101
|
von Siebenthal M, Besic M, Gheinani AH, Akshay A, Lizun-Platoni S, Kunz N, Burkhard FC, Monastyrskaya K. Urinary miRNA profiles discriminate between obstruction-induced bladder dysfunction and healthy controls. Sci Rep 2021; 11:10204. [PMID: 33986358 PMCID: PMC8119692 DOI: 10.1038/s41598-021-89535-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/28/2021] [Indexed: 02/07/2023] Open
Abstract
Urgency, frequency and incomplete emptying are the troublesome symptoms often shared between benign prostatic obstruction-induced (BLUTD) and neurogenic (NLUTD) lower urinary tract dysfunction. Previously, using bladder biopsies, we suggested a panel of miRNA biomarkers for different functional phenotypes of the bladder. Urine is a good source of circulating miRNAs, but sex- and age-matched controls are important for urinary metabolite comparison. In two groups of healthy subjects (average age 32 and 57 years old, respectively) the total protein and RNA content was very similar between age groups, but the number of secreted extracellular vesicles (uEVs) and expression of several miRNAs were higher in the young healthy male volunteers. Timing of urine collection was not important for these parameters. We also evaluated the suitability of urinary miRNAs for non-invasive diagnosis of bladder outlet obstruction (BOO). A three urinary miRNA signature (miR-10a-5p, miR-301b-3p and miR-363-3p) could discriminate between controls and patients with LUTD (BLUTD and NLUTD). This panel of representative miRNAs can be further explored to develop a non-invasive diagnostic test for BOO. The age-related discrepancy in the urinary miRNA content observed in this study points to the importance of selecting appropriate, age-matched controls.
Collapse
Affiliation(s)
- Michelle von Siebenthal
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, Bern, Switzerland
| | - Mustafa Besic
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, Bern, Switzerland
| | - Ali Hashemi Gheinani
- Urological Diseases Research Center, Boston Children's Hospital, Harvard Medical School, Boston, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Akshay Akshay
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, Bern, Switzerland
| | | | - Nadine Kunz
- Department of Urology, Inselspital University Hospital, 3010, Bern, Switzerland
| | - Fiona C Burkhard
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, Bern, Switzerland.,Department of Urology, Inselspital University Hospital, 3010, Bern, Switzerland
| | - Katia Monastyrskaya
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, Bern, Switzerland. .,Department of Urology, Inselspital University Hospital, 3010, Bern, Switzerland.
| |
Collapse
|
102
|
Dolati S, Shakouri SK, Dolatkhah N, Yousefi M, Jadidi-Niaragh F, Sanaie S. The role of exosomal non-coding RNAs in aging-related diseases. Biofactors 2021; 47:292-310. [PMID: 33621363 DOI: 10.1002/biof.1715] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/25/2021] [Indexed: 12/17/2022]
Abstract
Aging is a biological process caused by the accumulation of senescent cells with a permanent proliferative arrest. To the influence of aging on human life expectancy, there is essential for new biomarkers which possibly will assistance in recognizing age-associated pathologies. Exosomes, which are cell-secreted nanovesicles, make available a new biomarker detection and therapeutic approach for the transfer of different molecules with high capacity. Recently, non-coding RNAs (ncRNA) which are contained in exosomes have developed as important molecules regulating the complexity of aging and relevant human diseases. The discovery of ncRNA provided perceptions into an innovative regulatory platform that could interfere with cellular senescence. The non-coding transcriptome includes a different of RNA species, spanning from short ncRNAs (<200 nucleotides) to long ncRNAs, that are >200 bp long. Upgraded evidence displays that targeting ncRNAs possibly will influence senescence pathways. In this article, we will address ncRNAs that participated in age-related and cellular senescence diseases. Growing conception of ncRNAs in the aging process possibly will be responsible for new understandings into the improvement of age-related diseases and elongated life span.
