1
|
Yuan X, Wu Y, Zhang Y, Yao B, Zhang L, Liu Z, Ma Q, Ma F. Endocytosis of sialic acid Neu5Gc and elevated p53 contribute to preimplantation embryonic arrest during in vitro fertilization. J Biol Chem 2025:110254. [PMID: 40404013 DOI: 10.1016/j.jbc.2025.110254] [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: 11/21/2024] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/24/2025] Open
Abstract
The persistently low success rate of human in vitro fertilization (IVF) remains a major clinical challenge, despite significant technological advancements. While culture media composition is known to influence outcomes, the specific components affecting embryonic development are poorly understood. In this study, we identify N-glycolylneuraminic acid (Neu5Gc), a foreign sialic acid contaminant prevalent in animal-derived IVF media components, as a micro-environmental factor triggering developmental arrest. We demonstrate that Neu5Gc accumulates in arrested human embryos via endocytosis and upregulates p53, leading to caspase-3/PARP -dependent apoptotic pathways. This mechanism was validated in human embryonic stem cells (hESCs) and zebrafish embryos, with antibody-mediated Neu5Gc neutralization rescuing developmental defects. Our findings not only elucidate a novel pathway contributing to IVF inefficiency but also provide actionable insights for optimizing culture media formulations.
Collapse
Affiliation(s)
- Xin Yuan
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yilun Wu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Bin Yao
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Linyu Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhenqing Liu
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
| | - Qianhong Ma
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Fang Ma
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China; Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Prakash N, Cha Y, Koh WG, Park H, Bello AB, Lee SH. Derivation of Mesenchymal Stem Cells through Sequential Presentation of Growth Factors via Gelatin Microparticles in Pluripotent Stem Cell Spheroids. Biomater Res 2025; 29:0184. [PMID: 40303481 PMCID: PMC12038162 DOI: 10.34133/bmr.0184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 05/02/2025] Open
Abstract
The use of mesenchymal stem cells (MSCs) in regenerative medicine has gained considerable attention in recent years with the development of clinically relevant MSCs from induced pluripotent stem cells (iPSCs) and embryonic stem cells. Through sequential presentations of appropriate growth factors (GFs), iPSCs can be differentiated into mesodermal cells and then into MSCs. Furthermore, the formation of 3-dimensional cell spheroids, known as embryoid bodies, can be used to mimic in vivo conditions. However, the compact nature of embryoid bodies restricts the efficient and uniform delivery of GFs, leading to the formation of necrotic zones and hindered differentiation. To address this, we developed 2 types of gelatin microparticles (GelMPs) with distinct degradation rates for sequential delivery of GFs to enhance differentiation while preventing necrotic zones. In 2-dimensional culture, bone morphogenetic protein-4 (BMP4) and fibroblast growth factor 2 (FGF2) were identified as key proteins inducing iPSC differentiation into mesodermal cells and MSCs. The sequential presentation of these GFs was optimized for a 3-dimensional culture system by engineering fast-degrading GelMPs conjugated with BMP4 and slow-degrading GelMPs conjugated with FGF2. Our approach facilitated efficient iPSC differentiation into induced mesenchymal stem cells (iMSCs), as demonstrated by enhanced expression of mesodermal markers during the early stages of differentiation and MSC-specific markers at later stages. The resulting iMSCs exhibited characteristic surface markers (e.g., CD73, CD90, CD105, and CD44) and trilineage differentiation capability and were genetically stable. Compared to adult-derived MSCs, iMSCs showed superior proliferative capacity and reduced senescence, making them advantageous for cell therapy and regenerative medicine. This innovative approach of generating iMSCs has vast potential for therapeutic applications.
Collapse
Affiliation(s)
- Nityanand Prakash
- Department of Biomedical Engineering,
Dongguk University, Seoul 04620, Republic of Korea
| | - Young Cha
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry,
Harvard Medical School, Belmont, MA 02478, USA
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering,
Yonsei University, Seoul 03722, Republic of Korea
| | - Hansoo Park
- School of Integrative Engineering,
Chung-Ang University, Seoul 06911, Republic of Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering,
Dongguk University, Seoul 04620, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Engineering,
Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
3
|
Winston T, Song Y, Shi H, Yang J, Alsudais M, Kontaridis MI, Wu Y, Gaborski TR, Meng Q, Cooney RN, Ma Z. Lineage-Specific Mesenchymal Stromal Cells Derived from Human iPSCs Showed Distinct Patterns in Transcriptomic Profile and Extracellular Vesicle Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308975. [PMID: 38757640 PMCID: PMC11267277 DOI: 10.1002/advs.202308975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/16/2024] [Indexed: 05/18/2024]
Abstract
Over the past decades, mesenchymal stromal cells (MSCs) have been extensively investigated as a potential therapeutic cell source for the treatment of various disorders. Differentiation of MSCs from human induced pluripotent stem cells (iMSCs) has provided a scalable approach for the biomanufacturing of MSCs and related biological products. Although iMSCs shared typical MSC markers and functions as primary MSCs (pMSCs), there is a lack of lineage specificity in many iMSC differentiation protocols. Here, a stepwise hiPSC-to-iMSC differentiation method is employed via intermediate cell stages of neural crest and cytotrophoblast to generate lineage-specific MSCs with varying differentiation efficiencies and gene expression. Through a comprehensive comparison between early developmental cell types (hiPSCs, neural crest, and cytotrophoblast), two lineage-specific iMSCs, and six source-specific pMSCs, are able to not only distinguish the transcriptomic differences between MSCs and early developmental cells, but also determine the transcriptomic similarities of iMSC subtypes to postnatal or perinatal pMSCs. Additionally, it is demonstrated that different iMSC subtypes and priming conditions affected EV production, exosomal protein expression, and cytokine cargo.
Collapse
Affiliation(s)
- Tackla Winston
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
| | - Yuanhui Song
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
| | - Huaiyu Shi
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
| | - Junhui Yang
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
| | - Munther Alsudais
- Departments of Biomedical and Chemical EngineeringRochester Institute of TechnologyOne Lomb Memorial DriveRochesterNY14623USA
| | - Maria I. Kontaridis
- Department of Biomedical Research and Translational MedicineMasonic Medical Research Institute2150 Bleecker StreetUticaNY13501USA
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical CenterHarvard Medical School330 Brookline AveBostonMA02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBuilding C, 240 Longwood AveBostonMA02115USA
| | - Yaoying Wu
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
- Department of Microbiology & ImmunologySUNY Upstate Medical University766 Irving AvenueSyracuseNY13210USA
| | - Thomas R. Gaborski
- Departments of Biomedical and Chemical EngineeringRochester Institute of TechnologyOne Lomb Memorial DriveRochesterNY14623USA
| | - Qinghe Meng
- Department of SurgeryState University of New York Upstate Medical University750 East Adams StreetSyracuseNY13210USA
- Sepsis Interdisciplinary Research CenterState University of New York Upstate Medical University766 Irving AvenueSyracuseNY13210USA
| | - Robert N. Cooney
- Department of SurgeryState University of New York Upstate Medical University750 East Adams StreetSyracuseNY13210USA
- Sepsis Interdisciplinary Research CenterState University of New York Upstate Medical University766 Irving AvenueSyracuseNY13210USA
| | - Zhen Ma
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
- Department of BiologySyracuse University107 College PlSyracuseNY13210USA
| |
Collapse
|
4
|
Zhang Q, Lei X, Wang F, He X, Liu L, Hou Y, Liu Y, Jin F, Chen C, Li B. ERK1-mediated immunomodulation of mesenchymal stem cells ameliorates inflammatory disorders. iScience 2023; 26:107868. [PMID: 37790278 PMCID: PMC10543658 DOI: 10.1016/j.isci.2023.107868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 10/05/2023] Open
Abstract
Immune system disorders, especially T cell disorders, are important therapeutic targets of mesenchymal stem cells (MSCs) in many autoimmune diseases (ADs). Although extracellular regulated protein kinases (ERKs) play a role in MSC therapy by promoting T cell apoptosis, the mechanism remains unclear. Our findings indicate that ERK1-/- bone marrow MSCs (BMMSCs), but not ERK2-/- BMMSCs, failed to promote T cell apoptosis due to incapacity to activate the ETS2/AURKA/NF-κB/Fas/MCP-1 cascade. Moreover, ERK1-/- BMMSCs were unable to upregulate regulatory T cells and suppress T helper 17 cells. Licochalcone A (LA), which promotes ERK pathway activation, enhanced the therapeutic efficacy of MSC therapy in ulcerative colitis and collagen-induced arthritis mice. Our findings suggest that ERK1, but not ERK2, plays a crucial role in regulating T cells in MSCs. LA-treated MSCs provide a strategy to improve the efficacy of MSC-based treatments for ADs.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Xiao Lei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Fang Wang
- Department of Blood Purification, General Hospital of Central Theater Command of PLA, 68 Huangpu Road, Wuhan, Hubei 430010, China
| | - Xiaoning He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Lu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yuxia Hou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Yuan Liu
- The Affiliated Northwest Women’s and Children’s Hospital of Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi 710061, China
| | - Fang Jin
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| |
Collapse
|
5
|
Prakash N, Kim J, Jeon J, Kim S, Arai Y, Bello AB, Park H, Lee SH. Progress and emerging techniques for biomaterial-based derivation of mesenchymal stem cells (MSCs) from pluripotent stem cells (PSCs). Biomater Res 2023; 27:31. [PMID: 37072836 PMCID: PMC10114339 DOI: 10.1186/s40824-023-00371-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/26/2023] [Indexed: 04/20/2023] Open
Abstract
The use of mesenchymal stem cells (MSCs) for clinical purposes has skyrocketed in the past decade. Their multilineage differentiation potentials and immunomodulatory properties have facilitated the discovery of therapies for various illnesses. MSCs can be isolated from infant and adult tissue sources, which means they are easily available. However, this raises concerns because of the heterogeneity among the various MSC sources, which limits their effective use. Variabilities arise from donor- and tissue-specific differences, such as age, sex, and tissue source. Moreover, adult-sourced MSCs have limited proliferation potentials, which hinders their long-term therapeutic efficacy. These limitations of adult MSCs have prompted researchers to develop a new method for generating MSCs. Pluripotent stem cells (PSCs), such as embryonic stem cells and induced PSCs (iPSCs), can differentiate into various types of cells. Herein, a thorough review of the characteristics, functions, and clinical importance of MSCs is presented. The existing sources of MSCs, including adult- and infant-based sources, are compared. The most recent techniques for deriving MSCs from iPSCs, with a focus on biomaterial-assisted methods in both two- and three-dimensional culture systems, are listed and elaborated. Finally, several opportunities to develop improved methods for efficiently producing MSCs with the aim of advancing their various clinical applications are described.
