated ER stress and increased the levels of SERCA2b, which serves as a regulator of ER stress. Additionally, the levels of let-7 miRNA were regulated by fenofibrate; let-7 was found to target the 3′ untranslated region of SERCA2b. The present data suggest that the protective effects of fenofibrate against insulin resistance and its suppressive activity against excessive hepatic lipid accumulation may be related to the alteration of the let-7/SERCA2b axis and alleviation of ER stress.
Collapse
Affiliation(s)
- Dan Zhang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Shanzhuang Niu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Yicheng Ma
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Hang Chen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Yu Wen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Mingke Li
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Bo Zhou
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Yi Deng
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Chunjing Shi
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Guangyu Pu
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Meng Yang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Xianmei Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Chenggang Zou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Yuanli Chen
- Faculty of Basic Medicine, Kunming Medical University, Kunming, China
- *Correspondence: Yuanli Chen, ; Lanqing Ma,
| | - Lanqing Ma
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
- *Correspondence: Yuanli Chen, ; Lanqing Ma,
| |
Collapse
|
24
|
Overexpression of Lin28A in neural progenitor cells in vivo does not lead to brain tumor formation but results in reduced spine density. Acta Neuropathol Commun 2021; 9:185. [PMID: 34801069 PMCID: PMC8606090 DOI: 10.1186/s40478-021-01289-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/02/2021] [Indexed: 11/10/2022] Open
Abstract
LIN28A overexpression has been identified in malignant brain tumors called embryonal tumors with multilayered rosettes (ETMR) but its specific role during brain development remains largely unknown. Radial glia cells of the ventricular zone (VZ) are proposed as a cell of origin for ETMR. We asked whether an overexpression of LIN28A in such cells might affect brain development or result in the formation of brain tumors.Constitutive overexpression of LIN28A in hGFAP-cre::lsl-Lin28A (GL) mice led to a transient increase of proliferation in the cortical VZ at embryonic stages but no postnatal brain tumor formation. Postnatally, GL mice displayed a pyramidal cell layer dispersion of the hippocampus and altered spine and dendrite morphology, including reduced dendritic spine densities in the hippocampus and cortex. GL mice displayed hyperkinetic activity and differential quantitative MS-based proteomics revealed altered time dependent molecular functions regarding mRNA processing and spine morphogenesis. Phosphoproteomic analyses indicated a downregulation of mTOR pathway modulated proteins such as Map1b being involved in microtubule dynamics.In conclusion, we show that Lin28A overexpression transiently increases proliferation of neural precursor cells but it is not sufficient to drive brain tumors in vivo. In contrast, Lin28A impacts on protein abundancy patterns related to spine morphogenesis and phosphorylation levels of proteins involved in microtubule dynamics, resulting in decreased spine densities of neurons in the hippocampus and cortex as well as in altered behavior. Our work provides new insights into the role of LIN28A for neuronal morphogenesis and development and may reveal future targets for treatment of ETMR patients.
Collapse
|
25
|
Cirillo F, Catellani C, Lazzeroni P, Sartori C, Street ME. The Role of MicroRNAs in Influencing Body Growth and Development. Horm Res Paediatr 2021; 93:7-15. [PMID: 31914447 DOI: 10.1159/000504669] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/08/2019] [Indexed: 11/19/2022] Open
Abstract
Body growth and development are regulated among others by genetic and epigenetic factors. MicroRNAs (miRNAs) are epigenetic regulators of gene expression that act at the post-transcriptional level, thereby exerting a strong influence on regulatory gene networks. Increasing studies suggest the importance of miRNAs in the regulation of the growth plate and growth hormone (GH)-insulin-like growth factor (IGF) axis during the life course in a broad spectrum of animal species, contributing to longitudinal growth. This review summarizes the role of miRNAs in regulating growth in different in vitro and in vivo models acting on GH, GH receptor (GHR), IGFs, and IGF1R genes besides current knowledge in humans, and highlights that this regulatory system is of importance for growth.
Collapse
Affiliation(s)
- Francesca Cirillo
- Department of Mother and Child, Azienda USL - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Cecilia Catellani
- Department of Mother and Child, Azienda USL - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Pietro Lazzeroni
- Department of Mother and Child, Azienda USL - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Chiara Sartori
- Department of Mother and Child, Azienda USL - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Maria Elisabeth Street
- Department of Mother and Child, Azienda USL - IRCCS di Reggio Emilia, Reggio Emilia, Italy,
| |
Collapse
|
26
|
Shvedova M, Kobayashi T. MicroRNAs in cartilage development and dysplasia. Bone 2020; 140:115564. [PMID: 32745689 PMCID: PMC7502492 DOI: 10.1016/j.bone.2020.115564] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022]
Abstract
Small regulatory microRNAs (miRNAs) post-transcriptionally suppress gene expression. MiRNAs expressed in skeletal progenitor cells and chondrocytes regulate diverse aspects of cellular function and thus skeletal development. In this review, we discuss the role of miRNAs in skeletal development, particularly focusing on those whose physiological roles were revealed in vivo. Deregulation of miRNAs is found in multiple acquired diseases such as cancer; however congenital diseases caused by mutations in miRNA genes are very rare. Among those are mutations in miR-140 and miR-17~92 miRNAs which cause skeletal dysplasias. We also discuss pathological mechanisms underlining these skeletal dysplasias.
Collapse
Affiliation(s)
- Maria Shvedova
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tatsuya Kobayashi
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Sato T, Kataoka K, Ito Y, Yokoyama S, Inui M, Mori M, Takahashi S, Akita K, Takada S, Ueno-Kudoh H, Asahara H. Lin28a/let-7 pathway modulates the Hox code via Polycomb regulation during axial patterning in vertebrates. eLife 2020; 9:53608. [PMID: 32479258 PMCID: PMC7259951 DOI: 10.7554/elife.53608] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 05/18/2020] [Indexed: 01/06/2023] Open
Abstract
The body plan along the anteroposterior axis and regional identities are specified by the spatiotemporal expression of Hox genes. Multistep controls are required for their unique expression patterns; however, the molecular mechanisms behind the tight control of Hox genes are not fully understood. In this study, we demonstrated that the Lin28a/let-7 pathway is critical for axial elongation. Lin28a–/– mice exhibited axial shortening with mild skeletal transformations of vertebrae, which were consistent with results in mice with tail bud-specific mutants of Lin28a. The accumulation of let-7 in Lin28a–/– mice resulted in the reduction of PRC1 occupancy at the Hox cluster loci by targeting Cbx2. Consistently, Lin28a loss in embryonic stem-like cells led to aberrant induction of posterior Hox genes, which was rescued by the knockdown of let-7. These results suggest that the Lin28/let-7 pathway is involved in the modulation of the ‘Hox code’ via Polycomb regulation during axial patterning.