Collapse
Affiliation(s)
- Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Kazem Shakouri
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Dolatkhah
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sarvin Sanaie
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
103
|
Yuan D, Luo J, Sun Y, Hao L, Zheng J, Yang Z. PCOS follicular fluid derived exosomal miR-424-5p induces granulosa cells senescence by targeting CDCA4 expression. Cell Signal 2021; 85:110030. [PMID: 33930499 DOI: 10.1016/j.cellsig.2021.110030] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 01/10/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous reproductive disease, characterized by increased ovarian androgen biosynthesis, chronic anovulation and polycystic ovaries. The objective of this study was to identify the altered miRNA expression profiles in follicular fluid derived exosomes isolated from PCOS patients and to investigate the molecular functions of exosomal miR-424-5p. Herein, small RNA sequencing showed that twenty-five miRNAs were differentially expressed between control and PCOS group. The alterations in the miRNA profile were related to the endocrine resistance, cell growth and proliferation, cellular senescence and insulin signaling pathway. Among these differentially expressed miRNAs, we found that the expression of miR-424-5p was significantly decreased in PCOS exosomes and primary granulosa cells (GCs). Exosome-enriched miR-424-5p significantly promoted GCs senescence and suppressed cell proliferation. Similar to the results obtained in the cells transfected with miR-424-5p mimic, miR-424-5p mimic significantly decreased cell proliferation ability and induced senescence, but treatment with miR-424-5p inhibitor got the opposite results. In addition, cell division cycle associated 4 (CDCA4) gene displayed an inverse expression pattern to those of miR-424-5p, was identified as the direct target of miR-424-5p. Overexpression of CDCA4 reversed the effects of exosomal miR-424-5p on GCs via activation of Rb/E2F1 signaling pathway. These results demonstrate that exosomal miR-424-5p inhibits GCs proliferation and induces cellular senescence in PCOS through blocking CDCA4-mediated Rb/E2F1 signaling. Our findings provide new information on abnormal follicular development in PCOS.
Collapse
Affiliation(s)
- Dong Yuan
- Department of Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Jing Luo
- Department of Pathology, Basic Medical College of Chongqing Medical University, 400016, PR China
| | - Yixuan Sun
- Department of Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Lijuan Hao
- Department of Reproductive Endocrinology, Chongqing Health Center for Women and Children, Chongqing 401147, PR China
| | - Jing Zheng
- Department of Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Zhu Yang
- Department of Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China.
| |
Collapse
|
104
|
Kynurenine induces an age-related phenotype in bone marrow stromal cells. Mech Ageing Dev 2021; 195:111464. [PMID: 33631183 DOI: 10.1016/j.mad.2021.111464] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/08/2021] [Accepted: 02/21/2021] [Indexed: 01/02/2023]
Abstract
Advanced age is one of the important contributing factors for musculoskeletal deterioration. Although the exact mechanism behind this degeneration is unknown, it has been previously established that nutritional signaling plays a vital role in musculoskeletal pathophysiology. Our group established the vital role of the essential amino acid, tryptophan, in aging musculoskeletal health. With advanced age, inflammatory factors activate indoleamine 2,3-dioxygenase (IDO1) and accumulate excessive intermediate tryptophan metabolites such as Kynurenine (KYN). With age, Kynurenine accumulates and suppresses osteogenic differentiation, impairs autophagy, promotes early senescence, and alters cellular bioenergetics of bone marrow stem cells. Recent studies have shown that Kynurenine negatively impacts bone marrow stromal cells (BMSCs) and, consequently, promotes bone loss. Overall, understanding the mechanism behind BMSCs losing their ability for osteogenic differentiation can provide insight into the prevention of osteoporosis and the development of targeted therapies. Therefore, in this article, we review Kynurenine and how it plays a vital role in BMSC dysfunction and bone loss with age.
Collapse
|
105
|
Cai J, Qi H, Yao K, Yao Y, Jing D, Liao W, Zhao Z. Non-Coding RNAs Steering the Senescence-Related Progress, Properties, and Application of Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:650431. [PMID: 33816501 PMCID: PMC8017203 DOI: 10.3389/fcell.2021.650431] [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: 01/19/2021] [Accepted: 02/12/2021] [Indexed: 02/05/2023] Open
Abstract
The thirst to postpone and even reverse aging progress has never been quenched after all these decades. Unequivocally, mesenchymal stem cells (MSCs), with extraordinary abilities such as self-renewal and multi-directional differentiation, deserve the limelight in this topic. Though having several affable clinical traits, MSCs going through senescence would, on one hand, contribute to age-related diseases and, on the other hand, lead to compromised or even counterproductive therapeutical outcomes. Notably, increasing evidence suggests that non-coding RNAs (ncRNAs) could invigorate various regulatory processes. With even a slight dip or an uptick of expression, ncRNAs would make a dent in or even overturn cellular fate. Thereby, a systematic illustration of ncRNAs identified so far to steer MSCs during senescence is axiomatically an urgent need. In this review, we introduce the general properties and mechanisms of senescence and its relationship with MSCs and illustrate the ncRNAs playing a role in the cellular senescence of MSCs. It is then followed by the elucidation of ncRNAs embodied in extracellular vesicles connecting senescent MSCs with other cells and diversified processes in and beyond the skeletal system. Last, we provide a glimpse into the clinical methodologies of ncRNA-based therapies in MSC-related fields. Hopefully, the intricate relationship between senescence and MSCs will be revealed one day and our work could be a crucial stepping-stone toward that future.