Collapse
Affiliation(s)
- Nityanand Prakash
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jiseong Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jieun Jeon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Siyeon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| |
Collapse
|
6
|
Han X, Li B, Zhang S. MIR503HG: A potential diagnostic and therapeutic target in human diseases. Biomed Pharmacother 2023; 160:114314. [PMID: 36736276 DOI: 10.1016/j.biopha.2023.114314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
LncRNAs are involved in many physiological and pathological processes, including chromatin remodeling, transcription, posttranscriptional gene expression, mRNA stability, translation, and posttranslational modification, and their functions depend on subcellular localization. MIR503HG is a lncRNA as well as a host gene for the miRNAs miR-503 and miR-424. MIR503HG functions independently or synergistically with miR-503. MIR503HG affects cell proliferation, invasion, metastasis, apoptosis, angiogenesis, and other biological behaviors. The mechanism of MIR503HG in disease includes interaction with protein, sponging miRNA to regulate downstream target gene, and participation in NF-κB, TGF-β, ERK/MAPK, and PI3K/AKT signaling pathways. In this review, we summarize the molecular mechanisms of MIR503HG in disease and its potential applications in diagnosis, prognosis, and treatment. We also raise some unanswered questions in this area, providing insights for future research.
Collapse
Affiliation(s)
- Xue Han
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning Province, China.
| | - Bo Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning Province, China. libo--
| | - Shitai Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning Province, China.
| |
Collapse
|
7
|
Magnetic Nanomaterials Mediate Electromagnetic Stimulations of Nerves for Applications in Stem Cell and Cancer Treatments. J Funct Biomater 2023; 14:jfb14020058. [PMID: 36826857 PMCID: PMC9960824 DOI: 10.3390/jfb14020058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Although some progress has been made in the treatment of cancer, challenges remain. In recent years, advancements in nanotechnology and stem cell therapy have provided new approaches for use in regenerative medicine and cancer treatment. Among them, magnetic nanomaterials have attracted widespread attention in the field of regenerative medicine and cancer; this is because they have high levels of safety and low levels of invasibility, promote stem cell differentiation, and affect biological nerve signals. In contrast to pure magnetic stimulation, magnetic nanomaterials can act as amplifiers of an applied electromagnetic field in vivo, and by generating different effects (thermal, electrical, magnetic, mechanical, etc.), the corresponding ion channels are activated, thus enabling the modulation of neuronal activity with higher levels of precision and local modulation. In this review, first, we focused on the relationship between biological nerve signals and stem cell differentiation, and tumor development. In addition, the effects of magnetic nanomaterials on biological neural signals and the tumor environment were discussed. Finally, we introduced the application of magnetic-nanomaterial-mediated electromagnetic stimulation in regenerative medicine and its potential in the field of cancer therapy.
Collapse
|
8
|
Liu P, Li Y, Wang W, Bai Y, Jia H, Yuan Z, Yang Z. Role and mechanisms of the NF-ĸB signaling pathway in various developmental processes. Biomed Pharmacother 2022; 153:113513. [DOI: 10.1016/j.biopha.2022.113513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 11/02/2022] Open
|
9
|
Identification of circRNA Expression Profiles in BMSCs from Glucocorticoid-Induced Osteoporosis Model. Stem Cells Int 2022; 2022:3249737. [PMID: 35154330 PMCID: PMC8837445 DOI: 10.1155/2022/3249737] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 01/09/2022] [Indexed: 02/07/2023] Open
Abstract
Background. Circular RNAs (circRNAs) contribute to the regulation of many diseases. However, little is known about the role of circRNAs in the development of glucocorticoid-induced osteoporosis (GIOP). The present study is aimed at systematically characterizing the circRNA expression profiles in GIOP and predict the potential functions of the associated regulatory networks. Methods. A small animal GIOP model was developed in Sprague-Dawley rats given daily intraperitoneal doses of the synthetic glucocorticoid dexamethasone. Micro-CT and bone histomorphometry were performed to characterize the bone loss. Alizarin red S (ARS) staining and alkaline phosphatase (ALP) activity were assessed to determine the osteogenic differentiation potential of BMSCs. RNA sequencing was performed to identify differentially expressed circRNAs in BMSCs between the GIOP and normal groups, which were validated by qRT-PCR. siRNA interference experiments were used to demonstrate their function. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to predict the functions of differentially expressed circRNAs. The microRNA (miRNA) targets of the circRNAs and circRNA-miRNA interactions were predicted. Results. Micro-CT and bone histomorphometry confirmed the rat GIOP model. Both ARS intensity and ALP activity were decreased in GIOP BMSCs. Seventeen circRNAs were identified by
,
, and
, of which 7 were upregulated and 10 were downregulated. The qRT-PCR results of the selected circRNAs were consistent with the RNA-seq results and showed that circARSB and circAKT3 were significantly upregulated, while circPTEN and circTRPM7 were downregulated in the GIOP group. Further functional experiments found that downregulation of circARSB and circPTEN expression resulted in a corresponding change in osteogenic differentiation, suggesting that circARSB negatively, while circPTEN positively, regulates BMSC osteogenic differentiation. Analysis of circRNA-targeted miRNAs predicted that miR-135a-5p was associated with circARSB and circAKT3, and miR-881-3p was associated with circPTEN and circTRPM7. Furthermore, the signalling pathways associated with these differentially expressed circRNAs were predicted. Conclusions. The present study identified circARSB, circAKT3, circPTEN, and circTRPM7 as being associated with osteogenic differentiation during GIOP through a circRNA-targeted miRNA-mRNA axis, which might provide insight into the pathophysiological mechanism of GIOP.
Collapse
|
10
|
Bishop TF, Beck CW. Bacterial lipopolysaccharides can initiate regeneration of the Xenopus tadpole tail. iScience 2021; 24:103281. [PMID: 34765912 PMCID: PMC8571501 DOI: 10.1016/j.isci.2021.103281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 06/12/2021] [Accepted: 10/13/2021] [Indexed: 11/18/2022] Open
Abstract
Tadpoles of the frog Xenopus laevis can regenerate tails except for a short "refractory" period in which they heal rather than regenerate. Rapid and sustained production of ROS by NADPH oxidase (Nox) is critical for regeneration. Here, we show that tail amputation results in rapid, transient activation of the ROS-activated transcription factor NF-κB and expression of its direct target cox2 in the wound epithelium. Activation of NF-κB is also sufficient to rescue refractory tail regeneration. We propose that bacteria on the tadpole's skin could influence tail regenerative outcomes, possibly via LPS-TLR4-NF-κB signaling. When raised in antibiotics, fewer tadpoles in the refractory stage attempted regeneration, whereas addition of LPS rescued regeneration. Short-term activation of NF-κB using small molecules enhanced regeneration of tadpole hindlimbs, but not froglet forelimbs. We propose a model in which host microbiome contributes to creating optimal conditions for regeneration, via regulation of NF-κB by the innate immune system.
