1
|
Bhat OM, Mir RA, Nehvi IB, Wani NA, Dar AH, Zargar MA. Emerging role of sphingolipids and extracellular vesicles in development and therapeutics of cardiovascular diseases. IJC HEART & VASCULATURE 2024; 53:101469. [PMID: 39139609 PMCID: PMC11320467 DOI: 10.1016/j.ijcha.2024.101469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024]
Abstract
Sphingolipids are eighteen carbon alcohol lipids synthesized from non-sphingolipid precursors in the endoplasmic reticulum (ER). The sphingolipids serve as precursors for a vast range of moieties found in our cells that play a critical role in various cellular processes, including cell division, senescence, migration, differentiation, apoptosis, pyroptosis, autophagy, nutrition intake, metabolism, and protein synthesis. In CVDs, different subclasses of sphingolipids and other derived molecules such as sphingomyelin (SM), ceramides (CERs), and sphingosine-1-phosphate (S1P) are directly related to diabetic cardiomyopathy, dilated cardiomyopathy, myocarditis, ischemic heart disease (IHD), hypertension, and atherogenesis. Several genome-wide association studies showed an association between genetic variations in sphingolipid pathway genes and the risk of CVDs. The sphingolipid pathway plays an important role in the biogenesis and secretion of exosomes. Small extracellular vesicles (sEVs)/ exosomes have recently been found as possible indicators for the onset of CVDs, linking various cellular signaling pathways that contribute to the disease progression. Important features of EVs like biocompatibility, and crossing of biological barriers can improve the pharmacokinetics of drugs and will be exploited to develop next-generation drug delivery systems. In this review, we have comprehensively discussed the role of sphingolipids, and sphingolipid metabolites in the development of CVDs. In addition, concise deliberations were laid to discuss the role of sEVs/exosomes in regulating the pathophysiological processes of CVDs and the exosomes as therapeutic targets.
Collapse
Affiliation(s)
- Owais Mohmad Bhat
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Rakeeb Ahmad Mir
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | | | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Abid Hamid Dar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - M Afzal Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| |
Collapse
|
2
|
Liu R, Li L, Wang Z, Zhu J, Ji Y. Acetylated Histone Modifications: Intersection of Diabetes and Atherosclerosis. J Cardiovasc Pharmacol 2024; 83:207-219. [PMID: 37989137 DOI: 10.1097/fjc.0000000000001516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
ABSTRACT Worldwide, type 2 diabetes is predominant form of diabetes, and it is mainly affected by the environment. Furthermore, the offspring of patients with type 2 diabetes and metabolic disorder syndrome may have a higher risk of diabetes and cardiovascular disease, which indicates that the environmental impact on diabetes prevalence can be transmitted across generations. In the process of diabetes onset and intergenerational transmission, the genetic structure of the individual is not directly changed but is regulated by epigenetics. In this process, genes or histones are modified, resulting in selective expression of proteins. This modification will affect not only the onset of diabetes but also the related onset of atherosclerosis. Acetylation and deacetylation may be important regulatory factors for the above lesions. Therefore, in this review, based on the whole process of atherosclerosis evolution, we explored the possible existence of acetylation/deacetylation caused by diabetes. However, because of the lack of atherosclerosis-related acetylation studies directly based on diabetic models, we also used a small number of experiments involving nondiabetic models of related molecular mechanisms.
Collapse
Affiliation(s)
| | | | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; and
| | - Jie Zhu
- Department of Cardiology, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu' an People's Hospital, Lu'an, China
| | | |
Collapse
|
3
|
Siracusa C, Carino A, Carabetta N, Manica M, Sabatino J, Cianflone E, Leo I, Strangio A, Torella D, De Rosa S. Mechanisms of Cardiovascular Calcification and Experimental Models: Impact of Vitamin K Antagonists. J Clin Med 2024; 13:1405. [PMID: 38592207 PMCID: PMC10932386 DOI: 10.3390/jcm13051405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 04/10/2024] Open
Abstract
Cardiovascular calcification is a multifactorial and complex process involving an array of molecular mechanisms eventually leading to calcium deposition within the arterial walls. This process increases arterial stiffness, decreases elasticity, influences shear stress events and is related to an increased risk of morbidity and mortality associated with cardiovascular disease. In numerous in vivo and in vitro models, warfarin therapy has been shown to cause vascular calcification in the arterial wall. However, the exact mechanisms of calcification formation with warfarin remain largely unknown, although several molecular pathways have been identified. Circulating miRNA have been evaluated as biomarkers for a wide range of cardiovascular diseases, but their exact role in cardiovascular calcification is limited. This review aims to describe the current state-of-the-art research on the impact of warfarin treatment on the development of vascular calcification and to highlight potential molecular targets, including microRNA, within the implicated pathways.
Collapse
Affiliation(s)
- Chiara Siracusa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.S.); (A.C.); (N.C.); (M.M.); (E.C.)
| | - Annarita Carino
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.S.); (A.C.); (N.C.); (M.M.); (E.C.)
| | - Nicole Carabetta
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.S.); (A.C.); (N.C.); (M.M.); (E.C.)
| | - Marzia Manica
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.S.); (A.C.); (N.C.); (M.M.); (E.C.)
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (J.S.); (I.L.); (A.S.); (D.T.)
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.S.); (A.C.); (N.C.); (M.M.); (E.C.)
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (J.S.); (I.L.); (A.S.); (D.T.)
| | - Antonio Strangio
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (J.S.); (I.L.); (A.S.); (D.T.)
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (J.S.); (I.L.); (A.S.); (D.T.)
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.S.); (A.C.); (N.C.); (M.M.); (E.C.)
| |
Collapse
|
4
|
Pultar M, Oesterreicher J, Hartmann J, Weigl M, Diendorfer A, Schimek K, Schädl B, Heuser T, Brandstetter M, Grillari J, Sykacek P, Hackl M, Holnthoner W. Analysis of extracellular vesicle microRNA profiles reveals distinct blood and lymphatic endothelial cell origins. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e134. [PMID: 38938681 PMCID: PMC11080916 DOI: 10.1002/jex2.134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/22/2023] [Accepted: 12/22/2023] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are crucial mediators of cell-to-cell communication in physiological and pathological conditions. Specifically, EVs released from the vasculature into blood were found to be quantitatively and qualitatively different in diseases compared to healthy states. However, our understanding of EVs derived from the lymphatic system is still scarce. In this study, we compared the mRNA and microRNA (miRNA) expression in blood vascular (BEC) and lymphatic (LEC) endothelial cells. After characterization of the EVs by fluorescence-triggered flow cytometry, nanoparticle tracking analysis and cryo-transmission electron microscopy (cryo-TEM) we utilized small RNA-sequencing to characterize miRNA signatures in the EVs and identify cell-type specific miRNAs in BEC and LEC. We found miRNAs specifically enriched in BEC and LEC on the cellular as well as the extracellular vesicle level. Our data provide a solid basis for further functional in vitro and in vivo studies addressing the role of EVs in the blood and lymphatic vasculature.
Collapse
Affiliation(s)
- Marianne Pultar
- Ludwig Boltzmann Institute for TraumatologyThe Research Centre in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
- TAmiRNA GmbHViennaAustria
| | - Johannes Oesterreicher
- Ludwig Boltzmann Institute for TraumatologyThe Research Centre in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | | | - Moritz Weigl
- Ludwig Boltzmann Institute for TraumatologyThe Research Centre in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
- TAmiRNA GmbHViennaAustria
| | | | - Katharina Schimek
- Technische Universität Berlin, Medical BiotechnologyBerlinGermany
- TissUse GmbHBerlinGermany
| | - Barbara Schädl
- Ludwig Boltzmann Institute for TraumatologyThe Research Centre in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
- University Clinic of DentistryMedical University of ViennaViennaAustria
| | - Thomas Heuser
- Vienna Biocenter Core Facilities GmbH, EM FacilityViennaAustria
| | | | - Johannes Grillari
- Ludwig Boltzmann Institute for TraumatologyThe Research Centre in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
- Department of Biotechnology, Institute of Molecular BiotechnologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Peter Sykacek
- Department of Biotechnology, Institute of Computational BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | | | - Wolfgang Holnthoner
- Ludwig Boltzmann Institute for TraumatologyThe Research Centre in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| |
Collapse
|
5
|
Wang G, Luo Y, Gao X, Liang Y, Yang F, Wu J, Fang D, Luo M. MicroRNA regulation of phenotypic transformations in vascular smooth muscle: relevance to vascular remodeling. Cell Mol Life Sci 2023; 80:144. [PMID: 37165163 PMCID: PMC11071847 DOI: 10.1007/s00018-023-04793-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/10/2023] [Accepted: 04/27/2023] [Indexed: 05/12/2023]
Abstract
Alterations in the vascular smooth muscle cells (VSMC) phenotype play a critical role in the pathogenesis of several cardiovascular diseases, including hypertension, atherosclerosis, and restenosis after angioplasty. MicroRNAs (miRNAs) are a class of endogenous noncoding RNAs (approximately 19-25 nucleotides in length) that function as regulators in various physiological and pathophysiological events. Recent studies have suggested that aberrant miRNAs' expression might underlie VSMC phenotypic transformation, appearing to regulate the phenotypic transformations of VSMCs by targeting specific genes that either participate in the maintenance of the contractile phenotype or contribute to the transformation to alternate phenotypes, and affecting atherosclerosis, hypertension, and coronary artery disease by altering VSMC proliferation, migration, differentiation, inflammation, calcification, oxidative stress, and apoptosis, suggesting an important regulatory role in vascular remodeling for maintaining vascular homeostasis. This review outlines recent progress in the discovery of miRNAs and elucidation of their mechanisms of action and functions in VSMC phenotypic regulation. Importantly, as the literature supports roles for miRNAs in modulating vascular remodeling and for maintaining vascular homeostasis, this area of research will likely provide new insights into clinical diagnosis and prognosis and ultimately facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Gang Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yulin Luo
- GCP Center, Affiliated Hospital (Traditional Chinese Medicine) of Southwest Medical University, Luzhou, China
| | - Xiaojun Gao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Liang
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Feifei Yang
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Dan Fang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China.
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China.