Collapse
Affiliation(s)
- Jingyi Cai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hexu Qi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ke Yao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dian Jing
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wen Liao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, Osaka Dental University, Hirakata, Japan
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
106
|
Zhang Y, Ravikumar M, Ling L, Nurcombe V, Cool SM. Age-Related Changes in the Inflammatory Status of Human Mesenchymal Stem Cells: Implications for Cell Therapy. Stem Cell Reports 2021; 16:694-707. [PMID: 33636113 PMCID: PMC8072029 DOI: 10.1016/j.stemcr.2021.01.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Human mesenchymal stem/stromal cell (hMSC)-based cell therapies are promising for treating a variety of diseases. The unique immunomodulatory properties of hMSCs have extended their therapeutic potential beyond tissue regeneration. However, extensive pre-clinical culture expansion inevitably drives cells toward replicative “aging” and a consequent decline in quality. These “in vitro-aged” hMSCs resemble biologically aged cells, which have been reported to show senescence signatures, diminished immunosuppressive capacity, and weakened regenerative potential as well as pro-inflammatory features. In this review, we have surveyed the literature to explore the intimate relationship between the inflammatory status of hMSCs and their in vitro aging process. We posit that a shift from an anti-inflammatory to a pro-inflammatory phenotype of culture-expanded hMSCs contributes to a deterioration in their therapeutic efficacy. Potential molecular and cellular mechanisms underpinning this phenomenon have been discussed. We have also highlighted studies that leverage these mechanisms to make culture-expanded hMSCs more amenable for clinical use. Aged MSCs have reduced immunosuppressive potential Chronic inflammatory microenvironments can exacerbate MSC senescence and aging The immunomodulatory potential of MSCs should be assessed prior to clinical use MSC immunomodulatory properties may be modified in vitro by bioengineering means
Collapse
Affiliation(s)
- Ying Zhang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Maanasa Ravikumar
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore
| | - Ling Ling
- Institute of Medical Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore 636921, Singapore
| | - Simon M Cool
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore.
| |
Collapse
|
107
|
Xu M, Su X, Xiao X, Yu H, Li X, Keating A, Wang S, Zhao RC. Hydrogen Peroxide-Induced Senescence Reduces the Wound Healing-Promoting Effects of Mesenchymal Stem Cell-Derived Exosomes Partially via miR-146a. Aging Dis 2021; 12:102-115. [PMID: 33532131 PMCID: PMC7801275 DOI: 10.14336/ad.2020.0624] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have beneficial effects on wound healing. MSCs function through direct cell-cell communication or indirectly through paracrine secretion of exosomes. Here, we found that MSC-derived exosomes had pro-wound healing effects via promotion of angiogenesis; however, this promoting effect was significantly reduced when senescence was induced in parental MSCs by hydrogen peroxide (H2O2). Further experiments showed that decreased miR-146a expression in exosomes derived from senescent MSCs (s-exo) contributed to these findings. In vitro, the pro-angiogenic effect of s-exo on tube formation in human umbilical vein endothelial cells was significantly reduced compared with that of exosomes derived from control MSCs (c-exo). In vivo, higher tube numbers and longer tube lengths were observed in the c-exo group compared with the s-exo group. Using microarray analysis, we found that miR-146a level in s-exo was lower than that in c-exo. Knockdown of miR-146a in c-exo decreased its capacity to promote angiogenesis, and overexpression of miR-146a in s-exo partially rescued its impaired pro-angiogenic capacity, thereby confirming that downregulation of miR-146a contributed to the reduced pro-wound healing capacity of s-exo. Our study is the first to demonstrate that cell senescence induced by H2O2 alters the pro-angiogenic ability of exosomes by modulating the expression of exosomal miRNAs, especially miR-146a, thus providing new insights into the correlation between parental cell state and exosome content and function.