Collapse
Affiliation(s)
- Thomas F. Bishop
- Department of Zoology, University of Otago, 340 Great King Street, Dunedin, Otago 9016, New Zealand
| | - Caroline W. Beck
- Department of Zoology, University of Otago, 340 Great King Street, Dunedin, Otago 9016, New Zealand
| |
Collapse
|
11
|
Pluripotent-derived Mesenchymal Stem/stromal Cells: an Overview of the Derivation Protocol Efficacies and the Differences Among the Derived Cells. Stem Cell Rev Rep 2021; 18:94-125. [PMID: 34545529 DOI: 10.1007/s12015-021-10258-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) are remarkable tools for regenerative medicine. Therapeutic approaches using these cells can promote increased activity and viability in several cell types through diverse mechanisms such as paracrine and immunomodulatory activities, contributing substantially to tissue regeneration and functional recovery. However, biological samples of human MSCs, usually obtained from adult tissues, often exhibit variable behavior during in vitro culture, especially with respect to cell population heterogeneity, replicative senescence, and consequent loss of functionality. Accordingly, it is necessary to establish standard protocols to generate high-quality, stable cell cultures, for example, by using pluripotent stem cells (PSCs) in derivation protocols of MSC-like cells since PSCs maintain their characteristics consistently during culture. However, the available protocols seem to generate distinct populations of PSC-derivedMSCs (PSC-MSCs) with peculiar attributes, which do not always resemble bona fide primary MSCs. The present review addresses the developmental basis behind some of these derivation protocols, exposing the differences among them and discussing the functional properties of PSC-MSCs, shedding light on elements that may help determine standard characterizations and criteria to evaluate and define these cells.
Collapse
|
12
|
Shrestha M, Nguyen TT, Park J, Choi JU, Yook S, Jeong JH. Immunomodulation effect of mesenchymal stem cells in islet transplantation. Biomed Pharmacother 2021; 142:112042. [PMID: 34403963 DOI: 10.1016/j.biopha.2021.112042] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) therapy has brought a great enthusiasm to the treatment of various immune disorders, tissue regeneration and transplantation therapy. MSCs are being extensively investigated for their immunomodulatory actions. MSCs can deliver immunomodulatory signals to inhibit allogeneic T cell immune responses by downregulating pro-inflammatory cytokines and increasing regulatory cytokines and growth factors. Islet transplantation is a therapeutic alternative to the insulin therapy for the treatment of type 1 diabetes mellitus (T1DM). However, the acute loss of islets due to the lack of vasculature and hypoxic milieu in the immediate post-transplantation period may lead to treatment failure. Moreover, despite the use of potent immunosuppressive drugs, graft failure persists because of immunological rejection. Many in vitro and in vivo researches have demonstrated the multipotency of MSCs as a mediator of immunomodulation and a great approach for enhancement of islet engraftment. MSCs can interact with immune cells of the innate and adaptive immune systems via direct cell-cell contact or through secretomes containing numerous soluble growth and immunomodulatory factors or mitochondrial transfer. This review highlights the interactions between MSCs and different immune cells to mediate immunomodulatory functions along with the importance of MSCs therapy for the successful islet transplantation.
Collapse
Affiliation(s)
- Manju Shrestha
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Tiep Tien Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jooho Park
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Jeong Uk Choi
- College of Pharmacy, Chonnam University, Gwangju 61186, Republic of Korea
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea.
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
13
|
Li HD, Xu Y, Zhu X, Liu Q, Omenn GS, Wang J. ClusterMine: A knowledge-integrated clustering approach based on expression profiles of gene sets. J Bioinform Comput Biol 2021; 18:2040009. [PMID: 32698720 DOI: 10.1142/s0219720020400090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Clustering analysis of gene expression data is essential for understanding complex biological data, and is widely used in important biological applications such as the identification of cell subpopulations and disease subtypes. In commonly used methods such as hierarchical clustering (HC) and consensus clustering (CC), holistic expression profiles of all genes are often used to assess the similarity between samples for clustering. While these methods have been proven successful in identifying sample clusters in many areas, they do not provide information about which gene sets (functions) contribute most to the clustering, thus limiting the interpretability of the resulting cluster. We hypothesize that integrating prior knowledge of annotated gene sets would not only achieve satisfactory clustering performance but also, more importantly, enable potential biological interpretation of clusters. Here we report ClusterMine, an approach that identifies clusters by assessing functional similarity between samples through integrating known annotated gene sets in functional annotation databases such as Gene Ontology. In addition to the cluster membership of each sample as provided by conventional approaches, it also outputs gene sets that most likely contribute to the clustering, thus facilitating biological interpretation. We compare ClusterMine with conventional approaches on nine real-world experimental datasets that represent different application scenarios in biology. We find that ClusterMine achieves better performances and that the gene sets prioritized by our method are biologically meaningful. ClusterMine is implemented as an R package and is freely available at: www.genemine.org/clustermine.php.
Collapse
Affiliation(s)
- Hong-Dong Li
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 400083, P. R. China
| | - Yunpei Xu
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 400083, P. R. China
| | - Xiaoshu Zhu
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 400083, P. R. China.,School of Computer Science and Engineering, Yulin Normal University, Yulin, Guangxi, P. R. China
| | - Quan Liu
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 400083, P. R. China
| | - Gilbert S Omenn
- Departments of Computational Medicine and Bioinformatics, Internal Medicine, Human Genetics and School of Public Health, University of Michigan, Ann Arbor, MI 48109-2218, USA
| | - Jianxin Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 400083, P. R. China
| |
Collapse
|
14
|
Dumortier C, Danopoulos S, Velard F, Al Alam D. Bone Cells Differentiation: How CFTR Mutations May Rule the Game of Stem Cells Commitment? Front Cell Dev Biol 2021; 9:611921. [PMID: 34026749 PMCID: PMC8139249 DOI: 10.3389/fcell.2021.611921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 04/12/2021] [Indexed: 12/30/2022] Open
Abstract
Cystic fibrosis (CF)-related bone disease has emerged as a significant comorbidity of CF and is characterized by decreased bone formation and increased bone resorption. Both osteoblast and osteoclast differentiations are impacted by cystic fibrosis transmembrane conductance regulator (CFTR) mutations. The defect of CFTR chloride channel or the loss of CFTRs ability to interact with other proteins affect several signaling pathways involved in stem cell differentiation and the commitment of these cells toward bone lineages. Specifically, TGF-, nuclear factor-kappa B (NF-B), PI3K/AKT, and MAPK/ERK signaling are disturbed by CFTR mutations, thus perturbing stem cell differentiation. High inflammation in patients changes myeloid lineage secretion, affecting both myeloid and mesenchymal differentiation. In osteoblast, Wnt signaling is impacted, resulting in consequences for both bone formation and resorption. Finally, CFTR could also have a direct role in osteoclasts resorptive function. In this review, we summarize the existing literature on the role of CFTR mutations on the commitment of induced pluripotent stem cells to bone cells.