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
6
|
Williams MJ, White SC, Joseph Z, Hruska KA. Updates in the chronic kidney disease-mineral bone disorder show the role of osteocytic proteins, a potential mechanism of the bone-Vascular paradox, a therapeutic target, and a biomarker. Front Physiol 2023; 14:1120308. [PMID: 36776982 PMCID: PMC9909112 DOI: 10.3389/fphys.2023.1120308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
The chronic kidney disease-mineral bone disorder (CKD-MBD) is a complex multi-component syndrome occurring during kidney disease and its progression. Here, we update progress in the components of the syndrome, and synthesize recent investigations, which suggest a potential mechanism of the bone-vascular paradox. The discovery that calcified arteries in chronic kidney disease inhibit bone remodeling lead to the identification of factors produced by the vasculature that inhibit the skeleton, thus providing a potential explanation for the bone-vascular paradox. Among the factors produced by calcifying arteries, sclerostin secretion is especially enlightening. Sclerostin is a potent inhibitor of bone remodeling and an osteocyte specific protein. Its production by the vasculature in chronic kidney disease identifies the key role of vascular cell osteoblastic/osteocytic transdifferentiation in vascular calcification and renal osteodystrophy. Subsequent studies showing that inhibition of sclerostin activity by a monoclonal antibody improved bone remodeling as expected, but stimulated vascular calcification, demonstrate that vascular sclerostin functions to brake the Wnt stimulation of the calcification milieu. Thus, the target of therapy in the chronic kidney disease-mineral bone disorder is not inhibition of sclerostin function, which would intensify vascular calcification. Rather, decreasing sclerostin production by decreasing the vascular osteoblastic/osteocytic transdifferentiation is the goal. This might decrease vascular calcification, decrease vascular stiffness, decrease cardiac hypertrophy, decrease sclerostin production, reduce serum sclerostin and improve skeletal remodeling. Thus, the therapeutic target of the chronic kidney disease-mineral bone disorder may be vascular osteoblastic transdifferentiation, and sclerostin levels may be a useful biomarker for the diagnosis of the chronic kidney disease-mineral bone disorder and the progress of its therapy.
Collapse
Affiliation(s)
- Matthew J. Williams
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Sarah C. White
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Zachary Joseph
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Keith A. Hruska
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
- Departments of Medicine and Cell Biology, Washington University, Saint Louis, MO, United States
| |
Collapse
|
7
|
Pisano C, Terriaca S, Scioli MG, Nardi P, Altieri C, Orlandi A, Ruvolo G, Balistreri CR. The Endothelial Transcription Factor ERG Mediates a Differential Role in the Aneurysmatic Ascending Aorta with Bicuspid or Tricuspid Aorta Valve: A Preliminary Study. Int J Mol Sci 2022; 23:10848. [PMID: 36142762 PMCID: PMC9502538 DOI: 10.3390/ijms231810848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
The pathobiology of ascending aorta aneurysms (AAA) onset and progression is not well understood and only partially characterized. AAA are also complicated in case of bicuspid aorta valve (BAV) anatomy. There is emerging evidence about the crucial role of endothelium-related pathways, which show in AAA an altered expression and function. Here, we examined the involvement of ERG-related pathways in the differential progression of disease in aortic tissues from patients having a BAV or tricuspid aorta valve (TAV) with or without AAA. Our findings identified ERG as a novel endothelial-specific regulator of TGF-β-SMAD, Notch, and NO pathways, by modulating a differential fibrotic or calcified AAA progression in BAV and TAV aortas. We provided evidence that calcification is correlated to different ERG expression (as gene and protein), which appears to be under control of Notch signaling. The latter, when increased, associated with an early calcification in aortas with BAV valve and aneurysmatic, was demonstrated to favor the progression versus severe complications, i.e., dissection or rupture. In TAV aneurysmatic aortas, ERG appeared to modulate fibrosis. Therefore, we proposed that ERG may represent a sensitive tissue biomarker to monitor AAA progression and a target to develop therapeutic strategies and influence surgical procedures.
Collapse
Affiliation(s)
- Calogera Pisano
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Sonia Terriaca
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Maria Giovanna Scioli
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Paolo Nardi
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Claudia Altieri
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Augusto Orlandi
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
- Department of Biomedical Sciences, Catholic University of Our Lady of Good Counsel, 1001 Tirana, Albania
| | - Giovanni Ruvolo
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy
| |
Collapse
|
8
|
Wang ZX, Luo ZW, Li FXZ, Cao J, Rao SS, Liu YW, Wang YY, Zhu GQ, Gong JS, Zou JT, Wang Q, Tan YJ, Zhang Y, Hu Y, Li YY, Yin H, Wang XK, He ZH, Ren L, Liu ZZ, Hu XK, Yuan LQ, Xu R, Chen CY, Xie H. Aged bone matrix-derived extracellular vesicles as a messenger for calcification paradox. Nat Commun 2022; 13:1453. [PMID: 35304471 PMCID: PMC8933454 DOI: 10.1038/s41467-022-29191-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Adipocyte differentiation of bone marrow mesenchymal stem/stromal cells (BMSCs) instead of osteoblast formation contributes to age- and menopause-related marrow adiposity and osteoporosis. Vascular calcification often occurs with osteoporosis, a contradictory association called “calcification paradox”. Here we show that extracellular vesicles derived from aged bone matrix (AB-EVs) during bone resorption favor BMSC adipogenesis rather than osteogenesis and augment calcification of vascular smooth muscle cells. Intravenous or intramedullary injection of AB-EVs promotes bone-fat imbalance and exacerbates Vitamin D3 (VD3)-induced vascular calcification in young or old mice. Alendronate (ALE), a bone resorption inhibitor, down-regulates AB-EVs release and attenuates aging- and ovariectomy-induced bone-fat imbalance. In the VD3-treated aged mice, ALE suppresses the ovariectomy-induced aggravation of vascular calcification. MiR-483-5p and miR-2861 are enriched in AB-EVs and essential for the AB-EVs-induced bone-fat imbalance and exacerbation of vascular calcification. Our study uncovers the role of AB-EVs as a messenger for calcification paradox by transferring miR-483-5p and miR-2861. This study uncovers the role of extracellular vesicles from bone matrix as a messenger in the development of osteoporosis and vascular calcification (calcification paradox) during skeletal aging and menopause by transferring miR-483-5p and miR-2861.
Collapse
Affiliation(s)
- Zhen-Xing Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhong-Wei Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fu-Xing-Zi Li
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Cao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shan-Shan Rao
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Yi-Wei Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi-Yi Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guo-Qiang Zhu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiang-Shan Gong
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing-Tao Zou
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiang Wang
- Department of Laboratory Medicine, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yi-Juan Tan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Zhang
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yin Hu
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - You-You Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Yin
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao-Kai Wang
- Department of Emergency Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ze-Hui He
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lu Ren
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng-Zhao Liu
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Organ Injury, Aging and Regenerative Medicine, Changsha, Hunan, China.,Hunan Key Laboratory of Bone Joint Degeneration and Injury, Changsha, Hunan, China
| | - Xiong-Ke Hu
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ling-Qing Yuan
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ran Xu
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chun-Yuan Chen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hui Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Organ Injury, Aging and Regenerative Medicine, Changsha, Hunan, China. .,Hunan Key Laboratory of Bone Joint Degeneration and Injury, Changsha, Hunan, China.
| |
Collapse
|
9
|
Brown SV, Dewitt S, Clayton A, Waddington RJ. Identifying the Efficacy of Extracellular Vesicles in Osteogenic Differentiation: An EV-Lution in Regenerative Medicine. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.849724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have long been the focus for regenerative medicine and the restoration of damaged or aging cells throughout the body. However, the efficacy of MSCs in cell-based therapy still remains unpredictable and carries with it enumerable risks. It is estimated that only 3-10% of MSCs survive transplantation, and there remains undefined and highly variable heterogeneous biological potency within these administered cell populations. The mode of action points to secreted factors produced by MSCs rather than the reliance on engraftment. Hence harnessing such secreted elements as a replacement for live-cell therapies is attractive. Extracellular vesicles (EVs) are heterogenous lipid bounded structures, secreted by cells. They comprise a complex repertoire of molecules including RNA, proteins and other factors that facilitate cell-to-cell communication. Described as protected signaling centers, EVs can modify the cellular activity of recipient cells and are emerging as a credible alternative to cell-based therapies. EV therapeutics demonstrate beneficial roles for wound healing by preventing apoptosis, moderating immune responses, and stimulating angiogenesis, in addition to promoting cell proliferation and differentiation required for tissue matrix synthesis. Significantly, EVs maintain their signaling function following transplantation, circumventing the issues related to cell-based therapies. However, EV research is still in its infancy in terms of their utility as medicinal agents, with many questions still surrounding mechanistic understanding, optimal sourcing, and isolation of EVs for regenerative medicine. This review will consider the efficacy of using cell-derived EVs compared to traditional cell-based therapies for bone repair and regeneration. We discuss the factors to consider in developing productive lines of inquiry and establishment of standardized protocols so that EVs can be harnessed from optimal secretome production, to deliver reproducible and effective therapies.
Collapse
|
10
|
Zheng H, Liu J, Yu J, McAlinden A. Expression profiling of mitochondria-associated microRNAs during osteogenic differentiation of human MSCs. Bone 2021; 151:116058. [PMID: 34144232 PMCID: PMC8944210 DOI: 10.1016/j.bone.2021.116058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/28/2021] [Accepted: 06/08/2021] [Indexed: 12/31/2022]
Abstract
Small non-coding microRNAs (miRNAs) have the ability to target and bind to many mRNAs within the cytosol resulting in reduced protein expression and modulation of a number of cellular pathways and networks. In addition to the cytosol, miRNAs have been identified in other cellular compartments and organelles, including the mitochondria. While a few mitochondria-associated miRNAs (mitomiRs) are predicted to be derived from the mitochondrial genome, the majority appear to be transcribed from nuclear DNA and somehow transported into the mitochondria. These findings raise interesting questions about why miRNAs are located in the mitochondria and if they play a role in regulating processes within these organelles. Previously published work from our laboratory showed that miR-181a/b can regulate osteogenesis, in part, by enhancing mitochondrial metabolism. In other published studies, miR-181 paralogs and many other miRNAs have been identified in mitochondrial extracts derived from common cell lines and specific primary cells and tissues. Taken together, we were motivated to identify mitomiR expression profiles during in vitro osteogenesis. Specifically, we obtained RNA from purified mitochondrial extracts of human bone marrow-derived mesenchymal stem/stromal cells (MSCs) and from whole cell extracts of MSCs at day 0 or following osteogenic induction for 3, 7 and 14 days. Utilizing Affymetrix GeneChip™ miRNA 4.0 arrays, mitomiR expression signatures were determined at each time point. Based on the Affymetrix detection above background algorithm, the total number of miRNAs detected in MSC mitochondria extracts was 527 (non-induced MSCs), 627 (day 3 induced), 372 (day 7 induced) and 498 (day 14 induced). In addition, we identified significantly differentially-expressed mitomiRs at day 7 and day 14 of osteogenic induction when compared to day 0 (fold change ≥1.5; adjusted p value <0.05). In general, the most pronounced and highly significant changes in mitomiR expression during osteogenesis were observed at the day 7 time point. Interestingly, most miRNAs found to be differentially-expressed in mitochondria extracts did not show significantly altered expression in whole cell extracts at the same time points during osteoblast differentiation. This array study provides novel information on miRNAs associated with the mitochondria in MSCs during differentiation toward the osteoblast phenotype. These findings will guide future research to identify new miRNA candidates that may function in regulating mitochondrial function and/or bone formation, homeostasis or repair.