Collapse
Affiliation(s)
- Meiqian Xu
- 1Institute of Basic Medical Sciences Chinese Academy of Medical Sciences,School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Xiaodong Su
- 2Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing 100070, China
| | - Xian Xiao
- 1Institute of Basic Medical Sciences Chinese Academy of Medical Sciences,School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Hongliang Yu
- 1Institute of Basic Medical Sciences Chinese Academy of Medical Sciences,School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Xiaoxia Li
- 3Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Armand Keating
- 4Cell Therapy Translational Research Laboratory, Princess Margaret, Cancer Centre, Toronto, Ontario M5G 2M9, Canada.,5Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5G2M9, Canada.,6Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | - Shihua Wang
- 1Institute of Basic Medical Sciences Chinese Academy of Medical Sciences,School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Robert Chunhua Zhao
- 1Institute of Basic Medical Sciences Chinese Academy of Medical Sciences,School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing 100005, China
| |
Collapse
|
108
|
Abstract
MicroRNAs (miRNAs) are a class of short RNA molecules that mediate the regulation of gene activity through interactions with target mRNAs and subsequent silencing of gene expression. It has become increasingly clear the miRNAs regulate many diverse aspects of bone biology, including bone formation and bone resorption processes. The role of miRNAs specifically in osteoclasts has been of recent investigation, due to clinical interest in discovering new paradigms to control excessive bone resorption, as is observed in multiple conditions including aging, estrogen deprivation, cancer metastases or glucocorticoid use. Therefore understanding the role that miRNAs play during osteoclastic differentiation is of critical importance. In this review, we highlight and discuss general aspects of miRNA function in osteoclasts, including exciting data demonstrating that miRNAs encapsulated in extracellular vesicles (EVs) either originating from osteoclasts, or signaling to osteoclast from divergent sites, have important roles in bone homeostasis.
Collapse
Affiliation(s)
- Megan M Weivoda
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Sun-Kyeong Lee
- Department of Medicine, UCONN Center on Aging, University Connecticut Health Center, Farmington, CT 06030, USA
| | - David G Monroe
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Robert and Arlene Kogod Center on Aging, Rochester, MN 55905, USA.
| |
Collapse
|
109
|
Hensley AP, McAlinden A. The role of microRNAs in bone development. Bone 2021; 143:115760. [PMID: 33220505 PMCID: PMC8019264 DOI: 10.1016/j.bone.2020.115760] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/06/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Epigenetic regulation is critical for proper bone development. Evidence from a large body of published literature informs us that microRNAs (miRNAs) are important epigenetic factors that control many aspects of bone development, homeostasis, and repair processes. These small non-coding RNAs function at the post-transcriptional level to suppress expression of specific target genes. Many target genes may be affected by one miRNA resulting in alteration in cellular pathways and networks. Therefore, changes in levels or activity of a specific miRNA (e.g. via genetic mutations, disease scenarios, or by over-expression or inhibition strategies in vitro or in vivo) can lead to substantial changes in cell processes including proliferation, metabolism, apoptosis and differentiation. In this review, Section 1 briefly covers general background information on processes that control bone development as well as the biogenesis and function of miRNAs. In Section 2, we discuss the importance of miRNAs in skeletal development based on findings from in vivo mouse models and human clinical reports. Section 3 focuses on describing more recent data from the last three years related to miRNA regulation of osteoblast differentiation in vitro. Some of these studies also involve utilization of an in vivo rodent model to study the effects of miRNA modulation in scenarios of osteoporosis, bone repair or ectopic bone formation. In Section 4, we provide some recent information from studies analyzing the potential of miRNA-mediated crosstalk in bone and how exosomes containing miRNAs from one bone cell may affect the differentiation or function of another bone cell type. We then conclude by summarizing where the field currently stands with respect to miRNA-mediated regulation of osteogenesis and how information gained from developmental processes can be instructive in identifying potential therapeutic miRNA targets for the treatment of certain bone conditions.