Collapse
Affiliation(s)
- Tempei Sato
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Kensuke Kataoka
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yoshiaki Ito
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Research Core, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigetoshi Yokoyama
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,Laboratory of Metabolism, National Institutes of Health, Bethesda, United States
| | - Masafumi Inui
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,Laboratory of Animal Regeneration Systemology, Meiji University, Kanagawa, Japan
| | - Masaki Mori
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Medical Chemistry, Shiga University of Medical Science, Shiga, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
| | - Keiichi Akita
- Department of Clinical Anatomy, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hiroe Ueno-Kudoh
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,Reproduction Center, Yokohama City University, Yokohama, Japan
| | - Hiroshi Asahara
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo, Japan.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States
| |
Collapse
|
28
|
Choi SY, Han EC, Hong SH, Kwon TG, Lee Y, Lee HJ. Regulating Osteogenic Differentiation by Suppression of Exosomal MicroRNAs. Tissue Eng Part A 2020; 25:1146-1154. [PMID: 30520703 DOI: 10.1089/ten.tea.2018.0257] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPACT STATEMENT We investigated the role of exosomes in osteogenesis and the use of miRNA inhibitor-transfected exosomes to control osteogenic differentiation. RNA-sequencing (RNA-seq) of exosomal miRNAs revealed that growth condition of milieu of preosteoblast exosomes harbors high levels of let-7, which plays a critical role in osteogenesis regulation. We modified exosomes by transfecting let-7 inhibitor into exosomes under growth condition in MC3T3-E1 cells and revealed that exosomes whose let-7 was inactivated by engineering lost the ability to recover osteogenic differentiation. Genetically modified exosomes may serve as powerful biomaterials for developmental control, including of osteogenesis regulation.
Collapse
Affiliation(s)
- Song-Yi Choi
- 1Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Eun-Chong Han
- 1Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Su-Hyung Hong
- 1Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Tae-Geon Kwon
- 2Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Youngkyun Lee
- 3Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Heon-Jin Lee
- 1Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, Korea.,4Brain Science and Engineering Institute, Kyungpook National University, Daegu, Korea
| |
Collapse
|
29
|
Ahkin Chin Tai JK, Freeman JL. Zebrafish as an integrative vertebrate model to identify miRNA mechanisms regulating toxicity. Toxicol Rep 2020; 7:559-570. [PMID: 32373477 PMCID: PMC7195498 DOI: 10.1016/j.toxrep.2020.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
Zebrafish are an established vertebrate model for toxicity studies. Zebrafish have a fully sequenced genome and the capability to create genetic models. Zebrafish have over 80 % homology for genes related to human disease. Functions of miRNAs in the zebrafish genome are being characterized. Zebrafish are ideal for mechanistic studies on how miRNAs regulate toxicity.
Zebrafish (Danio rerio) are an integrative vertebrate model ideal for toxicity studies. The zebrafish genome is sequenced with detailed characterization of all life stages. With their genetic similarity to humans, zebrafish models are established to study biological processes including development and disease mechanisms for translation to human health. The zebrafish genome, similar to other eukaryotic organisms, contains microRNAs (miRNAs) which function along with other epigenetic mechanisms to regulate gene expression. Studies have now established that exposure to toxins and xenobiotics can change miRNA expression profiles resulting in various physiological and behavioral alterations. In this review, we cover the intersection of miRNA alterations from toxin or xenobiotic exposure with a focus on studies using the zebrafish model system to identify miRNA mechanisms regulating toxicity. Studies to date have addressed exposures to toxins, particulate matter and nanoparticles, various environmental contaminants including pesticides, ethanol, and pharmaceuticals. Current limitations of the completed studies and future directions for this research area are discussed.
Collapse
Affiliation(s)
| | - Jennifer L Freeman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907 USA
| |
Collapse
|
30
|
Liu G, Lu T, Li Y, Liu Y, Ji X, Jia W, Sun M, Luo Y. Exosomal proteome from the serum, bone marrow, and palm and toe pustular skin tissues of a single patient with SAPHO syndrome. J Proteomics 2020; 216:103673. [PMID: 32001333 DOI: 10.1016/j.jprot.2020.103673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 01/10/2020] [Accepted: 01/26/2020] [Indexed: 12/24/2022]
Abstract
Exosome proteomic analysis may reveal differentially abundant proteins that are of significance for clarifying the pathogenesis of SAPHO (Synovitis, Acne, Pustulosis, Hyperostosis and Osteitis) syndrome. Exosomes were isolated from the serum, bone marrow and skin tissue of the palm and toe pustular areas in a unique patient with SAPHO syndrome. The exosomes were not different from those of healthy subjects in size (114.1 ± 73.7 nm) or morphology. Label-free exosome proteomic analysis identified 198 more abundant proteins and 183 less abundant compared with those of healthy subjects. Gene ontology enrichment analysis revealed that these proteins were involved in binding with a variety of biological molecules and participated in biological processes related to autoimmunity or inflammation. A total of 243 KEGG (Kyoto Encyclopedia of Gene and Genomes) pathways were enriched, of which 43 were related to immune function. It was speculated that five differentially abundant proteins, Mitogen-activated protein kinase 1 (MAPK1/MK01), Tyrosine protein kinase (SYK), Integrin beta-3 (ITB3), Serine/threonine-protein phosphatase 2a catalytic subunit alpha isoform (PP2AA) and Serine/threonine-protein phosphatase 2a 65 kDa regulatory subunit A beta isoform (2AAB), associated with multiple KEGG pathways, forms an interaction network that may be involved in the occurrence, development and prognosis of SAPHO syndrome. SIGNIFICANCE: Exosomes of SAPHO syndrome patient were not significantly different from those of healthy subjects in size and morphology. Label-free proteomic analysis of exosomal proteins in patient with SAPHO syndrome speculated 5 proteins MAPK1, SYK, ITB3, PP2AA and 2AAB, which may be involved in the occurrence, development and prognosis of SAPHO syndrome by binding with other biological molecules. It is speculated for the first time that proteins Histone H2A type 1-J and Histone H4 were related to SAPHO syndrome. Clinic relevance. Exosome proteomics can suggest novel pathological data in patients with SAPHO.