Collapse
Affiliation(s)
- Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Jin Liu
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Jinsheng Yu
- Genome Technology Access Center, Washington University School of Medicine, St Louis, MO, United States of America.
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, United States of America; Shriners Hospital for Children - St Louis, St Louis, MO, United States of America.
| |
Collapse
|
11
|
Aryankalayil MJ, Martello S, Bylicky MA, Chopra S, May JM, Shankardass A, MacMillan L, Sun L, Sanjak J, Vanpouille-Box C, Eke I, Coleman CN. Analysis of lncRNA-miRNA-mRNA expression pattern in heart tissue after total body radiation in a mouse model. J Transl Med 2021; 19:336. [PMID: 34364390 PMCID: PMC8349067 DOI: 10.1186/s12967-021-02998-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
Background Radiation therapy is integral to effective thoracic cancer treatments, but its application is limited by sensitivity of critical organs such as the heart. The impacts of acute radiation-induced damage and its chronic effects on normal heart cells are highly relevant in radiotherapy with increasing lifespans of patients. Biomarkers for normal tissue damage after radiation exposure, whether accidental or therapeutic, are being studied as indicators of both acute and delayed effects. Recent research has highlighted the potential importance of RNAs, including messenger RNAs (mRNAs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) as biomarkers to assess radiation damage. Understanding changes in mRNA and non-coding RNA expression will elucidate biological pathway changes after radiation. Methods To identify significant expression changes in mRNAs, lncRNAs, and miRNAs, we performed whole transcriptome microarray analysis of mouse heart tissue at 48 h after whole-body irradiation with 1, 2, 4, 8, and 12 Gray (Gy). We also validated changes in specific lncRNAs through RT-qPCR. Ingenuity Pathway Analysis (IPA) was used to identify pathways associated with gene expression changes. Results We observed sustained increases in lncRNAs and mRNAs, across all doses of radiation. Alas2, Aplnr, and Cxc3r1 were the most significantly downregulated mRNAs across all doses. Among the significantly upregulated mRNAs were cell-cycle arrest biomarkers Gdf15, Cdkn1a, and Ckap2. Additionally, IPA identified significant changes in gene expression relevant to senescence, apoptosis, hemoglobin synthesis, inflammation, and metabolism. LncRNAs Abhd11os, Pvt1, Trp53cor1, and Dino showed increased expression with increasing doses of radiation. We did not observe any miRNAs with sustained up- or downregulation across all doses, but miR-149-3p, miR-6538, miR-8101, miR-7118-5p, miR-211-3p, and miR-3960 were significantly upregulated after 12 Gy. Conclusions Radiation-induced RNA expression changes may be predictive of normal tissue toxicities and may indicate targetable pathways for radiation countermeasure development and improved radiotherapy treatment plans. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02998-w.
Collapse
Affiliation(s)
- Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA.
| | - Shannon Martello
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | - Michelle A Bylicky
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | - Sunita Chopra
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | - Jared M May
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | - Aman Shankardass
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | | | - Landy Sun
- Gryphon Scientific, Takoma Park, MD, 20912, USA
| | | | | | - Iris Eke
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA.,Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA.,Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD, 20850, USA
| |
Collapse
|
12
|
Zheng D, Huo M, Li B, Wang W, Piao H, Wang Y, Zhu Z, Li D, Wang T, Liu K. The Role of Exosomes and Exosomal MicroRNA in Cardiovascular Disease. Front Cell Dev Biol 2021; 8:616161. [PMID: 33511124 PMCID: PMC7835482 DOI: 10.3389/fcell.2020.616161] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
Exosomes are small vesicles (30–150 nm in diameter) enclosed by a lipid membrane bilayer, secreted by most cells in the body. They carry various molecules, including proteins, lipids, mRNA, and other RNA species, such as long non-coding RNA, circular RNA, and microRNA (miRNA). miRNAs are the most numerous cargo molecules in the exosome. They are endogenous non-coding RNA molecules, approximately 19–22-nt-long, and important regulators of protein biosynthesis. Exosomes can be taken up by neighboring or distant cells, where they play a role in post-transcriptional regulation of gene expression by targeting mRNA. Exosomal miRNAs have diverse functions, such as participation in inflammatory reactions, cell migration, proliferation, apoptosis, autophagy, and epithelial–mesenchymal transition. There is increasing evidence that exosomal miRNAs play an important role in cardiovascular health. Exosomal miRNAs are widely involved in the occurrence and development of cardiovascular diseases, such as atherosclerosis, acute coronary syndrome, heart failure (HF), myocardial ischemia reperfusion injury, and pulmonary hypertension. In this review, we present a systematic overview of the research progress into the role of exosomal miRNAs in cardiovascular diseases, and present new ideas for the diagnosis and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Dongdong Zheng
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Ming Huo
- Department of Day Operating Room, The Second Hospital of Jilin University, Changchun, China
| | - Bo Li
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Weitie Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Hulin Piao
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yong Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Zhicheng Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Dan Li
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Tiance Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Wang SS, Wang C, Chen H. MicroRNAs are critical in regulating smooth muscle cell mineralization and apoptosis during vascular calcification. J Cell Mol Med 2020; 24:13564-13572. [PMID: 33089928 PMCID: PMC7754013 DOI: 10.1111/jcmm.16005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/27/2020] [Accepted: 10/01/2020] [Indexed: 02/01/2023] Open
Abstract
Vascular calcification refers to the pathological deposition of calcium and phosphate minerals into the vasculature. It is prevalent in atherosclerosis, ageing, type 2 diabetes mellitus and chronic kidney disease, thus, increasing morbidity and mortality from these conditions. Vascular calcification shares similar mechanisms with bone mineralization, with smooth muscle cells playing a critical role in both processes. In the last decade, a variety of microRNAs have been identified as key regulators for the differentiation, phenotypic switch, proliferation, apoptosis, cytokine production and matrix deposition in vascular smooth muscle cells during vascular calcification. Therefore, this review mainly discusses the roles of microRNAs in the pathophysiological mechanisms of vascular calcification in smooth muscle cells and describes several interventions against vascular calcification by regulating microRNAs. As the exact mechanisms of calcification remain not fully elucidated, having a better understanding of microRNA involvement in vascular calcification may give impetus to development of novel therapeutics for the control and treatment of vascular calcification.
Collapse
Affiliation(s)
- Shan-Shan Wang
- Department of Cardiology, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Wang
- Department of Cardiology, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Han Chen
- Department of Cardiology, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
14
|
Jiao J, Feng G, Wu M, Wang Y, Li R, Liu J. MiR-140-5p promotes osteogenic differentiation of mouse embryonic bone marrow mesenchymal stem cells and post-fracture healing of mice. Cell Biochem Funct 2020; 38:1152-1160. [PMID: 33047358 PMCID: PMC7756898 DOI: 10.1002/cbf.3585] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 07/09/2020] [Accepted: 08/01/2020] [Indexed: 12/13/2022]
Abstract
MiR-140-5p is high expressed in normal fracture healing, but its specific role and mechanism in tissue-to-bone healing are rarely reported. Therefore, this study investigated the effects of miR-140-5p on tissue-to-bone healing. Clone formation experiment, flow cytometry, Alizarin Red S Staining and Oil Red O Staining were performed to investigate the biological characteristics of mouse embryonic bone marrow mesenchymal stem cells C3H10T1/2. MiR-140-5p mimic was transfected into osteogenic medium (OS)-treated C3H10T1/2 cells to investigate the effects of miR-140-5p on osteogenic differentiation. MiR-140-5p transgenic mouse model and the transgenic fracture model were established, and the effects of miR-140-5p on osteogenic differentiation, bone mineral density (BMD) and bone mass of bone tissues were detected by haematoxylin and eosin staining and computed tomography scan. The expressions of osteocalcin, differentiation-related genes (Runx2, ALP, Spp1 and Bglap3) and miR-140-5p were determined by quantitative real-time polymerase chain reaction. C3H10T1/2 cells showed the abilities of forming cloned differentiation of osteogenesis, fat cells, and its phenotypes including CD44, CD90.1 and Sca-1 but excluding CD45 haematopoietic stem cell marker. Overexpression of miR-140-5p promoted the expressions of differentiation-related genes and calcium deposition of OS-treated C3H10T1/2 cells. MiR-140-5p increased the expression of osteocalcin, BMD and bone mass and promoted bone healing of miR-140-5p-transgenic mice with fracture. MiR-140-5p promoted osteogenic differentiation of mouse embryonic bone marrow mesenchymal stem cells and post-fracture healing in mice. SIGNIFICANCE OF THE STUDY: C3H10T1/2 cells showed the abilities of forming cloned differentiation of osteogenesis, fat cells and its phenotypes including CD44, CD90.1 and Sca-1 but excluding CD45 haematopoietic stem cell marker. Overexpression of miR-140-5p promoted the expressions of differentiation-related genes and calcium deposition of osteogenic medium-treated C3H10T1/2 cells. MiR-140-5p increased the expression of osteocalcin and bone mineral density and bone mass and promoted bone healing of miR-140-5p-transgenic mice with fracture. Our results showed that miR-140-5p promoted osteogenic differentiation of mouse embryonic bone marrow mesenchymal stem cells and post-fracture healing in mice, which may be a therapeutic target for treating fractures and promoting bone healing.
Collapse
Affiliation(s)
- Jianhang Jiao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Guang Feng
- The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Minfei Wu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Yang Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Rui Li
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jun Liu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Ryu J, Ahn Y, Kook H, Kim YK. The roles of non-coding RNAs in vascular calcification and opportunities as therapeutic targets. Pharmacol Ther 2020; 218:107675. [PMID: 32910935 DOI: 10.1016/j.pharmthera.2020.107675] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is characterized by an accumulation of calcium phosphate crystals inside the vessel wall. VC is often associated with diabetes, chronic kidney disease (CKD), atherosclerosis, and cardiovascular disease (CVD). Even though the number of patients with VC remains prevalent, there are still no approved therapies for the treatment of VC. Since the pathogenesis of VC is diverse and involves multiple factors and mechanisms, it is critical to reveal the novel mechanisms involved in VC. Although protein-coding RNAs involved in VC have been extensively studied, the roles of non-coding RNAs (ncRNAs) are not yet fully understood. The field of ncRNAs has recently received attention, and accumulating evidence from studies in VC suggests that ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), play an important role in the regulation of VC. NcRNAs can modulate VC by acting as promoters or inhibitors and may be useful in the clinical diagnosis and treatment of VC. In this article, we review and discuss ncRNAs that regulate VC and present the therapeutic implications of these ncRNAs.