Collapse
Affiliation(s)
- Claire Dumortier
- Division of Neonatology, Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States.,Universit de Reims Champagne-Ardenne, BIOS EA 4691, Reims, France
| | - Soula Danopoulos
- Division of Neonatology, Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Frdric Velard
- Universit de Reims Champagne-Ardenne, BIOS EA 4691, Reims, France
| | - Denise Al Alam
- Division of Neonatology, Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
| |
Collapse
|
15
|
Ren Y, Zhang K, Wang J, Meng X, Du X, Shi Z, Xue Y, Hong W. HOTAIRM1 promotes osteogenic differentiation and alleviates osteoclast differentiation by inactivating the NF-κB pathway. Acta Biochim Biophys Sin (Shanghai) 2021; 53:201-211. [PMID: 33404645 DOI: 10.1093/abbs/gmaa164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis (OP), one of the most prevalent chronic progressive bone diseases, is caused by deficiency in bone formation by osteoblasts or excessive bone resorption by osteoclasts and subsequently increases the risk of bone fractures. Emerging evidence has indicated that long noncoding RNAs (lncRNAs) play key roles in many biological processes and various disorders. However, the role and mechanism of HOX antisense intergenic RNA myeloid 1 (HOTAIRM1), a myeloid-specific lncRNA, in osteoclast differentiation, osteogenic differentiation, and OP remain unclear. In this study, we found that HOTAIRM1 was upregulated during ossification of ligamentum flavum and osteogenic differentiation, while it was downregulated in osteoclast differentiation and in the bone and serum of human and mouse with OP. Further investigation revealed that silencing Hotairm1 decreased the expression of the osteogenic markers and attenuated osteogenesis. Moreover, forced Hotairm1 expression inhibited the expressions of the osteoclastogenesis markers and alleviated receptor activator of nuclear factor kappa B (NF-κB) ligand (RANKL)-induced osteoclast differentiation. Mechanically, Hotairm1 repressed the phosphorylation of p65 and inhibitor of κBα (IκBα) and attenuated RANKL-mediated enhancement of phos-p65 and IκBα, suggesting that Hotairm1 inhibits RANKL-induced osteoclastogenesis through the NF-κB pathway. In conclusion, our data identified a crucial role of HOTAIRM1 in OP, providing a proof of this molecule as a potential diagnostic marker and a possible therapeutic target against OP.
Collapse
Affiliation(s)
- Yi Ren
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Kun Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jingzhao Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiaoxiang Meng
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiaoxiao Du
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zhemin Shi
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yuan Xue
- Department of Orthopedic Surgery, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Wei Hong
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease of Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
16
|
Mills WT, Nassar NN, Ravindra D, Li X, Meffert MK. Multi-Level Regulatory Interactions between NF-κB and the Pluripotency Factor Lin28. Cells 2020; 9:E2710. [PMID: 33348917 PMCID: PMC7767241 DOI: 10.3390/cells9122710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/22/2022] Open
Abstract
An appreciation for the complex interactions between the NF-κB transcription factor and the Lin28 RNA binding protein/let-7 microRNA pathways has grown substantially over the past decade. Both the NF-κB and Lin28/let-7 pathways are master regulators impacting cell survival, growth and proliferation, and an understanding of how interfaces between these pathways participate in governing pluripotency, progenitor differentiation, and neuroplastic responses remains an emerging area of research. In this review, we provide a concise summary of the respective pathways and focus on the function of signaling interactions at both the transcriptional and post-transcriptional levels. Regulatory loops capable of providing both reinforcing and extinguishing feedback have been described. We highlight convergent findings in disparate biological systems and indicate future directions for investigation.
Collapse
Affiliation(s)
- William T. Mills
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (W.T.M.IV); (N.N.N.); (D.R.); (X.L.)
| | - Noor N. Nassar
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (W.T.M.IV); (N.N.N.); (D.R.); (X.L.)
| | - Deepa Ravindra
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (W.T.M.IV); (N.N.N.); (D.R.); (X.L.)
| | - Xinbei Li
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (W.T.M.IV); (N.N.N.); (D.R.); (X.L.)
| | - Mollie K. Meffert
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (W.T.M.IV); (N.N.N.); (D.R.); (X.L.)
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
17
|
Deng P, Yu Y, Hong C, Wang CY. Growth differentiation factor 6, a repressive target of EZH2, promotes the commitment of human embryonic stem cells to mesenchymal stem cells. Bone Res 2020; 8:39. [PMID: 33298857 PMCID: PMC7672114 DOI: 10.1038/s41413-020-00116-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/13/2020] [Accepted: 08/14/2020] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived from human embryonic stem cells (hESCs) have significant potential for cell-mediated bone regeneration. Our recent study revealed that inhibiting the epigenetic regulator EZH2 plays a key role in promoting the mesodermal differentiation of hESCs. In this study, an epigenome-wide analysis of hESCs and MSCs revealed that growth differentiation factor 6 (GDF6), which is involved in bone formation, was the most upregulated gene associated with MSCs compared to hESCs. Furthermore, we identified GDF6 as a repressive target of EZH2 and found that ectopic GDF6 selectively promoted hESC differentiation towards the mesodermal lineage and enriched the MSC population. Our results provide molecular insights governing the mesenchymal commitment of hESCs and identify an inducing factor that offers strong promise for the future of regenerative medicine.
Collapse
Affiliation(s)
- Pend Deng
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yongxin Yu
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Christine Hong
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Cun-Yu Wang
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, Broad Stem Cell Research Institute and Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
18
|
Wu Y, Tang Y, Zhang X, Chu Z, Liu Y, Tang C. MMP-1 promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via the JNK and ERK pathway. Int J Biochem Cell Biol 2020; 129:105880. [PMID: 33157237 DOI: 10.1016/j.biocel.2020.105880] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/05/2020] [Accepted: 10/12/2020] [Indexed: 01/31/2023]
Abstract
Enhancing the functions of mesenchymal stem cells (MSCs) is considered a potential approach for promoting tissue regeneration. In this study, we investigated the effects of Matrix Metalloproteinase-1 (MMP-1) on bone marrow mesenchymal stem cells (BMSCs) and its mechanism. Our results showed that knockdown of MMP-1 impeded scratch closure, attenuated proliferation, inhibited ALP activity, ALP denser staining and mineralization in vitro, and decreased expression of RUNX2, OSX, OPN and OCN in BMSCs, while 20 ng/mL recombinant human MMP-1 protein (rhMMP-1) significantly accelerated scratch closure, enhanced proliferation, ALP activity, ALP denser staining and mineralization in vitro, and increased expression of RUNX2, OSX, OPN and OCN. In addition, knockdown of MMP-1 inhibited the expression of phosphorylated c-Jun N-terminal kinase (p-JNK) and phosphorylated extracellular regulated protein kinases (p-ERK), while 20 ng/mL rhMMP-1 increased the expression of p-JNK and p-ERK in BMSCs. Furthermore, inhibition of c-Jun N-terminal kinase (JNK) and extracellular regulated protein kinases (ERK) by their inhibitor SP600125 and PD98059 dramatically blocked MMP-1-enhanced ALP activity and mineralization in BMSCs. Our results revealed that MMP-1 could accelerate the osteogenic differentiation potentials of BMSCs via the JNK and ERK pathway, providing the mechanism underlying MSC biology and identifying a potential target for improving bone tissue regeneration.
Collapse
Affiliation(s)
- Yizhen Wu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China
| | - Yi Tang
- Second Dental Center, School and Hospital of Stomatology, Peking University, Beijing, 100081, PR China
| | - Xiaozhen Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China; Department of Dental Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Zhuangzhuang Chu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China
| | - Yajing Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China
| | - Chunbo Tang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China; Department of Dental Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| |
Collapse
|
19
|
Antagonism of Protease-Activated Receptor 4 Protects Against Traumatic Brain Injury by Suppressing Neuroinflammation via Inhibition of Tab2/NF-κB Signaling. Neurosci Bull 2020; 37:242-254. [PMID: 33111257 DOI: 10.1007/s12264-020-00601-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 07/19/2020] [Indexed: 12/23/2022] Open
Abstract
Traumatic brain injury (TBI) triggers the activation of the endogenous coagulation mechanism, and a large amount of thrombin is released to curb uncontrollable bleeding through thrombin receptors, also known as protease-activated receptors (PARs). However, thrombin is one of the most critical factors in secondary brain injury. Thus, the PARs may be effective targets against hemorrhagic brain injury. Since the PAR1 antagonist has an increased bleeding risk in clinical practice, PAR4 blockade has been suggested as a more promising treatment. Here, we explored the expression pattern of PAR4 in the brain of mice after TBI, and explored the effect and possible mechanism of BMS-986120 (BMS), a novel selective and reversible PAR4 antagonist on secondary brain injury. Treatment with BMS protected against TBI in mice. mRNA-seq analysis, Western blot, and qRT-PCR verification in vitro showed that BMS significantly inhibited thrombin-induced inflammation in astrocytes, and suggested that the Tab2/ERK/NF-κB signaling pathway plays a key role in this process. Our findings provide reliable evidence that blocking PAR4 is a safe and effective intervention for TBI, and suggest that BMS has a potential clinical application in the management of TBI.