Collapse
Affiliation(s)
- Guomin Liu
- Jilin Provincial Changbai Mountain Anti-tumor Medicine Engineering Center, Changchun, Jilin, China; Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin 130041, China.
| | - Tiancheng Lu
- Jilin Provincial Changbai Mountain Anti-tumor Medicine Engineering Center, Changchun, Jilin, China; Life Sciences College, Jilin Agricultural University, Changchun, Jilin 130118, China.
| | - Yalong Li
- Jilin Provincial Changbai Mountain Anti-tumor Medicine Engineering Center, Changchun, Jilin, China; Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin 130041, China.
| | - Yun Liu
- Department of Stomatology, The Second Hospital of Jilin University, Changchun, Jilin 130041, China; Jilin Provincial Changbai Mountain Anti-tumor Medicine Engineering Center, Changchun, Jilin, China.
| | - Xuan Ji
- Department of Stomatology, The Second Hospital of Jilin University, Changchun, Jilin 130041, China; Jilin Provincial Changbai Mountain Anti-tumor Medicine Engineering Center, Changchun, Jilin, China.
| | - Wenyuan Jia
- Jilin Provincial Changbai Mountain Anti-tumor Medicine Engineering Center, Changchun, Jilin, China; Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin 130041, China.
| | - Maolei Sun
- Department of Stomatology, The Second Hospital of Jilin University, Changchun, Jilin 130041, China; Jilin Provincial Changbai Mountain Anti-tumor Medicine Engineering Center, Changchun, Jilin, China.
| | - Yungang Luo
- Department of Stomatology, The Second Hospital of Jilin University, Changchun, Jilin 130041, China; Jilin Provincial Changbai Mountain Anti-tumor Medicine Engineering Center, Changchun, Jilin, China.
| |
Collapse
|
31
|
Gao L, Wu D, Wu Y, Yang Z, Sheng J, Lin X, Huang H. MiR-3940-5p promotes granulosa cell proliferation through targeting KCNA5 in polycystic ovarian syndrome. Biochem Biophys Res Commun 2020; 524:791-797. [PMID: 32019676 DOI: 10.1016/j.bbrc.2020.01.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 01/07/2020] [Indexed: 12/19/2022]
Abstract
Increased granulosa cell (GC) proliferation may contribute to abnormal folliculogenesis in patients with polycystic ovary syndrome (PCOS), which affects approximately 10% reproductive aged women. However, the mechanisms underlying increased GC proliferation in PCOS remain incompletely understood. In this study, we identified miR-3940-5p as a hub miRNA in GC from PCOS using weighted gene co-expression network analysis (WGCNA), and real-time polymerase chain reaction (RT-PCR) analysis confirmed that miR-3940-5p was significantly increased in GC from PCOS. Enrichment analysis of predicted target genes of miR-3940-5p indicated potential roles of miR-3940-5p in follicular development and cell proliferation regulation. Consistently, functional study confirmed that miR-3940-5p overexpression promoted granulosa cell proliferation. Integrated analysis of mRNA expression profiling data and predicted target genes of miR-3940-5p identified potassium voltage-gated channel subfamily A member 5 (KCNA5) as a potential target of miR-3940-5p, and was validated by luciferase reporter assay. Finally, functional analysis suggested that miR-3940-5p promoted GC proliferation in a KCNA5 dependent manner. In conclusion, miR-3940-5p was a hub miRNA upregulated in GC from PCOS, and promoted GC proliferation by targeting KCNA5.
Collapse
Affiliation(s)
- Ling Gao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Dandan Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Yanting Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Zuwei Yang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Jianzhong Sheng
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xianhua Lin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| | - Hefeng Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| |
Collapse
|
32
|
Yapijakis C. Regulatory Role of MicroRNAs in Brain Development and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1195:237-247. [PMID: 32468482 DOI: 10.1007/978-3-030-32633-3_32] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules of about 20-22 nucleotides. After their posttranscriptional maturation, miRNAs are loaded into the ribonucleoprotein complex RISC and modulate gene expression by binding to the 3' untranslated region of their target mRNAs through base-pairing, which in turn triggers mRNA degradation or translational inhibition. There is mounting evidence that miRNAs regulate various biological processes, including cell proliferation, differentiation, and apoptosis. Several studies have shown that miRNAs play an important role in neurogenesis and brain development.This review discusses recent progress on understanding the implication of precisely regulated miRNA expression in normal brain development and function. In addition, it reports known cases of dysregulation of miRNA expression and function implicated in the pathogenesis of neurodevelopmental disorders, craniofacial dysmorphic syndromes, neurodegenerative diseases, and psychiatric disorders. Current knowledge regarding the role of miRNAs in the brain in conjunction with the complex interplay between genetic and epigenetic factors are discussed.
Collapse
Affiliation(s)
- Christos Yapijakis
- 1st Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "Haghia Sophia" Hospital, Athens, Greece.