Collapse
Affiliation(s)
- Juhee Ryu
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Jeollanam-do, Republic of Korea; Department of Biochemistry, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Pharmacology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea
| | - Youngkeun Ahn
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Cardiology, Cardiovascular Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hyun Kook
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Jeollanam-do, Republic of Korea; Department of Pharmacology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.
| | - Young-Kook Kim
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Jeollanam-do, Republic of Korea; Department of Biochemistry, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.
| |
Collapse
|
16
|
Choe N, Shin S, Joung H, Ryu J, Kim Y, Ahn Y, Kook H, Kwon D. The microRNA miR-134-5p induces calcium deposition by inhibiting histone deacetylase 5 in vascular smooth muscle cells. J Cell Mol Med 2020; 24:10542-10550. [PMID: 32783377 PMCID: PMC7521311 DOI: 10.1111/jcmm.15670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 12/30/2022] Open
Abstract
Calcium deposition in vascular smooth muscle cells (VSMCs) is a form of ectopic ossification in blood vessels. It can result in rigidity of the vasculature and an increase in cardiac events. Here, we report that the microRNA miR-134-5p potentiates inorganic phosphate (Pi)-induced calcium deposition in VSMCs by inhibiting histone deacetylase 5 (HDAC5). Using miRNA microarray analysis of Pi-treated rat VSMCs, we first selected miR-134-5p for further evaluation. Quantitative RT-PCR confirmed that miR-134-5p was increased in Pi-treated A10 cells, a rat VSMC line. Transfection of miR-134-5p mimic potentiated the Pi-induced increase in calcium contents. miR-134-5p increased the amounts of bone runt-related transcription factor 2 (RUNX2) protein and bone morphogenic protein 2 (BMP2) mRNA in the presence of Pi but decreased the expression of osteoprotegerin (OPG). Bioinformatic analysis showed that the HDAC5 3'untranslated region (3'UTR) was one of the targets of miR-134-5p. The luciferase construct containing the 3'UTR of HDAC5 was down-regulated by miR-134-5p mimic in a dose-dependent manner in VSMCs. Overexpression of HDAC5 mitigated the calcium deposition induced by miR-134-5p. Our results suggest that a Pi-induced increase of miR-134-5p may cause vascular calcification through repression of HDAC5.
Collapse
Affiliation(s)
- Nakwon Choe
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Sera Shin
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Hosouk Joung
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Juhee Ryu
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Young‐Kook Kim
- Department of BiochemistryChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Youngkeun Ahn
- Department of CardiologyChonnam National University HospitalGwangjuRepublic of Korea
| | - Hyun Kook
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Duk‐Hwa Kwon
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| |
Collapse
|
17
|
Roles of Histone Acetylation Modifiers and Other Epigenetic Regulators in Vascular Calcification. Int J Mol Sci 2020; 21:ijms21093246. [PMID: 32375326 PMCID: PMC7247359 DOI: 10.3390/ijms21093246] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/28/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification (VC) is characterized by calcium deposition inside arteries and is closely associated with the morbidity and mortality of atherosclerosis, chronic kidney disease, diabetes, and other cardiovascular diseases (CVDs). VC is now widely known to be an active process occurring in vascular smooth muscle cells (VSMCs) involving multiple mechanisms and factors. These mechanisms share features with the process of bone formation, since the phenotype switching from the contractile to the osteochondrogenic phenotype also occurs in VSMCs during VC. In addition, VC can be regulated by epigenetic factors, including DNA methylation, histone modification, and noncoding RNAs. Although VC is commonly observed in patients with chronic kidney disease and CVD, specific drugs for VC have not been developed. Thus, discovering novel therapeutic targets may be necessary. In this review, we summarize the current experimental evidence regarding the role of epigenetic regulators including histone deacetylases and propose the therapeutic implication of these regulators in the treatment of VC.
Collapse
|
18
|
The Epigenetic Landscape of Vascular Calcification: An Integrative Perspective. Int J Mol Sci 2020; 21:ijms21030980. [PMID: 32024140 PMCID: PMC7037112 DOI: 10.3390/ijms21030980] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/26/2022] Open
Abstract
Vascular calcification (VC) is an important complication among patients of advanced age, those with chronic kidney disease, and those with diabetes mellitus. The pathophysiology of VC encompasses passive occurrence of physico-chemical calcium deposition, active cellular secretion of osteoid matrix upon exposure to metabolically noxious stimuli, or a variable combination of both processes. Epigenetic alterations have been shown to participate in this complex environment, through mechanisms including DNA methylation, non-coding RNAs, histone modifications, and chromatin changes. Despite such importance, existing reviews fail to provide a comprehensive view of all relevant reports addressing epigenetic processes in VC, and cross-talk between different epigenetic machineries is rarely examined. We conducted a systematic review based on PUBMED and MEDLINE databases up to 30 September 2019, to identify clinical, translational, and experimental reports addressing epigenetic processes in VC; we retrieved 66 original studies, among which 60.6% looked into the pathogenic role of non-coding RNA, followed by DNA methylation (12.1%), histone modification (9.1%), and chromatin changes (4.5%). Nine (13.6%) reports examined the discrepancy of epigenetic signatures between subjects or tissues with and without VC, supporting their applicability as biomarkers. Assisted by bioinformatic analyses blending in each epigenetic component, we discovered prominent interactions between microRNAs, DNA methylation, and histone modification regarding potential influences on VC risk.
Collapse
|
19
|
Zhu L, Li Q, Li Q, Qi D, Gao C, Yang H. MicroRNA‐2861 and microRNA‐5115 regulates myocardial ischemia–reperfusion injury through the GPR30/mTOR signaling pathway by binding to GPR30. J Cell Physiol 2020; 235:7791-7802. [PMID: 31930508 DOI: 10.1002/jcp.29427] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Lijie Zhu
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou P.R. China
- Department of Cardiology Fuwai Central China Cardiovascular Hospital Zhengzhou P.R. China
| | - Qingman Li
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou P.R. China
- Department of Cardiology Fuwai Central China Cardiovascular Hospital Zhengzhou P.R. China
| | - Qingmin Li
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou P.R. China
- Department of Cardiology Fuwai Central China Cardiovascular Hospital Zhengzhou P.R. China
| | - Datun Qi
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou P.R. China
- Department of Cardiology Fuwai Central China Cardiovascular Hospital Zhengzhou P.R. China
| | - Chuanyu Gao
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou P.R. China
- Department of Cardiology Fuwai Central China Cardiovascular Hospital Zhengzhou P.R. China
| | - Honghui Yang
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou P.R. China
- Department of Cardiology Fuwai Central China Cardiovascular Hospital Zhengzhou P.R. China
| |
Collapse
|
20
|
Liu H, Huang LH, Sun XY, Ouyang JM. High-phosphorus environment promotes calcification of A7R5 cells induced by hydroxyapatite nanoparticles. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 107:110228. [PMID: 31761154 DOI: 10.1016/j.msec.2019.110228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/28/2019] [Accepted: 09/18/2019] [Indexed: 12/19/2022]
Abstract
This study simulated the high-phosphorus (Pi) environment in patients with chronic kidney disease. Nano-hydroxyapatite (HAP) crystals were used to damage rat aortic smooth muscle cells (A7R5) pre-damaged with different concentrations of Pi solution to compare the differences in HAP-induced calcification in A7R5 cells before and after injury by high-Pi condition. After the A7R5 cells were damaged by high-Pi environment, the following were observed. HAP resulted in declined cell viability and lysosomal integrity, release of lactate dehydrogenase, and increased reactive oxygen species production. The ability of high-Pi damaged cells to internalize HAP crystals declined; crystal adhesion and calcium deposition on the cell surface and alkaline phosphatase activities increased. Osteopontin expression and level of Runt-related transcription factor 2 were increased, and HAP-induced osteogenic transformation was enhanced. High-Pi condition promoted the adhesion of A7R5 cells to nano-HAP crystals and inhibited HAP endocytosis, increasing the risk of calcification.
Collapse
Affiliation(s)
- Hong Liu
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou, 510632, China
| | - Ling-Hong Huang
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou, 510632, China
| | - Xin-Yuan Sun
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou, 510632, China
| | - Jian-Ming Ouyang
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
21
|
Huang XQ, Cen X, Sun WT, Xia K, Yu LY, Liu J, Zhao ZH. CircPOMT1 and circMCM3AP inhibit osteogenic differentiation of human adipose-derived stem cells by targeting miR-6881-3p. Am J Transl Res 2019; 11:4776-4788. [PMID: 31497198 PMCID: PMC6731423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 07/07/2019] [Indexed: 06/10/2023]
Abstract
Circular RNAs (circRNAs), novel endogenous non-coding RNAs with the special circular structure, have been found to play critical roles in various development of tissues and diseases. However, few studies have focused on the functions and mechanisms of circRNAs in the osteogenesis of human adipose-derived stem cells (hASCs). Here, we performed the circRNAs sequencing and bioinformatic analysis to investigate the expression profiles of hASCs during osteogenic differentiation. There were 150 upregulated circRNAs and 60 downregulated circRNAs expressed differentially. Among them, the expression of circPOMT1 and circMCM3AP were downregulated during the osteogenesis of hASCs. hsa-miR-6881-3p could promote the osteogenic differentiation of hASCs, while the expression of circPOMT1 and circMCM3AP were negatively correlated with it. Smad6 and Chordin, critical inhibitors of the BMPs signaling pathway, were predicted to be the targets of hsa-miR-6881-3p. Therefore, circPOMT1 and circMCM3AP might influence the osteogenic differentiation of hASCs by targeting hsa-miR-6881-3p via BMPs signaling pathway. CircPOMT1 and circMCM3AP are potential novel targets for the repairment of bone defects.