Collapse
|
20
|
Ruiz-Perera LM, Greiner JFW, Kaltschmidt C, Kaltschmidt B. A Matter of Choice: Inhibition of c-Rel Shifts Neuronal to Oligodendroglial Fate in Human Stem Cells. Cells 2020; 9:cells9041037. [PMID: 32331232 PMCID: PMC7226153 DOI: 10.3390/cells9041037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/18/2022] Open
Abstract
The molecular mechanisms underlying fate decisions of human neural stem cells (hNSCs) between neurogenesis and gliogenesis are critical during neuronal development and neurodegenerative diseases. Despite its crucial role in the murine nervous system, the potential role of the transcription factor NF-κB in the neuronal development of hNSCs is poorly understood. Here, we analyzed NF-κB subunit distribution during glutamatergic differentiation of hNSCs originating from neural crest-derived stem cells. We observed several peaks of specific NF-κB subunits. The most prominent nuclear peak was shown by c-REL subunit during a period of 2–5 days after differentiation onset. Furthermore, c-REL inhibition with pentoxifylline (PTXF) resulted in a complete shift towards oligodendroglial fate, as demonstrated by the presence of OLIG2+/O4+-oligodendrocytes, which showed PDGFRα, NG2 and MBP at the transcript level. In addition c-REL impairment further produced a significant decrease in neuronal survival. Transplantation of PTXF-treated predifferentiated hNSCs into an ex vivo oxidative-stress-mediated demyelination model of mouse organotypic cerebellar slices further led to integration in the white matter and differentiation into MBP+ oligodendrocytes, validating their functionality and therapeutic potential. In summary, we present a human cellular model of neuronal differentiation exhibiting a novel essential function of NF-κB-c-REL in fate choice between neurogenesis and oligodendrogenesis which will potentially be relevant for multiple sclerosis and schizophrenia.
Collapse
Affiliation(s)
| | | | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (J.F.W.G.); (C.K.)
| | - Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany;
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (J.F.W.G.); (C.K.)
- Correspondence:
| |
Collapse
|
21
|
Zhao X, Xie L, Wang Z, Wang J, Xu H, Han X, Bai D, Deng P. ZBP1 (DAI/DLM-1) promotes osteogenic differentiation while inhibiting adipogenic differentiation in mesenchymal stem cells through a positive feedback loop of Wnt/β-catenin signaling. Bone Res 2020; 8:12. [PMID: 32195010 PMCID: PMC7058036 DOI: 10.1038/s41413-020-0085-4] [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] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/19/2019] [Accepted: 07/01/2019] [Indexed: 02/05/2023] Open
Abstract
The lineage specification of mesenchymal stem/stromal cells (MSCs) is tightly regulated by a wide range of factors. Recently, the versatile functions of ZBP1 (also known as DAI or DLM-1) have been reported in the blood circulation and immune systems. However, the biological function of ZBP1 during the lineage specification of MSCs is still unknown. In the present study, we found that ZBP1 was upregulated during osteogenesis but downregulated during adipogenesis in mouse bone marrow-derived MSCs (mBMSCs). ZBP1 was highly expressed in osteoblasts but expressed at a relatively low level in marrow adipocytes. Knockdown of ZBP1 inhibited alkaline phosphataseactivity, extracellular matrix mineralization, and osteogenesis-related gene expression in vitro and reduced ectopic bone formation in vivo. Knockdown of ZBP1 also promoted adipogenesis in MSCs in vitro. Conversely, the overexpression of ZBP1 increased the osteogenesis but suppressed the adipogenesis of MSCs. When the expression of ZBP1 was rescued, the osteogenic capacity of ZBP1-depleted mBMSCs was restored at both the molecular and phenotypic levels. Furthermore, we demonstrated that ZBP1, a newly identified target of Wnt/β-catenin signaling, was required for β-catenin translocation into nuclei. Collectively, our results indicate that ZBP1 is a novel regulator of bone and fat transdifferentiation via Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Xuefeng Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Zhiyong Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Jiongke Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Hao Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Peng Deng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| |
Collapse
|
22
|
Onodera Y, Teramura T, Takehara T, Fukuda K. Transforming Growth Factor β-Activated Kinase 1 Regulates Mesenchymal Stem Cell Proliferation Through Stabilization of Yap1/Taz Proteins. Stem Cells 2019; 37:1595-1605. [PMID: 31461199 PMCID: PMC6916189 DOI: 10.1002/stem.3083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/17/2019] [Accepted: 08/08/2019] [Indexed: 01/02/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BMMSCs) are multipotent stem cells capable of differentiation into a variety of cell types, proliferation, and production of clinically useful secretory factors. These advantages make BMMSCs highly useful for cell transplantation therapy. However, the molecular network underlying BMMSC proliferation remains poorly understood. Here, we showed that TGFβ-activated kinase 1 (Tak1) is a critical molecule that regulates the activation of cell cycling and that Tak1 inhibition leads to quiescence in BMMSCs both in vivo and in vitro. Mechanistically, Tak1 was phosphorylated by growth factor stimulations, allowing it to bind and stabilize Yap1/Taz, which could then be localized to the nucleus. We also demonstrated that the quiescence induction by inhibiting Tak1 increased oxidized stress tolerance and improved BMMSC engraftment in intramuscular and intrabone marrow cell transplantation models. This study reveals a novel pathway controlling BMMSC proliferation and suggests a useful method to improve the therapeutic effect of BMMSC transplantation. Stem Cells 2019;37:1595-1605.
Collapse
Affiliation(s)
- Yuta Onodera
- Division of Cell Biology for Regenerative MedicineInstitute of Advanced Clinical Medicine, Kindai University Faculty of MedicineOsakaJapan
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative MedicineInstitute of Advanced Clinical Medicine, Kindai University Faculty of MedicineOsakaJapan
| | - Toshiyuki Takehara
- Division of Cell Biology for Regenerative MedicineInstitute of Advanced Clinical Medicine, Kindai University Faculty of MedicineOsakaJapan
| | - Kanji Fukuda
- Division of Cell Biology for Regenerative MedicineInstitute of Advanced Clinical Medicine, Kindai University Faculty of MedicineOsakaJapan
| |
Collapse
|
23
|
Joel MDM, Yuan J, Wang J, Yan Y, Qian H, Zhang X, Xu W, Mao F. MSC: immunoregulatory effects, roles on neutrophils and evolving clinical potentials. Am J Transl Res 2019; 11:3890-3904. [PMID: 31312397 PMCID: PMC6614638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/07/2019] [Indexed: 06/10/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent, non-hematopoietic stem cells capable of differentiating into varieties of mature cell types such as osteoblasts, chondrocytes, adipocytes, and myoblasts. MSCs can be isolated from different kinds of tissues and cultivated in vitro for amplification and passage easily. These cells have drawn researcher's attention lately due to their ability of tissue repair, properties of hematopoiesis support and function of immunoregulation through the secretion of a variety of cytokines and growth factors that have both paracrine and autocrine activities. MSCs can regulate the proliferation of T cells, the antibodies secretion of B cells, maturation of DC, polarization of macrophages and also have many effects on neutrophils such as the suppression of NO secretion, inhibition of apoptosis, reduction of their infiltration, decreasing of N-Formy l-L-Methionine-L-leucy l-L-phenylalanine, induction of respiratory bursts and promotion of survivals. In some conditions, MSCs exert their function of treatment through immunoregulation. We reviewed the multifaceted roles of MSCs in communicating with immune cells mainly neutrophils in both in vivo and in vitro experiments. MSCs may provide promising trends for cell therapy in future.