- Department of Molecular Genetics, Cephalogenetics Diagnostic Center, Athens, Greece.
| |
Collapse
|
33
|
Wang W, Wang X, Li X, Pu Q, Luo C, Xu L, Peng X, Liu S. Genetic Manipulation of MicroRNAs in the Silk Gland of Silkworm, Bombyx Mori. Biol Proced Online 2019; 21:16. [PMID: 31427900 PMCID: PMC6694536 DOI: 10.1186/s12575-019-0102-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/07/2019] [Indexed: 02/07/2023] Open
Abstract
Background MicroRNAs (miRNAs) are a class of non-coding RNAs with important post-transcriptional regulatory functions. To reveal the function of miRNAs in vivo, the critical step is to change their expression levels in the tissues or organs. In this work, we explored the application of several important genetic techniques in altering the expression of silk gland-specific miR-274 of silkworm (Bombyx mori). Results Injection of synthesized microRNA mimics and antagomirs exerted no effect on the expression of miR-274 in the silk gland, miR-274 sponge specifically absorbed miR-274 and down-regulated its expression, transgenic overexpression of miR-274 precursor significantly up-regulated miR-274, and finally tissue-specific CRISPR/Cas9 system achieved deletion of miR-274. Conclusions A practical technical system was established for studying the functions of miRNAs in silk gland of Bombyx mori. Our research provides methodological support for the functional study of miRNAs and other noncoding RNAs in the silk gland and more organs in other species.
Collapse
Affiliation(s)
- Wei Wang
- 1State Key Laboratory of Silkworm Genome Biology,Biological Science Research Center, Southwest University, Chongqing, 400715 People's Republic of China.,2Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715 People's Republic of China
| | - Xinran Wang
- 1State Key Laboratory of Silkworm Genome Biology,Biological Science Research Center, Southwest University, Chongqing, 400715 People's Republic of China.,2Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715 People's Republic of China
| | - Xuemei Li
- 1State Key Laboratory of Silkworm Genome Biology,Biological Science Research Center, Southwest University, Chongqing, 400715 People's Republic of China.,2Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715 People's Republic of China
| | - Qian Pu
- 1State Key Laboratory of Silkworm Genome Biology,Biological Science Research Center, Southwest University, Chongqing, 400715 People's Republic of China.,2Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715 People's Republic of China
| | - Chengyi Luo
- 1State Key Laboratory of Silkworm Genome Biology,Biological Science Research Center, Southwest University, Chongqing, 400715 People's Republic of China.,2Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715 People's Republic of China
| | - Lili Xu
- 1State Key Laboratory of Silkworm Genome Biology,Biological Science Research Center, Southwest University, Chongqing, 400715 People's Republic of China.,2Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715 People's Republic of China
| | - Xinyue Peng
- 1State Key Laboratory of Silkworm Genome Biology,Biological Science Research Center, Southwest University, Chongqing, 400715 People's Republic of China.,2Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715 People's Republic of China
| | - Shiping Liu
- 1State Key Laboratory of Silkworm Genome Biology,Biological Science Research Center, Southwest University, Chongqing, 400715 People's Republic of China.,2Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715 People's Republic of China
| |
Collapse
|
34
|
miR-26a-5p is a Stable Reference Gene for miRNA Studies in Chondrocytes from Developing Human Cartilage. Cells 2019; 8:cells8060631. [PMID: 31234552 PMCID: PMC6627695 DOI: 10.3390/cells8060631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
miRNAs are emerging as key regulators of complex biological systems in several developmental processes. qRT-PCR is a powerful tool to quantitatively assess the profiles and modulation of miRNA expression. In the emerging field of cartilage maturation studies, from precursor to hypertrophic chondrocytes, few data about miRNA regulation are available, and no consensus on the best reference gene (RG) has been reached. This is a crucial pitfall since reliable outcomes depend on proper data normalization. The aim of this work was to identify reliable and stable miRNA RGs, basing the analysis on available high throughput qRT-PCR miRNA data (from the NCBI Gene Expression Omnibus database, GSE49152) obtained from human embryonic cartilage tissues enriched in the precursor, differentiated, and hypertrophic chondrocytes. Four normalization approaches were used, and the stability was quantified by combining BestKeeper, delta-Ct, geNorm, and NormFinder statistical tools. An integrated approach allowed to identify miR-26a-5p as the most stable RG and miR-212-3p as the worst one. RNU44, used in original dataset analysis, performed as second best RG. Applications of different normalization strategies significantly impacted the profiles and modulation of miRNA expression. Herein presented results point out the crucial need of a consensus on data normalization studies aimed at dissecting miRNA role in human cartilage development, to avoid the postulation of unreliable biological conclusions.
Collapse
|
35
|
Bai M, Yin H, Zhao J, Li Y, Wu Y. miR-182-5p overexpression inhibits chondrogenesis by down-regulating PTHLH. Cell Biol Int 2019; 43:222-232. [PMID: 30095215 DOI: 10.1002/cbin.11047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 08/05/2018] [Indexed: 12/21/2022]
Abstract
Human bone marrow mesenchymal stem cells (hBM-MSC) have the ability of differentiating into chondrocytes and osteoblasts. miR-182-5p promotes osteoclastogenesis and bone metastasis by up-regulating the expression of parathyroid hormone-like hormone (PTHLH). However, the function of miR-182-5p in chondrogenesis is still unknown. Mimic or inhibitor of miR-182-5p was used to upregulate or knock-down miR-182-5p expression, respectively. We analyzed chondrogenesis by Safranin O staining and Blyscan™ Sulfated Glycosaminoglycan Assay. Immunohistochemistry, real-time PCR, and Western bolts were used to detect related makers. miR-182-5p overexpression inhibited chondrogenesis. Dual-luciferase reporter assay indicated that PTHLH was one of the target genes of miR-182-5p. Further studies showed that miR-182-5p overexpression down-regulated the expression of SOX-9 and COL2A1, but up-regulated COL1A1 and COL10A1. Consistently, miR-182-5p knock-down had the opposite effects. This effect of miR-182-5p in BM-MSCs can be rescued by PTHLH overexpression. miR-182-5p may play a negative role in chondrogenesis by down-regulating PTHLH.