Collapse
Affiliation(s)
- Xin-Qi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityNo. 14, 3rd Section, South Renmin Road, Chengdu 610041, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan UniversityChengdu, China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityNo. 14, 3rd Section, South Renmin Road, Chengdu 610041, Sichuan, China
- Department of Temporomandibular Joint, West China Hospital of Stomatology, Sichuan UniversityChengdu, China
| | - Wen-Tian Sun
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityNo. 14, 3rd Section, South Renmin Road, Chengdu 610041, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan UniversityChengdu, China
| | - Kai Xia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityNo. 14, 3rd Section, South Renmin Road, Chengdu 610041, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan UniversityChengdu, China
| | - Li-Yuan Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityNo. 14, 3rd Section, South Renmin Road, Chengdu 610041, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan UniversityChengdu, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityNo. 14, 3rd Section, South Renmin Road, Chengdu 610041, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan UniversityChengdu, China
| | - Zhi-He Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityNo. 14, 3rd Section, South Renmin Road, Chengdu 610041, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan UniversityChengdu, China
| |
Collapse
|
22
|
3D Printing PLA/Gingival Stem Cells/ EVs Upregulate miR-2861 and -210 during Osteoangiogenesis Commitment. Int J Mol Sci 2019; 20:ijms20133256. [PMID: 31269731 PMCID: PMC6651609 DOI: 10.3390/ijms20133256] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/14/2019] [Accepted: 06/27/2019] [Indexed: 02/07/2023] Open
Abstract
Bone tissue regeneration strategies require approaches that provide an osteogenic and angiogenic microenvironment able to drive the bone growth. Recently, the development of 3D printing biomaterials, including poly(lactide) (3D-PLA), enriched with mesenchymal stem cells (MSCs) and/or their derivatives, such as extracellular vesicles (EVs) has been achieving promising results. In this study, in vitro results showed an increased expression of osteogenic and angiogenic markers, as RUNX2, VEGFA, OPN and COL1A1 in the living construct 3D-PLA/human Gingival MSCs (hGMSCs)/EVs. Considering that EVs carry and transfer proteins, mRNA and microRNA into target cells, we evaluated miR-2861 and miR-210 expression related to osteoangiogenesis commitment. Histological examination of rats implanted with 3D-PLA/hGMSCs/EVs evidenced the activation of bone regeneration and of the vascularization process, confirmed also by MicroCT. In synthesis, an upregulation of miR-2861 and -210 other than RUNX2, VEGFA, OPN and COL1A1 was evident in cells cultured in the presence of the biomaterial and EVs. Then, these results evidenced that EVs may enhance bone regeneration in calvaria defects, in association with an enhanced vascularization offering a novel regulatory system in the osteoangiogenesis evolution. The application of new strategies to improve biomaterial engraftment is of great interest in the regenerative medicine and can represent a way to promote bone regeneration.
Collapse
|
23
|
Hu Y, Xu R, Chen CY, Rao SS, Xia K, Huang J, Yin H, Wang ZX, Cao J, Liu ZZ, Tan YJ, Luo J, Xie H. Extracellular vesicles from human umbilical cord blood ameliorate bone loss in senile osteoporotic mice. Metabolism 2019; 95:93-101. [PMID: 30668962 DOI: 10.1016/j.metabol.2019.01.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Senile osteoporosis is one of the most common age-related diseases worldwide. Accumulating evidences have indicated that young blood can reverse age-related impairments. Extracellular vesicles (EVs) exert therapeutic effects in a variety of diseases by delivering bioactive molecules such as microRNAs (miRNAs). The aim of the study is to evaluate the therapeutic potential of EVs from human umbilical cord blood plasma (UCB-EVs) on senile osteoporosis and to preliminarily clarify the underlying mechanism. METHODS UCB-EVs were injected into the tail vein of aged (16 months old) male C57BL/6 mice. Microcomputed tomography was performed to evaluate bone mass and microarchitecture of mice. The osteogenic and osteoclastic activities were determined by quantitative real-time PCR (qRT-PCR), histological examination and western blot analysis. In vitro, qRT-PCR assay was undertaken to explore the enrichment levels of a number of miRNAs that have positive effects in reducing bone loss. The efficacy of UCB-EVs on osteoblastic differentiation of bone marrow mesenchymal stromal cells (BMSCs) and osteoclastogenesis of RAW264.7 cells were assessed by cytochemical staining. Gene and protein expression changes were detected by qRT-PCR and western blotting respectively. Meanwhile, the roles of the selected miRNA in the regulatory effects of UCB-EVs on BMSCs and RAW264.7 cells were evaluated by using specific miRNA inhibitor. RESULTS The intravenous injection of UCB-EVs for two months attenuated bone loss in old mice, as defined by increased trabecular and cortical bone mass, enhanced osteoblast formation and reduced osteoclast formation compared to the control mice. In vitro, UCB-EVs could promote the osteogenic differentiation of BMSCs and inhibit the osteoclastogenesis of RAW264.7 cells. Moreover, it was confirmed that miR-3960 was highly enriched in UCB-EVs and miR-3960 inhibitor reversed the stimulatory effect of UCB-EVs on osteoblastic differentiation of BMSCs. CONCLUSION Our findings indicate that UCB-EVs ameliorate age-related bone loss by stimulating bone formation and inhibiting bone resorption, and miR-3960 mediated the osteogenic effect of UCB-EVs on BMSCs. Thus, UCB-EVs may represent a promising agent for prevention of senile osteoporosis.
Collapse
Affiliation(s)
- Yin Hu
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ran Xu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Chun-Yuan Chen
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shan-Shan Rao
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Xiangya Nursing School, Central South University, Changsha, Hunan 410013, China
| | - Kun Xia
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jie Huang
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hao Yin
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhen-Xing Wang
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jia Cao
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zheng-Zhao Liu
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yi-Juan Tan
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Juan Luo
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hui Xie
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan Key Laboratory of Organ Injury, Aging and Regenerative Medicine, Changsha, Hunan 410008, China; Hunan Key Laboratory of Bone Joint Degeneration and Injury, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
24
|
Zhang B, Miller VM, Miller JD. Influences of Sex and Estrogen in Arterial and Valvular Calcification. Front Endocrinol (Lausanne) 2019; 10:622. [PMID: 31620082 PMCID: PMC6763561 DOI: 10.3389/fendo.2019.00622] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/27/2019] [Indexed: 01/14/2023] Open
Abstract
Vascular and cardiac valvular calcification was once considered to be a degenerative and end stage product in aging cardiovascular tissues. Over the past two decades, however, a critical mass of data has shown that cardiovascular calcification can be an active and highly regulated process. While the incidence of calcification in the coronary arteries and cardiac valves is higher in men than in age-matched women, a high index of calcification associates with increased morbidity, and mortality in both sexes. Despite the ubiquitous portending of poor outcomes in both sexes, our understanding of mechanisms of calcification under the dramatically different biological contexts of sex and hormonal milieu remains rudimentary. Understanding how the critical context of these variables inform our understanding of mechanisms of calcification-as well as innovative strategies to target it therapeutically-is essential to advancing the fields of both cardiovascular disease and fundamental mechanisms of aging. This review will explore potential sex and sex-steroid differences in the basic biological pathways associated with vascular and cardiac valvular tissue calcification, and potential strategies of pharmacological therapy to reduce or slow these processes.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Virginia M. Miller
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Jordan D. Miller
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, MN, United States
- *Correspondence: Jordan D. Miller
| |
Collapse
|
25
|
Jiang S, Tan B, Zhang X. Identification of key lncRNAs in the carcinogenesis and progression of colon adenocarcinoma by co-expression network analysis. J Cell Biochem 2018; 120:6490-6501. [PMID: 30430631 DOI: 10.1002/jcb.27940] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/02/2018] [Indexed: 12/25/2022]
Abstract
Colon adenocarcinoma (COAD) is one of the most common cancers, and its carcinogenesis and progression is influenced by multiple long non-coding RNAs (lncRNA), especially through the miRNA sponge effect. In this study, more than 4000 lncRNAs were re-annotated from the microarray datasets through probe sequence mapping to obtain reliable lncRNA expression profiles. As a systems biology method for describing the correlation patterns among genes across microarray samples, weighted gene co-expression network analysis was conducted to identify lncRNA modules associated with the five stepwise stages from normal colonic samples to COAD (n = 94). In the most relevant module (R2 = -0.78, P = 4E-20), four hub lncRNAs were identified (CTD-2396E7.11, PCGF5, RP11-33O4.1, and RP11-164P12.5). Then, these four hub lncRNAs were validated using two other independent datasets including GSE20916 (n = 145) and GSE39582 (n = 552). The results indicated that all hub lncRNAs were significantly negatively correlated with the three-stage colonic carcinogenesis, as well as TNM stages in COAD (one-way analysis of variance P < 0.05). Kaplan-Meier survival curve showed that patients with higher expression of each hub lncRNA had a significantly higher overall survival rate and lower relapse risk (log-rank P < 0.05). In conclusion, through co-expression analysis, we identified and validated four key lncRNAs in association with the carcinogenesis and progression of COAD, and these lncRNAs might have important clinical implications for improving the risk stratification, therapeutic decision and prognosis prediction in COAD patients.
Collapse
Affiliation(s)
- Shi Jiang
- Department of Gastroenterology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, China
| | - Biyong Tan
- Department of Radiology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, China
| | - Xingqiang Zhang
- Department of Radiology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, China
| |
Collapse
|
26
|
Wei C, Huang H, Cong W, Li Z, Zhang X, Liu H, Wang R, Xiao J. Identification of the Differentially Expressed microRNAs Involved in Cleft Palate Induced by Retinoic Acid (RA) in Mouse Model. J HARD TISSUE BIOL 2018. [DOI: 10.2485/jhtb.27.243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Chao Wei
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Haitao Huang
- Department of Stomatology, the First Affiliated Hospital, Dalian Medical University
| | - Wei Cong
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Zhiguang Li
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University
| | - Xuehong Zhang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University
| | - Han Liu
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Ru Wang
- Department of Stomatology, the First Affiliated Hospital, Dalian Medical University
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Dalian Medical University
| | - Jing Xiao
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| |
Collapse
|
27
|
Zhang C, Zhang K, Huang F, Feng W, Chen J, Zhang H, Wang J, Luo P, Huang H. Exosomes, the message transporters in vascular calcification. J Cell Mol Med 2018; 22:4024-4033. [PMID: 29892998 PMCID: PMC6111818 DOI: 10.1111/jcmm.13692] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 04/18/2018] [Indexed: 12/17/2022] Open
Abstract
Vascular calcification (VC) is caused by hydroxyapatite deposition in the intimal and medial layers of the vascular wall, leading to severe cardiovascular events in patients with hypertension, chronic kidney disease and diabetes mellitus. VC occurrences involve complicated mechanism networks, such as matrix vesicles or exosomes production, osteogenic differentiation, reduced cell viability, aging and so on. However, with present therapeutic methods targeting at VC ineffectively, novel targets for VC treatment are demanded. Exosomes are proven to participate in VC and function as initializers for mineral deposition. Secreted exosomes loaded with microRNAs are also demonstrated to modulate VC procession in recipient vascular smooth muscle cells. In this review, we targeted at the roles of exosomes during VC, especially at their effects on transporting biological information among cells. Moreover, we will discuss the potential mechanisms of exosomes in VC.