Collapse
Affiliation(s)
- Mbobda Defo Marius Joel
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| | - Jintao Yuan
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong UniversityZhenjiang 212300, Jiangsu, P. R. China
| | - Jingyan Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| | - Yongmin Yan
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| | - Hui Qian
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| | - Wenrong Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| |
Collapse
|
24
|
Zhu Y, Long HT, Zeng L, Tang YF, Zhao RB, Lin ZY, Zhao SS, Cheng L. MiR-19b-3p regulates osteogenic differentiation of PDGFRα + muscle cells by specifically targeting PTEN. Cell Biol Int 2019; 43:565-573. [PMID: 30958604 DOI: 10.1002/cbin.11133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/16/2019] [Indexed: 01/08/2023]
Abstract
Heterotopic ossification (HO) is a common disturbing complication of intra-articular fractures. Its prevention and treatment are still difficult as its pathogenesis is unclear. It was reported that PDGFRα+ muscle cells in skeletal muscle may participate in the formation of HO; however, the specific mechanism is still unknown. This study investigated the function of miR-19b-3p in osteogenic differentiation of PDGFRα+ muscle cells. MiR-19b-3p was upregulated during PDGFRα+ muscle cell osteogenic differentiation. The exogenous expression of miR-19b-3p led to an increase in osteogenic marker gene transcription and translation during the osteogenic differentiation of PDGFRα+ muscle cells. Furthermore, both alkaline phosphatase and alizarin red staining increased in miR-19b-3p mimic transfected cells. Over-expression of miR-19b-3p led to the down-regulation of gene of phosphate and tension homology deleted on chromosome ten (PTEN). Additionally, the dual luciferase reporter assay demonstrated that PTEN was a direct target of miR-19b-3p. The increase of osteocalcin, osteopontin, and Runt-related transcription factor 2 protein levels induced by ectopic miR-19b-3p expression could be partially reversed by PTEN over-expression. In conclusion, our results suggested that miR-19b-3p may be a promising target in inhibiting PDGFRα+ muscle cell osteogenic differentiation and treatment of HO.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Orthopaedics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, People's Republic of China
| | - Hai-Tao Long
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, People's Republic of China
| | - Lei Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, People's Republic of China
| | - Yi-Fu Tang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, People's Republic of China
| | - Rui-Bo Zhao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, People's Republic of China
| | - Zhang-Yuan Lin
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, People's Republic of China
| | - Shu-Shan Zhao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, People's Republic of China
| | - Liang Cheng
- Department of Orthopaedics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, People's Republic of China
| |
Collapse
|
25
|
Abdal Dayem A, Lee SB, Kim K, Lim KM, Jeon TI, Seok J, Cho ASG. Production of Mesenchymal Stem Cells Through Stem Cell Reprogramming. Int J Mol Sci 2019; 20:ijms20081922. [PMID: 31003536 PMCID: PMC6514654 DOI: 10.3390/ijms20081922] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess a broad spectrum of therapeutic applications and have been used in clinical trials. MSCs are mainly retrieved from adult or fetal tissues. However, there are many obstacles with the use of tissue-derived MSCs, such as shortages of tissue sources, difficult and invasive retrieval methods, cell population heterogeneity, low purity, cell senescence, and loss of pluripotency and proliferative capacities over continuous passages. Therefore, other methods to obtain high-quality MSCs need to be developed to overcome the limitations of tissue-derived MSCs. Pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are considered potent sources for the derivation of MSCs. PSC-derived MSCs (PSC-MSCs) may surpass tissue-derived MSCs in proliferation capacity, immunomodulatory activity, and in vivo therapeutic applications. In this review, we will discuss basic as well as recent protocols for the production of PSC-MSCs and their in vitro and in vivo therapeutic efficacies. A better understanding of the current advances in the production of PSC-MSCs will inspire scientists to devise more efficient differentiation methods that will be a breakthrough in the clinical application of PSC-MSCs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology, Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| | | | | | | | | | | | | |
Collapse
|
26
|
Xu M, Shaw G, Murphy M, Barry F. Induced Pluripotent Stem Cell-Derived Mesenchymal Stromal Cells Are Functionally and Genetically Different From Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells 2019; 37:754-765. [PMID: 30779868 PMCID: PMC6591688 DOI: 10.1002/stem.2993] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/21/2018] [Accepted: 02/03/2019] [Indexed: 12/14/2022]
Abstract
There has been considerable interest in the generation of functional mesenchymal stromal cell (MSC) preparations from induced pluripotent stem cells (iPSCs) and this is now regarded as a potential source of unlimited, standardized, high‐quality cells for therapeutic applications in regenerative medicine. Although iMSCs meet minimal criteria for defining MSCs in terms of marker expression, there are substantial differences in terms of trilineage potential, specifically a marked reduction in chondrogenic and adipogenic propensity in iMSCs compared with bone marrow‐derived (BM) MSCs. To reveal the cellular basis underlying these differences, we conducted phenotypic, functional, and genetic comparisons between iMSCs and BM‐MSCs. We found that iMSCs express very high levels of both KDR and MSX2 compared with BM‐MSCs. In addition, BM‐MSCs had significantly higher levels of PDGFRα. These distinct gene expression profiles were maintained during culture expansion, suggesting that prepared iMSCs are more closely related to vascular progenitor cells (VPCs). Although VPCs can differentiate along the chondrogenic, osteogenic, and adipogenic pathways, they require different inductive conditions compared with BM‐MSCs. These observations suggest to us that iMSCs, based on current widely used preparation protocols, do not represent a true alternative to primary MSCs isolated from BM. Furthermore, this study highlights the fact that high levels of expression of typical MSC markers such as CD73, CD90, and CD105 are insufficient to distinguish MSCs from other mesodermal progenitors in differentiated induced pluripotent stem cell cultures. stem cells2019;37:754–765
Collapse
Affiliation(s)
- Maojia Xu
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Georgina Shaw
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Mary Murphy
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Frank Barry
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
27
|
Hou Q, Huang Y, Liu Y, Luo Y, Wang B, Deng R, Zhang S, Liu F, Chen D. Profiling the miRNA-mRNA-lncRNA interaction network in MSC osteoblast differentiation induced by (+)-cholesten-3-one. BMC Genomics 2018; 19:783. [PMID: 30373531 PMCID: PMC6206902 DOI: 10.1186/s12864-018-5155-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 10/10/2018] [Indexed: 02/06/2023] Open
Abstract
Background Our previous study showed that (+)-cholesten-3-one (CN) has the potential to induce the osteoblastic differentiation of mesenchymal stem cells (MSCs). However, the roles of CN in targeting miRNA-mRNA-lncRNA interactions to regulate osteoblast differentiation remain poorly understood. Results A total of 77 miRNAs (36 upregulated and 41 downregulated) and 295 lncRNAs (281 upregulated and 14 downregulated) were significantly differentially expressed during CN-induced MSC osteogenic differentiation. Bioinformatic analysis identified that several pathways may play vital roles in MSC osteogenic differentiation, such as the vitamin D receptor signalling, TNF signalling, PI3K-Akt signalling, calcium signalling, and mineral absorption pathways. Further bioinformatic analysis revealed 16 core genes, including 6 mRNAs (Vdr, Mgp, Fabp3, Fst, Cd38, and Col1a1), 5 miRNAs (miR-483, miR-298, miR-361, miR-92b and miR-155) and 5 lncRNAs (NR_046246.1, NR_046239.1, XR_086062.1, XR_145872.1 and XR_146737.1), that may play important roles in regulating the CN-induced osteogenic differentiation of MSCs. Verified by the luciferase reporter, AR-S, qRT-PCR and western blot assays, we identified one miRNA (miR-298) that may enhance the osteogenic differentiation potential of MSCs via the vitamin D receptor signalling pathway. Conclusions This study revealed the global expression profile of miRNAs and lncRNAs involved in the Chinese medicine active ingredient CN-induced osteoblast differentiation of MSCs for the first time and provided a foundation for future investigations of miRNA-mRNA-lncRNA interaction networks to completely illuminate the regulatory role of CN in MSC osteoblast differentiation. Electronic supplementary material The online version of this article (10.1186/s12864-018-5155-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiuke Hou
- Department of Anatomy, The Research Centre of Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China.,The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Yongquan Huang
- Department of Orthopaedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Yamei Liu
- Department of Diagnosis of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Yiwen Luo
- Department of Trauma, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Bin Wang
- Department of Trauma, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Rudong Deng
- Department of Anatomy, The Research Centre of Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Saixia Zhang
- Department of Anatomy, The Research Centre of Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Fengbin Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Dongfeng Chen
- Department of Anatomy, The Research Centre of Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, People's Republic of China.
| |
Collapse
|
28
|
MSX2 Initiates and Accelerates Mesenchymal Stem/Stromal Cell Specification of hPSCs by Regulating TWIST1 and PRAME. Stem Cell Reports 2018; 11:497-513. [PMID: 30033084 PMCID: PMC6092836 DOI: 10.1016/j.stemcr.2018.06.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023] Open
Abstract
The gap in knowledge of the molecular mechanisms underlying differentiation of human pluripotent stem cells (hPSCs) into the mesenchymal cell lineages hinders the application of hPSCs for cell-based therapy. In this study, we identified a critical role of muscle segment homeobox 2 (MSX2) in initiating and accelerating the molecular program that leads to mesenchymal stem/stromal cell (MSC) differentiation from hPSCs. Genetic deletion of MSX2 impairs hPSC differentiation into MSCs. When aided with a cocktail of soluble molecules, MSX2 ectopic expression induces hPSCs to form nearly homogeneous and fully functional MSCs. Mechanistically, MSX2 induces hPSCs to form neural crest cells, an intermediate cell stage preceding MSCs, and further differentiation by regulating TWIST1 and PRAME. Furthermore, we found that MSX2 is also required for hPSC differentiation into MSCs through mesendoderm and trophoblast. Our findings provide novel mechanistic insights into lineage specification of hPSCs to MSCs and effective strategies for applications of stem cells for regenerative medicine.