Collapse
Affiliation(s)
- Ming Bai
- Department of Minimally Invasive Spine Surgery, The Second Affiliated Hospital of Inner Mongolia Medical University, NO 1, Yingfang Road, Hohhot, Inner Mongolia 010000, P. R. China
| | - Heping Yin
- Department of Minimally Invasive Spine Surgery, The Second Affiliated Hospital of Inner Mongolia Medical University, NO 1, Yingfang Road, Hohhot, Inner Mongolia 010000, P. R. China
| | - Jian Zhao
- Department of Minimally Invasive Spine Surgery, The Second Affiliated Hospital of Inner Mongolia Medical University, NO 1, Yingfang Road, Hohhot, Inner Mongolia 010000, P. R. China
| | - Yang Li
- Department of Minimally Invasive Spine Surgery, The Second Affiliated Hospital of Inner Mongolia Medical University, NO 1, Yingfang Road, Hohhot, Inner Mongolia 010000, P. R. China
| | - Yimin Wu
- Department of Minimally Invasive Spine Surgery, The Second Affiliated Hospital of Inner Mongolia Medical University, NO 1, Yingfang Road, Hohhot, Inner Mongolia 010000, P. R. China
| |
Collapse
|
36
|
Rux D, Decker RS, Koyama E, Pacifici M. Joints in the appendicular skeleton: Developmental mechanisms and evolutionary influences. Curr Top Dev Biol 2018; 133:119-151. [PMID: 30902250 PMCID: PMC6988388 DOI: 10.1016/bs.ctdb.2018.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The joints are a diverse group of skeletal structures, and their genesis, morphogenesis, and acquisition of specialized tissues have intrigued biologists for decades. Here we review past and recent studies on important aspects of joint development, including the roles of the interzone and morphogenesis of articular cartilage. Studies have documented the requirement of interzone cells in limb joint initiation and formation of most, if not all, joint tissues. We highlight these studies and also report more detailed interzone dissection experiments in chick embryos. Articular cartilage has always received special attention owing to its complex architecture and phenotype and its importance in long-term joint function. We pay particular attention to mechanisms by which neonatal articular cartilage grows and thickens over time and eventually acquires its multi-zone structure and becomes mechanically fit in adults. These and other studies are placed in the context of evolutionary biology, specifically regarding the dramatic changes in limb joint organization during transition from aquatic to land life. We describe previous studies, and include new data, on the knee joints of aquatic axolotls that unlike those in higher vertebrates, are not cavitated, are filled with rigid fibrous tissues and resemble amphiarthroses. We show that when axolotls metamorph to life on land, their intra-knee fibrous tissue becomes sparse and seemingly more flexible and the articular cartilage becomes distinct and acquires a tidemark. In sum, there have been considerable advances toward a better understanding of limb joint development, biological responsiveness, and evolutionary influences, though much remains unclear. Future progress in these fields should also lead to creation of new developmental biology-based tools to repair and regenerate joint tissues in acute and chronic conditions.
Collapse
Affiliation(s)
- Danielle Rux
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, United States.
| | - Rebekah S Decker
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
37
|
|
38
|
Kobayashi T, Kozlova A. Lin28a overexpression reveals the role of Erk signaling in articular cartilage development. Development 2018; 145:dev.162594. [PMID: 30042178 DOI: 10.1242/dev.162594] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/08/2018] [Indexed: 12/12/2022]
Abstract
Adult articular cartilage shows limited tissue turnover, and therefore development of the proper structure of articular cartilage is crucial for life-long joint function. However, the mechanism by which the articular cartilage structure is developmentally regulated is poorly understood. In this study, we show evidence that activation of extracellular signal-regulated kinases (Erk1/2) in articular chondrocyte progenitors during developmental stages control articular cartilage thickness. We found that overexpression of Lin28a, an RNA-binding protein that regulates organismal growth and metabolism, in articular chondrocyte progenitor cells upregulated Erk signaling and increased articular cartilage thickness. Overexpression of a constitutively active Kras mimicked Lin28a overexpression, and inhibition of Erk signaling during embryonic stages normalized the cartilage phenotype of both Kras- and Lin28a-overexpressing mice. These results suggest that articular cartilage thickness is mainly determined during the process of embryonic synovial joint development, which is positively regulated by Erk signaling.
Collapse
Affiliation(s)
- Tatsuya Kobayashi
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Anastasia Kozlova
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
39
|
Luo W, Liu L, Yang L, Dong Y, Liu T, Wei X, Liu D, Gu H, Kong J, Yuan Z, Zhao Q. The vitamin D receptor regulates miR-140-5p and targets the MAPK pathway in bone development. Metabolism 2018; 85:139-150. [PMID: 29604361 DOI: 10.1016/j.metabol.2018.03.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 02/21/2018] [Accepted: 03/22/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Skeletal development is a complicated process. The status of vitamin D (VD) is closely related to fetal bone development in the embryonic period. Recently, miRNAs have been found to participate in the regulation of skeletal growth and development in several species. However, the mechanisms underlying the interactions among vitamin D, its receptor (VDR), and miRNAs during the process of bone development remain unclear. The aim of this study was to identify miRNAs that are regulated by 1,25(OH)2D3 in murine osteoblasts and to analyze the relationship and the effects of VD/VDR and miRNAs in vitro and in vivo. METHODS We performed miRNA sequencing in murine primary osteoblasts and in an osteoblast cell line treated with 1,25(OH)2D3 to identify miRNAs in these cells. After qRT-PCR validation, miR-140-5p was selected for further analysis. We assessed the pathways comprising predicted target genes for several expressed miRNAs, including miR-140-5p, validated predicted target genes in the MAPK pathway by qRT-PCR, and explored the correlation between VD/VDR and miR-140-5p in vitro and in vivo. RESULTS 88 miRNAs in murine primary osteoblasts and 49 miRNAs in osteoblast cell line were found to be differentially expressed. MiR-140-5p was upregulated in these 2 types of murine osteoblasts. The expression of miR-140-5p was promoted by 1,25(OH)2D3 through transcriptional activation by VDR, with targeted inhibition of MAPK signaling in osteoblasts. A positive correlation between vitamin D/VDR and miR-140-5p was observed in VDR-knockout mice and in 165 human serum specimens. These data show for the first time that VDR transcriptionally activates miR-140-5p. Therefore, the VD/VDR/miR-140-5p/MAPK signaling axis plays an important role in transmitting the effects of 1,25(OH)2D3. CONCLUSION Our results demonstrate a novel regulatory mechanism by which miR-140-5p targets the MAPK pathway by means of VD/VDR in vitro and in vivo. These findings provide a new reference for mechanistic research and therapeutic approaches for vitamin D-related bone diseases.