Collapse
Affiliation(s)
- Austin P Hensley
- Department of Biomedical Engineering, Washington University School of Medicine, St Louis, MO, United States of America
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, United States of America; Shriners Hospital for Children - St Louis, St Louis, MO, United States of America.
| |
Collapse
|
110
|
Exosome-Derived Noncoding RNAs as a Promising Treatment of Bone Regeneration. Stem Cells Int 2021; 2021:6696894. [PMID: 33542737 PMCID: PMC7843188 DOI: 10.1155/2021/6696894] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 02/05/2023] Open
Abstract
The reconstruction of large bone defects remains a crucial challenge in orthopedic surgery. The current treatments including autologous and allogenic bone grafting and bioactive materials have their respective drawbacks. While mesenchymal stem cell (MSC) therapy may address these limitations, growing researches have demonstrated that the effectiveness of MSC therapy depends on paracrine factors, particularly exosomes. This aroused great focus on the exosome-based cell-free therapy in the treatment of bone defects. Exosomes can transfer various cargoes, and noncoding RNAs are the most widely studied cargo through which exosomes exert their ability of osteoinduction. Here, we review the research status of the exosome-derived noncoding RNAs in bone regeneration, the potential application of exosomes, and the existing challenges.
Collapse
|
111
|
Herrmann M, Diederichs S, Melnik S, Riegger J, Trivanović D, Li S, Jenei-Lanzl Z, Brenner RE, Huber-Lang M, Zaucke F, Schildberg FA, Grässel S. Extracellular Vesicles in Musculoskeletal Pathologies and Regeneration. Front Bioeng Biotechnol 2021; 8:624096. [PMID: 33553127 PMCID: PMC7855463 DOI: 10.3389/fbioe.2020.624096] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
The incidence of musculoskeletal diseases is steadily increasing with aging of the population. In the past years, extracellular vesicles (EVs) have gained attention in musculoskeletal research. EVs have been associated with various musculoskeletal pathologies as well as suggested as treatment option. EVs play a pivotal role in communication between cells and their environment. Thereby, the EV cargo is highly dependent on their cellular origin. In this review, we summarize putative mechanisms by which EVs can contribute to musculoskeletal tissue homeostasis, regeneration and disease, in particular matrix remodeling and mineralization, pro-angiogenic effects and immunomodulatory activities. Mesenchymal stromal cells (MSCs) present the most frequently used cell source for EV generation for musculoskeletal applications, and herein we discuss how the MSC phenotype can influence the cargo and thus the regenerative potential of EVs. Induced pluripotent stem cell-derived mesenchymal progenitor cells (iMPs) may overcome current limitations of MSCs, and iMP-derived EVs are discussed as an alternative strategy. In the last part of the article, we focus on therapeutic applications of EVs and discuss both practical considerations for EV production and the current state of EV-based therapies.
Collapse
Affiliation(s)
- Marietta Herrmann
- Interdisciplinary Center for Clinical Research (IZKF) Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Solvig Diederichs
- Research Centre for Experimental Orthopaedics, Centre for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Svitlana Melnik
- Research Centre for Experimental Orthopaedics, Centre for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Drenka Trivanović
- Interdisciplinary Center for Clinical Research (IZKF) Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Shushan Li
- Department of Orthopedic Surgery, Experimental Orthopedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| | - Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - Rolf E. Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - Frank A. Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Susanne Grässel
- Department of Orthopedic Surgery, Experimental Orthopedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| |
Collapse
|
112
|
Contribution of extracellular vesicles in normal hematopoiesis and hematological malignancies. Heliyon 2021; 7:e06030. [PMID: 33521365 PMCID: PMC7820922 DOI: 10.1016/j.heliyon.2021.e06030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/05/2020] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed microparticles that have prominent roles in the intercellular crosstalk. EVs are secreted after fusion of endosomes with the plasma membrane (exosomes) or shed from the plasma membrane (microvesicles). These microparticles modulate bone marrow microenvironment and alter differentiation and expansion of normal hematopoietic cells. EVs originated from mesenchymal stromal cells have been shown to enhance expansion of myeloid-biased hematopoietic progenitor cells. In addition, megakaryocytic microparticles stimulate differentiation of hematopoietic stem and progenitor cells into mature megakaryocytes. The ability of EVs in induction of maturation and expansion of certain hematopoietic cells has implications in transfusion medicine and in targeted therapeutic modalities. Important prerequisites for these interventions are identification the specific targets of EVs, transferred biomolecules and molecular mechanisms underlying the fate decision in the target cells. EVs are also involved in the pathogenesis and progression of hematological malignancies including acute leukemia and multiples myeloma. In the current review, we provide a summary of studies which evaluated the significance of EVs in normal hematopoiesis and hematological malignancies.