Collapse
|
29
|
Abstract
Stem cell aging is a process in which stem cells progressively lose their ability to self-renew or differentiate, succumb to senescence or apoptosis, and eventually become functionally depleted. Unresolved oxidative stress and concomitant oxidative damages of cellular macromolecules including nucleic acids, proteins, lipids, and carbohydrates have been recognized to contribute to stem cell aging. Excessive production of reactive oxygen species and insufficient cellular antioxidant reserves compromise cell repair and metabolic homeostasis, which serves as a mechanistic switch for a variety of aging-related pathways. Understanding the molecular trigger, regulation, and outcomes of those signaling networks is critical for developing novel therapies for aging-related diseases by targeting stem cell aging. Here we explore the key features of stem cell aging biology, with an emphasis on the roles of oxidative stress in the aging process at the molecular level. As a concept of cytoprotection of stem cells in transplantation, we also discuss how systematic enhancement of endogenous antioxidant capacity before or during graft into tissues can potentially raise the efficacy of clinical therapy. Finally, future directions for elucidating the control of oxidative stress and developing preventive/curative strategies against stem cell aging are discussed.
Collapse
Affiliation(s)
- Feng Chen
- 1 State Key Discipline of Infectious Diseases and Chemical Biology Laboratory for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Yingxia Liu
- 1 State Key Discipline of Infectious Diseases and Chemical Biology Laboratory for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Nai-Kei Wong
- 1 State Key Discipline of Infectious Diseases and Chemical Biology Laboratory for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Jia Xiao
- 1 State Key Discipline of Infectious Diseases and Chemical Biology Laboratory for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China.,2 Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Kwok-Fai So
- 3 GMH Institute of CNS Regeneration, Guangdong Medical Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, China
| |
Collapse
|
30
|
Huang H, Dou L, Song J, Luo J. CBFA2T2 is required for BMP-2-induced osteogenic differentiation of mesenchymal stem cells. Biochem Biophys Res Commun 2018; 496:1095-1101. [DOI: 10.1016/j.bbrc.2018.01.144] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/23/2018] [Indexed: 01/06/2023]
|
31
|
Roux C, Saviane G, Pini J, Belaïd N, Dhib G, Voha C, Ibáñez L, Boutin A, Mazure NM, Wakkach A, Blin-Wakkach C, Rouleau M. Immunosuppressive Mesenchymal Stromal Cells Derived from Human-Induced Pluripotent Stem Cells Induce Human Regulatory T Cells In Vitro and In Vivo. Front Immunol 2018; 8:1991. [PMID: 29422893 PMCID: PMC5788894 DOI: 10.3389/fimmu.2017.01991] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/21/2017] [Indexed: 12/18/2022] Open
Abstract
Despite mesenchymal stromal cells (MSCs) are considered as a promising source of cells to modulate immune functions on cells from innate and adaptive immune systems, their clinical use remains restricted (few number, limited in vitro expansion, absence of a full phenotypic characterization, few insights on their in vivo fate). Standardized MSCs derived in vitro from human-induced pluripotent stem (huIPS) cells, remediating part of these issues, are considered as well as a valuable tool for therapeutic approaches, but their functions remained to be fully characterized. We generated multipotent MSCs derived from huiPS cells (huiPS-MSCs), and focusing on their immunosuppressive activity, we showed that human T-cell activation in coculture with huiPS-MSCs was significantly reduced. We also observed the generation of functional CD4+ FoxP3+ regulatory T (Treg) cells. Further tested in vivo in a model of human T-cell expansion in immune-deficient NSG mice, huiPS-MSCs immunosuppressive activity prevented the circulation and the accumulation of activated human T cells. Intracytoplasmic labeling of cytokines produced by the recovered T cells showed reduced percentages of human-differentiated T cells producing Th1 inflammatory cytokines. By contrast, T cells producing IL-10 and FoxP3+-Treg cells, absent in non-treated animals, were detected in huiPS-MSCs treated mice. For the first time, these results highlight the immunosuppressive activity of the huiPS-MSCs on human T-cell stimulation with a concomitant generation of human Treg cells in vivo. They may favor the development of new tools and strategies based on the use of huiPS cells and their derivatives for the induction of immune tolerance.
Collapse
Affiliation(s)
- Clémence Roux
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France.,Service d'Hématologie Clinique, CHU de Nice, Hôpital de l'Archet, Nice, France
| | - Gaëlle Saviane
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Jonathan Pini
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Nourhène Belaïd
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Gihen Dhib
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Christine Voha
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France.,Pôle d'Odontologie, CHU de Nice, Hôpital Saint-Roch, Nice, France
| | - Lidia Ibáñez
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Antoine Boutin
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Nathalie M Mazure
- Université Nice-Sophia Antipolis, Nice, France.,Institute for Research on Cancer and Aging of Nice, CNRS-UMR 7284-INSERM U108, Centre Antoine Lacassagne, Nice, France
| | - Abdelilah Wakkach
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Matthieu Rouleau
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| |
Collapse
|
32
|
Wu W, Yin Y, Zhong J, Peng Y, Li S, Zheng L, Cao H, Zhang J. Cell therapy could be a potential way to improve lipoprotein lipase deficiency. Lipids Health Dis 2017; 16:189. [PMID: 28969646 PMCID: PMC5625700 DOI: 10.1186/s12944-017-0577-4] [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: 10/07/2016] [Accepted: 09/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lipoprotein lipase (LPL) deficiency is an autosomal recessive genetic disorder characterized by extreme hypertriglyceridemia, with no cure presently available. The purpose of this study was to test the possibility of using cell therapy to alleviate LPL deficiency. METHODS The LPL coding sequence was cloned into the MSCV retrovirus vector, after which MSCV-hLPL and MSCV (empty construct without LPL coding sequence) virion suspensions were made using the calcium chloride method. A muscle cell line (C2C12), kidney cell line (HEK293T) and pre-adipocyte cell line (3 T3-L1) were transfected with the virus in order to express recombinant LPL in vitro. Finally, each transfected cell line was injected subcutaneously into nude mice to identify the cell type which could secret recombinant LPL in vivo. Control cells were transfected with the MSCV empty vector. LPL activity was analyzed using a radioimmunoassay. RESULTS After virus infection, the LPL activity at the cell surface of each cell type was significantly higher than in the control cells, which indicates that all three cell types can be used to generate functional LPL. The transfected cells were injected subcutaneously into nude mice, and the LPL activity of the nearby muscle tissue at the injection site in mice injected with 3 T3-L1 cells was more than 5 times higher at the injection sites than at non-injected control sites. The other two types of cells did not show this trend. CONCLUSION The subcutaneous injection of adipocytes overexpressing LPL can improve the LPL activity of the adjacent tissue of nude mice. This is a ground-breaking preliminary study for the treatment of LPL deficiency, and lays a good foundation for using cell therapy to correct LPL deficiency.
Collapse
Affiliation(s)
- Wenjing Wu
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Yajun Yin
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Jie Zhong
- College of life science and biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, China
| | - Yongjia Peng
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Shuncai Li
- College of life science and biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, China
| | - Libin Zheng
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Hong Cao
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China
| | - Jin Zhang
- College of Biological and Chemical Science and Engineering, Jiaxing University, Lianglin Campus,118 Jiahang Road, Jiaxing, 314001, China. .,College of life science and biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, China.
| |
Collapse
|
33
|
Seo S, Jeon HY, Kim H. Comparison of Cellular Transforming Activity of OCT4, NANOG, and SOX2 in Immortalized Astrocytes. DNA Cell Biol 2017; 36:1000-1009. [PMID: 28933914 DOI: 10.1089/dna.2017.3889] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Embryonic stem cell factors-OCT4, NANOG, and SOX2-contribute to the maintenance of stem cell properties and malignant progression in various cancers, including glioblastoma. Although functional roles of each of these genes are well documented in stem cell and cancer biology, no study has directly compared their cellular transforming activity under same experimental conditions. In this study, we compared the cellular transforming activity of OCT4, NANOG, and SOX2 using human immortalized astrocytes cultured under serum-free stem cell culture conditions. We found that SOX2 exhibited the strongest transforming activities, such as cell proliferation, neurosphere formation, resistance to cytotoxic drug, and cell migration/invasion, which may be associated with the activation of the nuclear factor kappa B (NFκB) signaling pathway. Thus, OCT4, NANOG, and SOX2, known to be frequently activated in various cancers and cancer stem cells, may play a distinct role in the regulation of cellular transformation.