Collapse
Affiliation(s)
- Wenting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lingli Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China; Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liping Yang
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China; Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yaping Dong
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China; Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tianjing Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China; Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaowei Wei
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Dan Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hui Gu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Juan Kong
- Nutrition Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qun Zhao
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China; Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
40
|
MicroRNA-140 Suppresses Human Chondrocytes Hypertrophy by Targeting SMAD1 and Controlling the Bone Morphogenetic Protein Pathway in Osteoarthritis. Am J Med Sci 2018; 355:477-487. [DOI: 10.1016/j.amjms.2018.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/06/2018] [Accepted: 01/18/2018] [Indexed: 12/19/2022]
|
41
|
Jee YH, Wang J, Yue S, Jennings M, Clokie SJ, Nilsson O, Lui JC, Baron J. mir-374-5p, mir-379-5p, and mir-503-5p Regulate Proliferation and Hypertrophic Differentiation of Growth Plate Chondrocytes in Male Rats. Endocrinology 2018; 159:1469-1478. [PMID: 29390136 PMCID: PMC5839730 DOI: 10.1210/en.2017-00780] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/17/2018] [Indexed: 11/19/2022]
Abstract
Growth plate chondrocytes undergo sequential differentiation to form the resting zone, the proliferative zone (PZ), and the hypertrophic zone (HZ). The important role of microRNAs (miRNAs) in the growth plate was previously revealed by cartilage-specific ablation of Dicer, an enzyme essential for biogenesis of many miRNAs. To identify specific miRNAs that regulate differentiation of PZ chondrocytes to HZ chondrocytes, we microdissected individual growth plate zones from juvenile rats and performed miRNA profiling using a solution hybridization method and miRNA sequencing. Thirty-four miRNAs were differentially expressed between the PZ and the HZ, and we hypothesized that some of the miRNAs that are preferentially expressed in the PZ may promote proliferation and inhibit hypertrophic differentiation. Consistent with this hypothesis, transfection of inhibitors for four of these miRNAs (mir-369-3p, mir-374-5p, mir-379-5p, and mir-503-5p) decreased proliferation in primary epiphyseal chondrocytes. The inhibitors for three of these miRNAs (mir-374-5p, mir-379-5p, and mir-503-5p) also increased expression of multiple genes that are associated with chondrocyte hypertrophic differentiation. We next hypothesized that preferential expression of these miRNAs in the PZ is driven by the parathyroid hormone-related protein (PTHrP) concentration gradient across the growth plate. Consistent with this hypothesis, treatment of primary chondrocytes with a parathyroid hormone (PTH)/PTHrP receptor agonist, PTH1-34, increased expression of mir-374-5p, mir-379-5p, and mir-503-5p. Taken together, our findings suggest that the PTHrP concentration gradient across the growth plate induces differential expression of mir-374-5p, mir-379-5p, and mir-503-5p between the PZ and the HZ. In the PZ, the higher expression levels of these miRNAs promote proliferation and inhibit hypertrophic differentiation. In the HZ, downregulation of these miRNAs inhibits proliferation and promotes hypertrophic differentiation.
Collapse
Affiliation(s)
- Youn Hee Jee
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Jinhee Wang
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Shanna Yue
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Melissa Jennings
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Samuel J. Clokie
- Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham B15 2TG, United Kingdom
| | - Ola Nilsson
- Karolinska Institutet, Department of Women's and Children's Health, SE-171 76 Stockholm, Sweden
- Örebro University, Department of Medical Sciences, SE-701 82 Örebro, Sweden
| | - Julian C. Lui
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Jeffrey Baron
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
- Correspondence: Jeffrey Baron, MD, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive MSC 1103, Bethesda, Maryland 20892. E-mail:
| |
Collapse
|
42
|
Jiang S, Yan W, Wang SE, Baltimore D. Let-7 Suppresses B Cell Activation through Restricting the Availability of Necessary Nutrients. Cell Metab 2018; 27:393-403.e4. [PMID: 29337138 DOI: 10.1016/j.cmet.2017.12.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 07/19/2017] [Accepted: 12/09/2017] [Indexed: 12/18/2022]
Abstract
The control of uptake and utilization of necessary extracellular nutrients-glucose and glutamine-is an important aspect of B cell activation. Let-7 is a family of microRNAs known to be involved in metabolic control. Here, we employed several engineered mouse models, including B cell-specific overexpression of Lin28a or the let-7a-1/let-7d/let-7f-1 cluster (let-7adf) and knockout of individual let-7 clusters to show that let-7adf specifically inhibits T cell-independent (TI) antigen-induced immunoglobulin (Ig)M antibody production. Both overexpression and deletion of let-7 in this cluster leads to altered TI-IgM production. Mechanistically, let-7adf suppresses the acquisition and utilization of key nutrients, including glucose and glutamine, through directly targeting hexokinase 2 (Hk2) and by repressing a glutamine transporter Slc1a5 and a key degradation enzyme, glutaminase (Gls), a mechanism mediated by regulation of c-Myc. Our results suggest a novel role of let-7adf as a "metabolic brake" on B cell antibody production.
Collapse
Affiliation(s)
- Shuai Jiang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Wei Yan
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shizhen Emily Wang
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA.