Collapse
Affiliation(s)
- Chao Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Kun Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Feifei Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Weijing Feng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Jie Chen
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China.,Department of Radiation Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huanji Zhang
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jingfeng Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Pei Luo
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Hui Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| |
Collapse
|
28
|
Schurgers LJ, Akbulut AC, Kaczor DM, Halder M, Koenen RR, Kramann R. Initiation and Propagation of Vascular Calcification Is Regulated by a Concert of Platelet- and Smooth Muscle Cell-Derived Extracellular Vesicles. Front Cardiovasc Med 2018; 5:36. [PMID: 29682509 PMCID: PMC5897433 DOI: 10.3389/fcvm.2018.00036] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/26/2018] [Indexed: 12/20/2022] Open
Abstract
The ageing population continues to suffer from its primary killer, cardiovascular disease (CVD). Despite recent advances in interventional medicinal and surgical therapies towards the end of the 20th century, the epidemic of cardiovascular disease has not been halted. Yet, rather than receding globally, the burden of CVD has risen to become a top cause of morbidity and mortality worldwide. Most CVD arises from thrombotic rupture of an atherosclerotic plaque, the pathologic thickening of coronary and carotid artery segments and subsequent distal ischemia in heart or brain. In fact, one-fifth of deaths are directly attributable to thrombotic rupture of a vulnerable plaque. Atherosclerotic lesion formation is caused by a concert of interactions between circulating leukocytes and platelets, interacting with the endothelial barrier, signalling into the arterial wall by the release of cytokines and extracellular vesicles (EVs). Both platelet- and cell-derived EVs represent a novel mechanism of cellular communication, particularly by the transport and transfer of cargo and by reprogramming of the recipient cell. These interactions result in phenotypic switching of vascular smooth muscle cells (VSMCs) causing migration and proliferation, and subsequent secretion of EVs. Loss of VSMCs attracts perivascular Mesenchymal Stem Cells (MSCs) from the adventitia, which are a source of VSMCs and contribute to repair after vascular injury. However, continuous stress stimuli eventually switch phenotype of cells into osteochondrogenic VSMCs facilitating vascular calcification. Although Virchow’s triad is over 100 years old, it is a reality that is accurate today. It can be briefly summarised as changes in the composition of blood (platelet EVs), alterations in the vessel wall (VSMC phenotypic switching, MSC infiltration and EV release) and disruption of blood flow (atherothrombosis). In this paper, we review the latest relevant advances in the identification of extracellular vesicle pathways as well as VSMCs and pericyte/MSC phenotypic switching, underlying vascular calcification.
Collapse
Affiliation(s)
- Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Asim C Akbulut
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Dawid M Kaczor
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Maurice Halder
- Division of Nephrology, RWTH Aachen University, Aachen, Germany
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Rafael Kramann
- Division of Nephrology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
29
|
Zhao M, Li L, Zhou J, Cui X, Tian Q, Jin Y, Zhu Y. MiR-2861 Behaves as a Biomarker of Lung Cancer Stem Cells and Regulates the HDAC5-ERK System Genes. Cell Reprogram 2018; 20:99-106. [PMID: 29620443 DOI: 10.1089/cell.2017.0045] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cancer stem cells (CSCs) are responsible for cancer initiating, recurrence, and drug resistance. Discovery of novel biomarkers for CSCs is helpful for early diagnosis and prognosis. Lung cancer stem cells (LCSCs) were closely related to the occurrence and development of lung cancer. In our study, the important role of miR-2861 in maintaining the stemness of LCSCs was investigated. The LCSC differentiation model was established through introducing serum into the medium of H460 spheres. miR-2861 expression was significantly higher in LCSCs no matter compared to the differentiation cells or normal cells. HDAC5 expression was positively correlated with miR-2861 in LCSCs, and knockdown of miR-2861 decreased the expression of HDAC5, which implied that HDAC5 may be involved in the differentiation of LCSCs mediated by miR-2861. The role of HDAC5 in the regulation of LCSC differentiation was further verified by the inhibitory effect of LMK-235 on the phosphorylation of ERK1/2, which was recognized as the regulator of CSC differentiation. Our study provided a better understanding of miR-2861 and HDAC5 axis in maintaining the stemness of LCSCs and laid a foundation for molecular targeted therapy.
Collapse
Affiliation(s)
- Mengya Zhao
- 1 CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences, Suzhou, China .,2 College of Life Sciences, Shanghai University , Shanghai, China
| | - Lin Li
- 1 CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences, Suzhou, China
| | - Jundong Zhou
- 3 Department of Radio Oncology, Affiliated Suzhou Hospital, Nanjing Medical University , Suzhou, China
| | - Xueyuan Cui
- 1 CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences, Suzhou, China .,2 College of Life Sciences, Shanghai University , Shanghai, China
| | - Qingmei Tian
- 1 CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences, Suzhou, China .,4 School of Pharmacy, Xi'an Jiaotong University , Xi'an, China
| | - Yaqing Jin
- 1 CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences, Suzhou, China .,5 University of Chinese Academy of Sciences , Beijing, China
| | - Yimin Zhu
- 1 CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences, Suzhou, China
| |
Collapse
|
30
|
Long Noncoding RNA Sponges miR-454 to Promote Osteogenic Differentiation in Maxillary Sinus Membrane Stem Cells. IMPLANT DENT 2018; 26:178-186. [PMID: 28301382 DOI: 10.1097/id.0000000000000569] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Although increasing evidence has shown that long noncoding RNAs (lncRNAs) play an important regulatory role in pluripotency and differentiation of mesenchymal stem cells, little is known about the role of lncRNA in maxillary sinus membrane stem cells (MSMSCs). The goal of this study was to investigate the expression profile and function of lncRNAs on osteogenic differentiation of MSMSCs. MATERIALS AND METHODS By using lncRNA microarray, we identify a novel osteogenesis differentiation-related lncRNA of MSMSCs (lncRNA-MODR). The functional role of lncRNA-MODR in regulating osteogenesis was evaluated by quantitative real-time polymerase chain reaction, western blot, and alizarin red staining. Bioinformatic analyses of the predicted target genes (gene ontology, pathway, and network analysis) were applied for further study of lncRNA-MODR. RESULTS We show that lncRNA-MODR is gradually upregulated during osteogenic differentiation. lncRNA-MODR overexpression upregulated, whereas lncRNA-MODR silencing decreased the expression of the osteogenic key marker, runt-related transcription factor 2 (RUNX2). In-depth analyses showed that lncRNA-MODR acts as a molecular sponge for microRNA-454 (miR-454) and that prevents RUNX2 from mi-454-mediated suppression. CONCLUSION The lncRNAs act as a competing endogenous RNA to sequester microRNA-454 (miR-454), leading to heightened RUNX2 expression and thus promotes osteogenesis of MSMSCs.
Collapse
|
31
|
Nanoudis S, Pikilidou M, Yavropoulou M, Zebekakis P. The Role of MicroRNAs in Arterial Stiffness and Arterial Calcification. An Update and Review of the Literature. Front Genet 2017; 8:209. [PMID: 29312437 PMCID: PMC5733083 DOI: 10.3389/fgene.2017.00209] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 11/28/2017] [Indexed: 12/20/2022] Open
Abstract
Arterial stiffness is an independent risk factor for fatal and non-fatal cardiovascular events, such as systolic hypertension, coronary artery disease, stroke, and heart failure. Moreover it reflects arterial aging which in many cases does not coincide with chronological aging, a fact that is in large attributed to genetic factors. In addition to genetic factors, microRNAs (miRNAs) seem to largely affect arterial aging either by advancing or by regressing arterial stiffness. MiRNAs are small RNA molecules, ~22 nucleotides long that can negatively control their target gene expression posttranscriptionally. Pathways that affect main components of stiffness such as fibrosis and calcification seem to be influenced by up or downregulation of specific miRNAs. Identification of this aberrant production of miRNAs can help identify epigenetic changes that can be therapeutic targets for prevention and treatment of vascular diseases. The present review summarizes the specific role of the so far discovered miRNAs that are involved in pathways of arterial stiffness.
Collapse
Affiliation(s)
- Sideris Nanoudis
- Hypertension Excellence Center, 1st Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Maria Pikilidou
- Hypertension Excellence Center, 1st Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Maria Yavropoulou
- Division of Endocrinology and Metabolism, AHEPA University Hospital, Thessaloniki, Greece
| | - Pantelis Zebekakis
- Hypertension Excellence Center, 1st Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
32
|
Liu Z, Chen T, Han Q, Chen M, You J, Fang F, Peng L, Wu B. HDAC inhibitor LMK‑235 promotes the odontoblast differentiation of dental pulp cells. Mol Med Rep 2017; 17:1445-1452. [PMID: 29138868 PMCID: PMC5780081 DOI: 10.3892/mmr.2017.8055] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 08/18/2017] [Indexed: 01/07/2023] Open
Abstract
The role of dental pulp cells (DPCs) in hard dental tissue regeneration had received increasing attention because DPCs can differentiate into odontoblasts and other tissue‑specific cells. In recent years, epigenetic modifications had been identified to serve an important role in cell differentiation, and histone deacetylase (HDAC) inhibitors have been widely studied by many researchers. However, the effects of HDAC4 and HDAC5 on the differentiation of DPCs and the precise molecular mechanisms remain unclear. The present study demonstrated that LMK‑235, a specific human HDAC4 and HDAC5 inhibitor, increased the expression of specific odontoblastic gene expression levels detected by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) in dental pulp cells, and did not reduce cell proliferation tested by MTT assay after 3 days in culture at a low concentration. In addition, the mRNA and protein expression levels of dentin sialophosphoprotein, runt‑related transcription factor 2, alkaline phosphatase (ALP) and osteocalcin were evaluated by RT‑qPCR and western blotting, respectively. The increased gene and protein expression of specific markers demonstrated, indicating that LMK‑235 promoted the odontoblast induction of DPCs. ALP activity and mineralised nodule formation were also enhanced due to the effect of LMK‑235, detected by an ALP activity test and Alizarin Red S staining, respectively. Additionally, the vascular endothelial growth factor (VEGF)/RAC‑gamma serine/threonine‑protein kinase (AKT)/mechanistic target of rapamycin (mTOR) signalling pathway was tested to see if it takes part in the differentiation of DPCs treated with LMK‑235, and it was demonstrated that the mRNA expression levels of VEGF, AKT and mTOR were upregulated. These findings indicated that LMK‑235 may serve a key role in the proliferation and odontoblast differentiation of DPCs, and could be used to accelerate dental tissue regeneration.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ting Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qianqian Han
- Department of Periodontics, Stomatology Hospital of Guangdong Province, Guangzhou, Guangdong 510260, P.R. China
| | - Ming Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jie You
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Fuchun Fang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ling Peng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Buling Wu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
33
|
Gošev I, Zeljko M, Đurić Ž, Nikolić I, Gošev M, Ivčević S, Bešić D, Legčević Z, Paić F. Epigenome alterations in aortic valve stenosis and its related left ventricular hypertrophy. Clin Epigenetics 2017; 9:106. [PMID: 29026447 PMCID: PMC5627415 DOI: 10.1186/s13148-017-0406-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022] Open
Abstract
Aortic valve stenosis is the most common cardiac valve disease, and with current trends in the population demographics, its prevalence is likely to rise, thus posing a major health and economic burden facing the worldwide societies. Over the past decade, it has become more than clear that our traditional genetic views do not sufficiently explain the well-known link between AS, proatherogenic risk factors, flow-induced mechanical forces, and disease-prone environmental influences. Recent breakthroughs in the field of epigenetics offer us a new perspective on gene regulation, which has broadened our perspective on etiology of aortic stenosis and other aortic valve diseases. Since all known epigenetic marks are potentially reversible this perspective is especially exciting given the potential for development of successful and non-invasive therapeutic intervention and reprogramming of cells at the epigenetic level even in the early stages of disease progression. This review will examine the known relationships between four major epigenetic mechanisms: DNA methylation, posttranslational histone modification, ATP-dependent chromatin remodeling, and non-coding regulatory RNAs, and initiation and progression of AS. Numerous profiling and functional studies indicate that they could contribute to endothelial dysfunctions, disease-prone activation of monocyte-macrophage and circulatory osteoprogenitor cells and activation and osteogenic transdifferentiation of aortic valve interstitial cells, thus leading to valvular inflammation, fibrosis, and calcification, and to pressure overload-induced maladaptive myocardial remodeling and left ventricular hypertrophy. This is especcialy the case for small non-coding microRNAs but was also, although in a smaller scale, convincingly demonstrated for other members of cellular epigenome landscape. Equally important, and clinically most relevant, the reported data indicate that epigenetic marks, particularly certain microRNA signatures, could represent useful non-invasive biomarkers that reflect the disease progression and patients prognosis for recovery after the valve replacement surgery.