Collapse
Affiliation(s)
- Sunyoung Seo
- 1 Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University , Seoul, Republic of Korea
| | - Hee-Young Jeon
- 1 Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University , Seoul, Republic of Korea.,2 Institute of Animal Molecular Biotechnology, Korea University , Seoul, Republic of Korea
| | - Hyunggee Kim
- 1 Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University , Seoul, Republic of Korea.,2 Institute of Animal Molecular Biotechnology, Korea University , Seoul, Republic of Korea
| |
Collapse
|
34
|
Yu Y, Deng P, Yu B, Szymanski JM, Aghaloo T, Hong C, Wang CY. Inhibition of EZH2 Promotes Human Embryonic Stem Cell Differentiation into Mesoderm by Reducing H3K27me3. Stem Cell Reports 2017; 9:752-761. [PMID: 28826853 PMCID: PMC5599223 DOI: 10.1016/j.stemcr.2017.07.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 01/16/2023] Open
Abstract
Mesoderm derived from human embryonic stem cells (hESCs) is a major source of the mesenchymal stem/stromal cells (MSCs) that can differentiate into osteoblasts and chondrocytes for tissue regeneration. While significant progress has been made in understanding of molecular mechanisms of hESC differentiation into mesodermal cells, little is known about epigenetic factors controlling hESC fate toward mesoderm and MSCs. Identifying potential epigenetic factors that control hESC differentiation will undoubtedly lead to advancements in regenerative medicine. Here, we conducted an epigenome-wide analysis of hESCs and MSCs and uncovered that EZH2 was enriched in hESCs and was downregulated significantly in MSCs. The specific EZH2 inhibitor GSK126 directed hESC differentiation toward mesoderm and generated more MSCs by reducing H3K27me3. Our results provide insights into epigenetic landscapes of hESCs and MSCs and suggest that inhibiting EZH2 promotes mesodermal differentiation of hESCs. Inhibiting EZH2 directs hESC differentiation to mesoderm and generates more MSCs H3K27me3 levels decrease on specific gene clusters as hESCs differentiate to MSCs EZH2 is downregulated as hESC differentiate to MSCs
Collapse
Affiliation(s)
- Yongxin Yu
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peng Deng
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bo Yu
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - John M Szymanski
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tara Aghaloo
- Section of Oral and Maxillofacial Surgery, Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christine Hong
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Cun-Yu Wang
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Broad Stem Cell Research Institute and Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
35
|
Takano A, Fukuda T, Shinjo T, Iwashita M, Matsuzaki E, Yamamichi K, Takeshita M, Sanui T, Nishimura F. Angiopoietin-like protein 2 is a positive regulator of osteoblast differentiation. Metabolism 2017; 69:157-170. [PMID: 28285646 DOI: 10.1016/j.metabol.2017.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 12/13/2016] [Accepted: 01/08/2017] [Indexed: 11/28/2022]
Abstract
INTRODUCTION AND AIMS Several studies have reported that angiopoietin-like protein 2 (Angptl2) is expressed abundantly in adipocytes and is associated with adipose tissue inflammation. In the present study, we found that osteoblasts and mesenchymal stem cells also expressed Angptl2 at high levels. The aim of this study was to understand the role of Angptl2 in osteoblastic cell differentiation. METHODS Angptl2 expression was examined during osteoblast and adipocyte differentiation. The role of Angptl2 on cell differentiation and associated signaling was analyzed by gene knockdown using Angptl2 small interfering ribonucleic acid (siRNA). RESULTS Angptl2 was highly expressed in MC3T3-E1 cells, ST2 cells and primary osteoblasts, but not in RAW264 cells. Inhibition of Angptl2 expression using siRNA markedly inhibited alkaline phosphatase (ALP) expression and osteoblastic differentiation in MC3T3-E1, ST2 cells and primary osteoblasts. Angptl2 siRNA also inhibited adipocyte differentiation in ST2 cells. Treatment of MC3T3-E1 cells with Angptl2 siRNA led to the down-regulation of the activities of several cell signaling pathways, including extracellular signal-regulated kinase (ERK), Jun amino-terminal kinase (JNK), Akt, and nuclear factor kappa B (NF-κB) signals. It also down-regulated the expression of Osterix, but not that of runt-related transcription factor 2 (Runx2), suggesting that Angptl2 is a positive activator of Osterix and its down-stream signals. Treatment of MC3T3-E1 cells with anti-Angptl2 antibodies suppressed ALP gene expression. In addition, treatment of Angptl2 siRNA-treated cells with culture supernatants of normal MC3T3-E1 cells restored ALP gene expression, indicating that Angptl2 acts in an autocrine manner. CONCLUSIONS The results suggest that Angptl2 is an autocrine positive regulator of cell differentiation. Thus, it is suggested that Angptl2 regulates not only adipose tissue metabolism but also bone metabolism.
Collapse
Affiliation(s)
- Aiko Takano
- Department of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takao Fukuda
- Department of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takanori Shinjo
- Department of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Misaki Iwashita
- Department of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Etsuko Matsuzaki
- Department of Operative Dentistry and Endodontology, Fukuoka Dental College, Fukuoka, Japan
| | - Kensuke Yamamichi
- Department of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masaaki Takeshita
- Department of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Terukazu Sanui
- Department of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Fusanori Nishimura
- Department of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
36
|
Differential long noncoding RNA/mRNA expression profiling and functional network analysis during osteogenic differentiation of human bone marrow mesenchymal stem cells. Stem Cell Res Ther 2017; 8:30. [PMID: 28173844 PMCID: PMC5297123 DOI: 10.1186/s13287-017-0485-6] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/22/2016] [Accepted: 01/20/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are the most promising cell types for bone regeneration and repair due to their osteogenic potential. MSC differentiation is precisely regulated and orchestrated by the mechanical and molecular signals from the extracellular environment, involving complex pathways regulated at both the transcriptional and post-transcriptional levels. However, the potential role of long noncoding RNA (lncRNA) in the osteogenic differentiation of human MSCs remains largely unclear. METHODS Here, we undertook the survey of differential coding and noncoding transcript expression profiling and functional network analysis during osteogenic differentiation of human bone marrow mesenchymal stem cells (BMSCs) using human whole transcriptome microarray. The key pathways, mRNAs, and lncRNAs controlling osteogenic differentiation of BMSCs were identified by further bioinformatic analysis. The role of lncRNA in the osteogenic differentiation of MSCs was verified by lncRNA overexpression or knockdown methods. RESULTS A total of 1269 coding transcripts with 648 genes significantly upregulated and 621 genes downregulated, and 1408 lncRNAs with 785 lncRNAs significantly upregulated and 623 lncRNAs downregulated were detected along with osteogenic differentiation. Bioinformatic analysis identified that several pathways may be associated with osteogenic differentiation potentials of BMSCs, such as the MAPK signaling pathway, the Jak-STAT signaling pathway, the Toll-like receptor signaling pathway, and the TGF-beta signaling pathway, etc. Bioinformatic analysis also revealed 13 core regulatory genes including seven mRNAs (GPX3, TLR2, BDKRB1, FBXO5, BRCA1, MAP3K8, and SCARB1), and six lncRNAs (XR_111050, NR_024031, FR374455, FR401275, FR406817, and FR148647). Based on the analysis, we identified one lncRNA, XR_111050, that could enhance the osteogenic differentiation potentials of MSCs. CONCLUSIONS The potential regulatory mechanisms were identified using bioinformatic analyses. We further predicted the interactions of differentially expressed coding and noncoding genes, and identified core regulatory factors by co-expression networks during osteogenic differentiation of BMSCs. Our results could lead to a better understanding of the molecular mechanisms of genes and lncRNAs, and their cooperation underlying MSC osteogenic differentiation and bone formation. We identified that one lncRNA, XR_111050, could be a potential target for bone tissue engineering.
Collapse
|
37
|
Rinkenbaugh AL, Baldwin AS. The NF-κB Pathway and Cancer Stem Cells. Cells 2016; 5:cells5020016. [PMID: 27058560 PMCID: PMC4931665 DOI: 10.3390/cells5020016] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
The NF-κB transcription factor pathway is a crucial regulator of inflammation and immune responses. Additionally, aberrant NF-κB signaling has been identified in many types of cancer. Downstream of key oncogenic pathways, such as RAS, BCR-ABL, and Her2, NF-κB regulates transcription of target genes that promote cell survival and proliferation, inhibit apoptosis, and mediate invasion and metastasis. The cancer stem cell model posits that a subset of tumor cells (cancer stem cells) drive tumor initiation, exhibit resistance to treatment, and promote recurrence and metastasis. This review examines the evidence for a role for NF-κB signaling in cancer stem cell biology.
Collapse
Affiliation(s)
- Amanda L Rinkenbaugh
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|