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
43
|
Valenti MT, Dalle Carbonare L, Mottes M. Role of microRNAs in progenitor cell commitment and osteogenic differentiation in health and disease (Review). Int J Mol Med 2018; 41:2441-2449. [PMID: 29393379 DOI: 10.3892/ijmm.2018.3452] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 01/09/2018] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs) are considered 'micro- managers of gene expression' and awareness of their fundamental role in the control of biological functions is constantly increasing. Bone formation and homeostasis are complex processes involving the differentiation and interaction of various cell types. Several miRNAs have been shown to be involved in different pathways and stages in the regulation of normal and abnormal bone formation and turnover. This present review focuses on the involvement of miRNAs in terms of their effect on the commitment of bone marrow mesenchymal stem cells towards osteogenesis, adipogenesis and chondrogenesis, respectively. The miRNAs involved in regulating osteoblast, chondroblast and osteoclast activity, are also taken into consideration, as are their interactions. miRNA expression levels, which may differ significantly in healthy versus pathological conditions, can be readily monitored and represent useful biomarkers. Several studies have suggested that miRNAs offer potential as useful biomarkers of bone pathologies, including osteoporosis and osteosarcoma. The development of efficient methods of delivering miRNA mimics or miRNA inhibitors into specific cells remains a challenge for novel therapeutic applications in the field of personalized medicine.
Collapse
Affiliation(s)
- Maria Teresa Valenti
- Department of Medicine, Biomedicine and Movement Sciences, Biology and Genetics Section, University of Verona, Verona I‑37134, Italy
| | - Luca Dalle Carbonare
- Department of Medicine, Biomedicine and Movement Sciences, Biology and Genetics Section, University of Verona, Verona I‑37134, Italy
| | - Monica Mottes
- Department of Neurosciences, Biomedicine and Movement Sciences, Biology and Genetics Section, University of Verona, Verona I‑37134, Italy
| |
Collapse
|
44
|
Nanoscale Assembly of High-Mobility Group AT-Hook 2 Protein with DNA Replication Fork. Biophys J 2018; 113:2609-2620. [PMID: 29262356 DOI: 10.1016/j.bpj.2017.10.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/24/2017] [Accepted: 10/12/2017] [Indexed: 01/31/2023] Open
Abstract
High mobility group AT-hook 2 (HMGA2) protein is composed of three AT-hook domains. HMGA2 expresses at high levels in both embryonic stem cells and cancer cells, where it interacts with and stabilizes replication forks (RFs), resulting in elevated cell proliferation rates. In this study, we demonstrated that HMGA2 knockdown reduces cell proliferation. To understand the features required for interaction between HMGA2 and RFs, we studied the solution structure of HMGA2, free and in complex with RFs, using an integrated host of biophysical techniques. Circular dichroism and NMR experiments confirmed the disordered state of unbound HMGA2. Dynamic light scattering and sedimentation velocity experiments demonstrated that HMGA2 and RF are monodisperse in solution, and form an equimolar complex. Small-angle x-ray scattering studies revealed that HMGA2 binds in a side-by-side orientation to RF where 3 AT-hooks act as a clamp to wrap around a distorted RF. Thus, our data provide insights into how HMGA2 interacts with stalled RFs and the function of the process.
Collapse
|
45
|
Li Q, Huang QP, Wang YL, Huang QS. Extracellular vesicle-mediated bone metabolism in the bone microenvironment. J Bone Miner Metab 2018; 36:1-11. [PMID: 28766139 DOI: 10.1007/s00774-017-0860-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 07/04/2017] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are phospholipid membrane-enclosed entities containing specific proteins, RNA, miRNA, and lncRNA. EVs are released by various cells and play a vital role in cell communication by transferring their contents from the host cells to the recipient cells. The role of EVs has been characterized in a wide range of physiological and pathophysiological processes. In this context, we highlight recent advances in our understanding of the regulatory effects of EVs, with a focus on bone metabolism and the bone microenvironment. The roles of EVs in cell communication among bone-related cells, stem cells, tumor cells, and other cells under physiological or pathological conditions are also discussed. In addition, promising applications for EVs in treating bone-related diseases are proposed.
Collapse
Affiliation(s)
- Qi Li
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an, 710072, Shaanxi, People's Republic of China.
| | - Qiu-Ping Huang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Yi-Lin Wang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Qing-Sheng Huang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an, 710072, Shaanxi, People's Republic of China
| |
Collapse
|
46
|
Abstract
Regulation of body growth remains a fascinating and unresolved biological mystery. One key component of body growth is skeletal and longitudinal bone growth. Children grow taller because their bones grew longer, and the predominant driver of longitudinal bone growth is a cartilaginous structure found near the ends of long bone named the growth plate. Numerous recent studies have started to unveil the importance of microRNAs in regulation of growth plate functions, therefore contributing to regulation of linear growth. In addition to longitudinal growth, other organs in our body need to increase in size and cell number as we grow, and the regulation of organ growth involves both systemic factors like hormones; and other intrinsic mechanisms, which we are just beginning to understand. This review aims to summarize some recent important findings on how microRNAs are involved in both of these processes: the regulation of longitudinal bone growth, and the regulation of organs and overall body growth.
Collapse
Affiliation(s)
- Julian C Lui
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC Rm 1-3330, 10 Center Drive, MSC-1103, Bethesda, MD, 20892-1103, United States.
| |
Collapse
|
47
|
Sun YX, Zhang JF, Xu J, Xu LL, Wu TY, Wang B, Pan XH, Li G. MicroRNA-144-3p inhibits bone formation in distraction osteogenesis through targeting Connexin 43. Oncotarget 2017; 8:89913-89922. [PMID: 29163798 PMCID: PMC5685719 DOI: 10.18632/oncotarget.20984] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/17/2017] [Indexed: 11/25/2022] Open
Abstract
Distraction osteogenesis (DO), one of effective therapies for bone regeneration, has been received more attention in recent years. However, the underlying mechanism remains elusive. Recently, microRNAs (miRNAs) have been reported to play important roles in regulating osteogenesis and bone formation. We therefore provided the hypothesis that miRNAs could involve in the DO-mediated bone regeneration. After successfully established the DO model of rats, a miRNA microarray was performed to find the differently expressed miRNAs in DO and control groups in this study. As one of the most downregulated miRNAs, miR-144-3p was found to be decreased during osteogenic differentiation in mesenchymal stem cells of rats (rBMSCs) and DO model. And miR-144-3p overexpression suppressed the osteogenesis while its inhibitor promoted osteogenesis. Furthermore, Connexin-43, an essential regulator for osteogenesis, was validated to be a novel target for miR-144-3p. Finally, miR-144-3p inhibitor modified MSCs promoted mineralization of distracted bone in rat DO model. In conclusion, miR-144-3p was found to regulate osteogenesis and inhibition of miR-144-3p resulted in acceleration of mineralization of DO, which not only give clues to understanding the mechanism of DO but also provide a potential therapeutic target in clinical practice.