Collapse
Affiliation(s)
- Igor Gošev
- Department of Surgery, University of Rochester Medical center, Rochester, NY USA
| | - Martina Zeljko
- Department of Cardiology, Clinical Unit of Internal Medicine, Clinical Hospital Merkur, Zajćeva 19, 10 000 Zagreb, Croatia
| | - Željko Đurić
- Department of Cardiac Surgery, University Hospital Center Zagreb, Kišpatićeva 12, 10 000 Zagreb, Croatia
| | - Ivana Nikolić
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115 USA
| | - Milorad Gošev
- School of Medicine, University of Josip Juraj Strossmayer, Trg Svetog trojstva 3, 31 000 Osijek, Croatia
| | - Sanja Ivčević
- Department of Physiology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| | - Dino Bešić
- Laboratory for Epigenetics and Molecular Medicine, Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| | - Zoran Legčević
- Laboratory for Epigenetics and Molecular Medicine, Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| | - Frane Paić
- Laboratory for Epigenetics and Molecular Medicine, Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| |
Collapse
|
34
|
Mellows B, Mitchell R, Antonioli M, Kretz O, Chambers D, Zeuner MT, Denecke B, Musante L, Ramachandra DL, Debacq-Chainiaux F, Holthofer H, Joch B, Ray S, Widera D, David AL, Huber TB, Dengjel J, De Coppi P, Patel K. Protein and Molecular Characterization of a Clinically Compliant Amniotic Fluid Stem Cell-Derived Extracellular Vesicle Fraction Capable of Accelerating Muscle Regeneration Through Enhancement of Angiogenesis. Stem Cells Dev 2017; 26:1316-1333. [PMID: 28679310 DOI: 10.1089/scd.2017.0089] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The secretome of human amniotic fluid stem cells (AFSCs) has great potential as a therapeutic agent in regenerative medicine. However, it must be produced in a clinically compliant manner before it can be used in humans. In this study, we developed a means of producing a biologically active secretome from AFSCs that is free of all exogenous molecules. We demonstrate that the full secretome is capable of promoting stem cell proliferation, migration, and protection of cells against senescence. Furthermore, it has significant anti-inflammatory properties. Most importantly, we show that it promotes tissue regeneration in a model of muscle damage. We then demonstrate that the secretome contains extracellular vesicles (EVs) that harbor much, but not all, of the biological activity of the whole secretome. Proteomic characterization of the EV and free secretome fraction shows the presence of numerous molecules specific to each fraction that could be key regulators of tissue regeneration. Intriguingly, we show that the EVs only contain miRNA and not mRNA. This suggests that tissue regeneration in the host is mediated by the action of EVs modifying existing, rather than imposing new, signaling pathways. The EVs harbor significant anti-inflammatory activity as well as promote angiogenesis, the latter may be the mechanistic explanation for their ability to promote muscle regeneration after cardiotoxin injury.
Collapse
Affiliation(s)
- Ben Mellows
- 1 School of Biological Sciences, University of Reading , Reading, United Kingdom
| | - Robert Mitchell
- 1 School of Biological Sciences, University of Reading , Reading, United Kingdom
| | - Manuela Antonioli
- 2 Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS 'Lazzaro Spallanzani' , Rome, Italy
| | - Oliver Kretz
- 3 Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf , Hamburg, Germany .,4 Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg , Freiburg, Germany .,5 Department of Medicine IV, Faculty of Medicine, University of Freiburg , Freiburg, Germany
| | - David Chambers
- 6 Wolfson Centre for Age-Related Diseases, King's College , London, United Kingdom
| | | | - Bernd Denecke
- 8 Interdisciplinary Centre for Clinical Research Aachen, RWTH Aachen University , Aachen, Germany
| | - Luca Musante
- 9 Centre for Bioanalytical Sciences (CBAS), Dublin City University , Dublin, Ireland
| | - Durrgah L Ramachandra
- 10 Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health , London, United Kingdom
| | | | - Harry Holthofer
- 9 Centre for Bioanalytical Sciences (CBAS), Dublin City University , Dublin, Ireland .,12 FRIAS Freiburg Institute for Advanced Studies, University of Freiburg , Freiburg, Germany
| | - Barbara Joch
- 5 Department of Medicine IV, Faculty of Medicine, University of Freiburg , Freiburg, Germany
| | - Steve Ray
- 13 Micregen, Biohub, Cheshire, United Kingdom
| | - Darius Widera
- 7 School of Pharmacy, University of Reading , Reading, United Kingdom
| | - Anna L David
- 14 Institute for Women's Health, University College London , London, United Kingdom .,15 NIHR University College London Hospitals Biomedical Research Centre , London, United Kingdom
| | - Tobias B Huber
- 3 Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf , Hamburg, Germany .,4 Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg , Freiburg, Germany .,12 FRIAS Freiburg Institute for Advanced Studies, University of Freiburg , Freiburg, Germany .,16 BIOSS Centre for Biological Signalling Studies and Centre for Systems Biology (ZBSA), Albert-Ludwigs University , Freiburg, Germany
| | - Joern Dengjel
- 12 FRIAS Freiburg Institute for Advanced Studies, University of Freiburg , Freiburg, Germany .,17 Department of Biology, University of Fribourg , Fribourg, Switzerland
| | - Paolo De Coppi
- 10 Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health , London, United Kingdom
| | - Ketan Patel
- 1 School of Biological Sciences, University of Reading , Reading, United Kingdom .,12 FRIAS Freiburg Institute for Advanced Studies, University of Freiburg , Freiburg, Germany
| |
Collapse
|
35
|
Kumar MM, Goyal R. LncRNA as a Therapeutic Target for Angiogenesis. Curr Top Med Chem 2017; 17:1750-1757. [PMID: 27848894 DOI: 10.2174/1568026617666161116144744] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/25/2016] [Accepted: 09/21/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Out of 3 billion base pairs in human genome only ~2% code for proteins; and out of 180,000 transcripts in human cells, about 20,000 code for protein, remaining 160,000 are non-coding transcripts. Most of these transcripts are more than 200 base pairs and constitute a group of long non-coding RNA (lncRNA). Many of the lncRNA have its own promoter, and are well conserved in mammals. Accumulating evidence indicates that lncRNAs act as molecular switches in cellular differentiation, movement, apoptosis, and in the reprogramming of cell states by altering gene expression patterns. However, the role of this important group of molecules in angiogenesis is not well understood. Angiogenesis is a complex process and depends on precise regulation of gene expression. CONCLUSION Dysregulation of transcription during this process may lead to several diseases including various cancers. As angiogenesis is an important process in cancer pathogenesis and treatment, lncRNA may be playing an important role in angiogenesis. In support of this, lncRNA microvascular invasion in hepatocellular carcinoma (MVIH) has been shown to activate angiogenesis. Furthermore, lncRNA-Meg3-knockout mouse showed increased expression of vascular endothelial growth factor pathway genes and increased cortical microvessel density. Overall, there is strong evidence that lncRNA is an important class of regulatory molecule, and a number of studies have demonstrated that these can be targeted to change cellular physiology and functions. In this review, we have attempted to summarize these studies and elucidate the potential of this novel regulatory molecule as a therapeutic target.
Collapse
Affiliation(s)
- Mohan M Kumar
- Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States
| | - Ravi Goyal
- Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, School of Medicine, Loma Linda, CA 92350, United States
| |
Collapse
|
36
|
Tao SC, Yuan T, Zhang YL, Yin WJ, Guo SC, Zhang CQ. Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics 2017; 7:180-195. [PMID: 28042326 PMCID: PMC5196895 DOI: 10.7150/thno.17133] [Citation(s) in RCA: 499] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 10/04/2016] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Osteoarthritis (OA) is the most common joint disease throughout the world. Exosomes derived from miR-140-5p-overexpressing synovial mesenchymal stem cells (SMSC-140s) may be effective in treating OA. We hypothesized that exosomes derived from SMSC-140 (SMSC-140-Exos) would enhance the proliferation and migration abilities of articular chondrocytes (ACs) without harming extracellular matrix (ECM) secretion. METHODS SMSCs were transfected with or without miR-140-5p. Exosomes derived from SMSCs or SMSC-140s (SMSC-Exos or SMSC-140-Exos) were isolated and identified. Proliferation, migration and ECM secretion were measured in vitro and compared between groups. The mechanism involving alternative Wnt signalling and activation of Yes-associated protein (YAP) was investigated using lentivirus, oligonucleotides or chemical drugs. The preventative effect of exosomes in vivo was measured using Safranin-O and Fast green staining and immunohistochemical staining. RESULTS Wnt5a and Wnt5b carried by exosomes activated YAP via the alternative Wnt signalling pathway and enhanced proliferation and migration of chondrocytes with the side-effect of significantly decreasing ECM secretion. Highly-expressed miR-140-5p blocked this side-effect via RalA. SMSC-140-Exos enhanced the proliferation and migration of ACs without damaging ECM secretion in vitro, while in vivo, SMSC-140-Exos successfully prevented OA in a rat model. CONCLUSIONS These findings highlight the promising potential of SMSC-140-Exos in preventing OA. We first found a potential source of exosomes and studied their merits and shortcomings. Based on our understanding of the molecular mechanism, we overcame the shortcomings by modifying the exosomes. Such exosomes derived from modified cells hold potential as future therapeutic strategies.
Collapse
Affiliation(s)
- Shi-Cong Tao
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Ting Yuan
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Yue-Lei Zhang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Wen-Jing Yin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Shang-Chun Guo
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
- ✉ Corresponding authors: Chang-Qing Zhang, Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China. E-mail: . Shang-Chun Guo, Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China. E-mail:
| | - Chang-Qing Zhang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
- ✉ Corresponding authors: Chang-Qing Zhang, Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China. E-mail: . Shang-Chun Guo, Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China. E-mail:
| |
Collapse
|
37
|
Function, Role, and Clinical Application of MicroRNAs in Vascular Aging. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6021394. [PMID: 28097140 PMCID: PMC5209603 DOI: 10.1155/2016/6021394] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/07/2016] [Accepted: 11/23/2016] [Indexed: 01/31/2023]
Abstract
Vascular aging, a specific type of organic aging, is related to age-dependent changes in the vasculature, including atherosclerotic plaques, arterial stiffness, fibrosis, and increased intimal thickening. Vascular aging could influence the threshold, process, and severity of various cardiovascular diseases, thus making it one of the most important risk factors in the high mortality of cardiovascular diseases. As endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are the main cell biological basis of these pathology changes of the vasculature, the structure and function of ECs and VSMCs play a key role in vascular aging. MicroRNAs (miRNAs), small noncoding RNAs, have been shown to regulate the expression of multiple messenger RNAs (mRNAs) posttranscriptionally, contributing to many crucial aspects of cell biology. Recently, miRNAs with functions associated with aging or aging-related diseases have been studied. In this review, we will summarize the reported role of miRNAs in the process of vascular aging with special emphasis on EC and VSMC functions. In addition, the potential application of miRNAs to clinical practice for the diagnosis and treatment of cardiovascular diseases will also be discussed.
Collapse
|
38
|
Budak F, Bal SH, Tezcan G, Guvenc F, Akalin EH, Goral G, Deniz G, Oral HB. MicroRNA Expression Patterns of CD8+ T Cells in Acute and Chronic Brucellosis. PLoS One 2016; 11:e0165138. [PMID: 27824867 PMCID: PMC5100978 DOI: 10.1371/journal.pone.0165138] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/06/2016] [Indexed: 01/18/2023] Open
Abstract
Although our knowledge about Brucella virulence factors and the host response increase rapidly, the mechanisms of immune evasion by the pathogen and causes of chronic disease are still unknown. Here, we aimed to investigate the immunological factors which belong to CD8+ T cells and their roles in the transition of brucellosis from acute to chronic infection. Using miRNA microarray, more than 2000 miRNAs were screened in CD8+ T cells of patients with acute or chronic brucellosis and healthy controls that were sorted from peripheral blood with flow cytometry and validated through qRT-PCR. Findings were evaluated using GeneSpring GX (Agilent) 13.0 software and KEGG pathway analysis. Expression of two miRNAs were determined to display a significant fold change in chronic group when compared with acute or control groups. Both miRNAs (miR-126-5p and miR-4753-3p) were decreased (p <0.05 or fold change > 2). These miRNAs have the potential to be the regulators of CD8+ T cell-related marker genes for chronic brucellosis infections. The differentially expressed miRNAs and their predicted target genes are involved in MAPK signaling pathway, cytokine-cytokine receptor interactions, endocytosis, regulation of actin cytoskeleton, and focal adhesion indicating their potential roles in chronic brucellosis and its progression. It is the first study of miRNA expression analysis of human CD8+ T cells to clarify the mechanism of inveteracy in brucellosis.
Collapse
Affiliation(s)
- Ferah Budak
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - S. Haldun Bal
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Gulcin Tezcan
- Department of Medical Biology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Furkan Guvenc
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - E. Halis Akalin
- Department of Clinical Microbiology and Infection Diseases, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Guher Goral
- Department of Medical Microbiology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Gunnur Deniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - H. Barbaros Oral
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
- * E-mail:
| |
Collapse
|
39
|
Brun J, Abruzzese T, Rolauffs B, Aicher WK, Hart ML. Choice of xenogenic-free expansion media significantly influences the myogenic differentiation potential of human bone marrow-derived mesenchymal stromal cells. Cytotherapy 2016; 18:344-59. [PMID: 26857228 DOI: 10.1016/j.jcyt.2015.11.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/16/2015] [Accepted: 11/25/2015] [Indexed: 01/29/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) have great potential for use in cell-based therapies for restoration of structure and function of many tissue types including smooth muscle. METHODS We compared proliferation, immunophenotype, differentiation capability and gene expression of bone marrow-derived MSCs expanded in different media containing human serum, plasma and platelet lysate in combination with commonly used protocols for myogenic, osteogenic, chondrogenic and adipogenic differentiation. Moreover, we developed a xenogenic-free protocol for myogenic differentiation of MSCs. RESULTS Expansion of MSCs in media complemented with serum, serum + platelet lysate or plasma + platelet lysate were multipotent because they differentiated toward four mesenchymal (myogenic, osteogenic, chondrogenic, adipogenic) lineages. Addition of platelet lysate to expansion media increased the proliferation of MSCs and their expression of CD146. Incubation of MSCs in medium containing human serum or plasma plus 5% human platelet lysate in combination with smooth muscle cell (SMC)-inducing growth factors TGFβ1, PDGF and ascorbic acid induced high expression of ACTA2, TAGLN, CNN1 and/or MYH11 contractile SMC markers. Osteogenic, adipogenic and chondrogenic differentiations served as controls. DISCUSSION Our study provides novel data on the myogenic differentiation potential of human MSCs toward the SMC lineage using different xenogenic-free cell culture expansion media in combination with distinct differentiation medium compositions. We show that the choice of expansion medium significantly influences the differentiation potential of human MSCs toward the smooth muscle cell, as well as osteogenic, adipogenic and chondrogenic lineages. These results can aid in designing studies using MSCs for tissue-specific therapeutic applications.
Collapse
Affiliation(s)
- Juliane Brun
- Clinical Research Group KFO 273, Department of Urology, University of Tübingen, Tübingen, Germany
| | - Tanja Abruzzese
- Clinical Research Group KFO 273, Department of Urology, University of Tübingen, Tübingen, Germany
| | - Bernd Rolauffs
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tübingen, Tübingen, Germany
| | - Wilhelm K Aicher
- Clinical Research Group KFO 273, Department of Urology, University of Tübingen, Tübingen, Germany
| | - Melanie L Hart
- Clinical Research Group KFO 273, Department of Urology, University of Tübingen, Tübingen, Germany; Siegfried Weller Institute for Trauma Research, BG Trauma Clinic Tuebingen, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
40
|
Ballantyne MD, McDonald RA, Baker AH. lncRNA/MicroRNA interactions in the vasculature. Clin Pharmacol Ther 2016; 99:494-501. [PMID: 26910520 PMCID: PMC4881297 DOI: 10.1002/cpt.355] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/16/2016] [Indexed: 12/14/2022]
Abstract
MicroRNA (miRNA) have gained widespread attention for their role in diverse vascular processes including angiogenesis, apoptosis, proliferation, and migration. Despite great understanding of miRNA expression and function, knowledge of long noncoding RNA (lncRNA) molecular mechanisms still remains limited. The influence of miRNA on lncRNA function, and the converse, is now beginning to emerge. lncRNA may regulate miRNA function by acting as endogenous sponges to regulate gene expression and miRNA have been shown to bind and regulate lncRNA stability. A detailed understanding of the molecular and cellular effects of lncRNA‐miRNA‐mediated interactions in vascular pathophysiology could pave the way for new diagnostic markers and therapeutic approaches, but first there is a requirement for a more detailed understanding of the impact of such regulatory networks.
Collapse
Affiliation(s)
- M D Ballantyne
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK.,Institute of Cardiovascular and Medical Science, British Heart Foundation Glasgow, Glasgow, UK
| | - R A McDonald
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - A H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| |
Collapse
|
41
|
The Regulatory Roles of MicroRNAs in Bone Remodeling and Perspectives as Biomarkers in Osteoporosis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1652417. [PMID: 27073801 PMCID: PMC4814634 DOI: 10.1155/2016/1652417] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 02/26/2016] [Accepted: 02/29/2016] [Indexed: 02/05/2023]
Abstract
MicroRNAs are involved in many cellular and molecular activities and played important roles in many biological and pathological processes, such as tissue formation, cancer development, diabetes, neurodegenerative diseases, and cardiovascular diseases. Recently, it has been reported that microRNAs can modulate the differentiation and activities of osteoblasts and osteoclasts, the key cells that are involved in bone remodeling process. Meanwhile, the results from our and other research groups showed that the expression profiles of microRNAs in the serum and bone tissues are significantly different in postmenopausal women with or without fractures compared to the control. Therefore, it can be postulated that microRNAs might play important roles in bone remodeling and that they are very likely to be involved in the pathological process of postmenopausal osteoporosis. In this review, we will present the updated research on the regulatory roles of microRNAs in osteoblasts and osteoclasts and the expression profiles of microRNAs in osteoporosis and osteoporotic fracture patients. The perspective of serum microRNAs as novel biomarkers in bone loss disorders such as osteoporosis has also been discussed.
Collapse
|
42
|
Szulc P. Abdominal aortic calcification: A reappraisal of epidemiological and pathophysiological data. Bone 2016; 84:25-37. [PMID: 26688274 DOI: 10.1016/j.bone.2015.12.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/30/2015] [Accepted: 12/09/2015] [Indexed: 12/16/2022]
Abstract
In men and women, there is a significant association between the risk of cardiovascular event (myocardial infarction, stroke) and risk of major fragility fracture (hip, vertebra). Abdominal aortic calcification (AAC) can be assessed using semiquantitative scores on spine radiographs and spine scans obtained by DXA. Severe AAC is associated with higher risk of major cardiovascular event. Not only does severe AAC reflect poor cardiovascular health status, but also directly disturbs blood flow in the vascular system. Severe (but not mild or moderate) AAC is associated with lower bone mineral density (BMD), faster bone loss and higher risk of major fragility fracture. The fracture risk remains increased after adjustment for BMD and other potential risk factors. The association between severe AAC and fracture risk was found in both sexes, mainly in the follow-ups of less than 10years. Many factors contribute to initiation and progression of AAC: lifestyle, co-morbidities, inorganic ions, dyslipidemia, hormones, cytokines (e.g. inflammatory cytokines, RANKL), matrix vesicles, microRNAs, structural proteins (e.g. elastin), vitamin K-dependent proteins, and medications (e.g. vitamin K antagonists). Osteogenic transdifferentiation of vascular smooth muscle cells (VSMC) and circulating osteoprogenitors penetrating into vascular wall plays a major role in the AAC initiation and progression. Vitamin K-dependent proteins protect vascular tunica media against formation of calcified deposits (matrix GLA protein, GLA-rich protein) and against VSMC apoptosis (Gas6). Further studies are needed to investigate clinical utility of AAC for the assessment of fracture and cardiovascular risk at the individual level and develop new medications permitting to prevent AAC progression.
Collapse
Affiliation(s)
- Pawel Szulc
- INSERM UMR 1033, University of Lyon, Hôpital Edouard Herriot, Lyon, France.
| |
Collapse
|