Collapse
Affiliation(s)
- Yu-Xin Sun
- Department of Orthopaedics and Traumatology, Bao-An District People's Hospital, Shenzhen, PR China.,Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Jin-Fang Zhang
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Jia Xu
- Department of Orthopaedics Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, PR China
| | - Liang-Liang Xu
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Tian-Yi Wu
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Bin Wang
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Xiao-Hua Pan
- Department of Orthopaedics and Traumatology, Bao-An District People's Hospital, Shenzhen, PR China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Bao-An District People's Hospital, Shenzhen, PR China.,Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China.,Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
48
|
Neumann JE, Wefers AK, Lambo S, Bianchi E, Bockstaller M, Dorostkar MM, Meister V, Schindler P, Korshunov A, von Hoff K, Nowak J, Warmuth-Metz M, Schneider MR, Renner-Müller I, Merk DJ, Shakarami M, Sharma T, Chavez L, Glass R, Chan JA, Taketo MM, Neumann P, Kool M, Schüller U. A mouse model for embryonal tumors with multilayered rosettes uncovers the therapeutic potential of Sonic-hedgehog inhibitors. Nat Med 2017; 23:1191-1202. [DOI: 10.1038/nm.4402] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/15/2017] [Indexed: 12/24/2022]
|
49
|
Li B, Bai L, Shen P, Sun Y, Chen Z, Wen Y. Identification of differentially expressed microRNAs in knee anterior cruciate ligament tissues surgically removed from patients with osteoarthritis. Int J Mol Med 2017; 40:1105-1113. [PMID: 28765881 PMCID: PMC5593459 DOI: 10.3892/ijmm.2017.3086] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 07/17/2017] [Indexed: 01/12/2023] Open
Abstract
The degradation of cruciate ligaments is frequently observed in degenerative joint diseases, such as osteoarthritis (OA). The present study aimed to identify the differentially expressed microRNAs (miRNAs or miRs) in knee anterior cruciate ligament (ACL) tissues derived from patients with OA and in health subjects (non-OA). By using Affymetrix miRNA 4.0 microarrays, a total of 22 miRNAs (including let-7f-5p, miR-26b-5p and miR-146a-5p) were found to be upregulated, while 17 (including miR-18a-3p, miR-138-5p and miR-485-3p) were downregulated in the osteoarthritic ACL tissues (fold change ≥2, P-value <0.05). The expression levels of 12 miRNAs were validated by quantitative PCR, and the corresponding results revealed an excellent correlation with the microarray data (R2=0.889). Genes (such as a disintegrin and metalloproteinase domain with thrombospondin type-1 motifs, bone morphogenetic protein-2, runt related transcription factor-2, collagen-1A1 and 2, interleukin-6 and transforming growth factor-β) involved in cartilage development and remodeling, collagen biosynthesis and degradation, inflammatory response and extracellular matrix homeostasis were predicted as potential targets of the dysregulated miRNAs. Moreover, a large set of putative genes were enriched in OA pathogenesis-associated pathways (such as mitogen-activated protein kinase and vascular endothelial growth factor signaling pathway). Collectively, the data from our study provides novel insight into the ligament injury-related miRNA dysregulation in patients with OA.
Collapse
Affiliation(s)
- Bin Li
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Lunhao Bai
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Peng Shen
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yue Sun
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhizuo Chen
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yu Wen
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|
50
|
Papaioannou G, Petit ET, Liu ES, Baccarini M, Pritchard C, Demay MB. Raf Kinases Are Essential for Phosphate Induction of ERK1/2 Phosphorylation in Hypertrophic Chondrocytes and Normal Endochondral Bone Development. J Biol Chem 2017; 292:3164-3171. [PMID: 28073913 PMCID: PMC5336153 DOI: 10.1074/jbc.m116.763342] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/04/2017] [Indexed: 01/18/2023] Open
Abstract
Hypophosphatemia causes rickets by impairing hypertrophic chondrocyte apoptosis. Phosphate induction of MEK1/2-ERK1/2 phosphorylation in hypertrophic chondrocytes is required for phosphate-mediated apoptosis and growth plate maturation. MEK1/2 can be activated by numerous molecules including Raf isoforms. A- and B-Raf ablation in chondrocytes does not alter skeletal development, whereas ablation of C-Raf decreases hypertrophic chondrocyte apoptosis and impairs vascularization of the growth plate. However, ablation of C-Raf does not impair phosphate-induced ERK1/2 phosphorylation in vitro, but leads to rickets by decreasing VEGF protein stability. To determine whether Raf isoforms are required for phosphate-induced hypertrophic chondrocyte apoptosis, mice lacking all three Raf isoforms in chondrocytes were generated. Raf deletion caused neonatal death and a significant expansion of the hypertrophic chondrocyte layer of the growth plate, accompanied by decreased cleaved caspase-9. This was associated with decreased phospho-ERK1/2 immunoreactivity in the hypertrophic chondrocyte layer and impaired vascular invasion. These data further demonstrated that Raf kinases are required for phosphate-induced ERK1/2 phosphorylation in cultured hypertrophic chondrocytes and perform essential, but partially redundant roles in growth plate maturation.
Collapse
Affiliation(s)
- Garyfallia Papaioannou
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School, Boston, Massachusetts 02115
| | - Elizabeth T Petit
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Eva S Liu
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School, Boston, Massachusetts 02115; Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Manuela Baccarini
- Department of Microbiology, Immunobiology and Genetics, Center of Molecular Biology, Max F. Perutz Laboratories, University of Vienna, Doktor-Bohr-Gasse 9, Vienna 1030, Austria
| | - Catrin Pritchard
- Department of Molecular and Cell Biology, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Marie B Demay
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School, Boston, Massachusetts 02115.
| |
Collapse
|