Collapse
|
113
|
Wu H, Zhou X, Wang X, Cheng W, Hu X, Wang Y, Luo B, Huang W, Gu J. miR-34a in extracellular vesicles from bone marrow mesenchymal stem cells reduces rheumatoid arthritis inflammation via the cyclin I/ATM/ATR/p53 axis. J Cell Mol Med 2021; 25:1896-1910. [PMID: 33465281 PMCID: PMC7882978 DOI: 10.1111/jcmm.15857] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 07/31/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (Evs) participate in the development of rheumatoid arthritis (RA), but the mechanisms remain unclear. This study aimed to determine the mechanism by which microRNA‐34a (miR‐34a) contained in bone marrow mesenchymal stem cell (BM‐MSC)‐derived Evs functions in RA fibroblast‐like synoviocytes (RA‐FLSs). BM‐MSC‐derived Evs and an Evs inhibitor were extracted. A rat model of RA was established. miR‐34a gain‐ and loss‐of‐function experiments were performed, and the inflammation in rat synovial fluid and tissues was detected. The role of miR‐34a in RA‐FLSs was also measured in vitro. The target gene of miR‐34a was predicted using the online software TargetScan and identified using a dual‐luciferase reporter gene assay, and the activation of the ATM/ATR/p53 signalling pathway was assessed. BM‐MSC‐derived Evs mainly elevated miR‐34a expression, which reduced RA inflammation in vivo and inhibited RA‐FLS proliferation and resistance to apoptosis in vitro, while inhibited miR‐34a expression enhanced RA development. In addition, miR‐34a could target cyclin I to activate the ATM/ATR/p53 signalling pathway, thus inhibiting abnormal RA‐FLS growth and RA inflammation. Our study showed that miR‐34a contained in BM‐MSC‐derived Evs could reduce RA inflammation by inhibiting the cyclin I/ATM/ATR/p53 signalling pathway.
Collapse
Affiliation(s)
- Huaiguo Wu
- Center for Precision Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Xike Zhou
- Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China.,Department of Pathology, The Fifth People's Hospital of Wuxi, The Medical School of Jiangnan University, Wuxi, China
| | - Xuedong Wang
- Center for Precision Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, China.,Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China
| | - Wei Cheng
- Center for Precision Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Xinjia Hu
- Center for Precision Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, China.,Department of Osteoarthropathy, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University and the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Yueping Wang
- Center for Precision Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, China.,Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China.,Department of Biology, College of Arts & Science, Massachusetts University, Boston, MA, USA
| | - Bing Luo
- Center for Precision Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, China.,Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China
| | - Wenjun Huang
- Center for Precision Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, China.,Department of Osteoarthropathy, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University and the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Juan Gu
- Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China.,Department of Pathology, The Fifth People's Hospital of Wuxi, The Medical School of Jiangnan University, Wuxi, China
| |
Collapse
|
114
|
Gholami L, Nooshabadi VT, Shahabi S, Jazayeri M, Tarzemany R, Afsartala Z, Khorsandi K. Extracellular vesicles in bone and periodontal regeneration: current and potential therapeutic applications. Cell Biosci 2021; 11:16. [PMID: 33436061 PMCID: PMC7802187 DOI: 10.1186/s13578-020-00527-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/31/2020] [Indexed: 12/12/2022] Open
Abstract
Oral mesenchymal stem cells (MSCs) and their secretomes are considered important factors in the field of medical tissue engineering and cell free biotherapy due to their ease of access, differentiation potential, and successful therapeutic outcomes. Extracellular vesicles (EVs) and the conditioned medium (CM) from MSCs are gaining more attraction as an alternative to cell-based therapies due to the less ethical issues involved, and their easier acquisition, preservation, long term storage, sterilization, and packaging. Bone and periodontal regenerative ability of EVs and CM have been the focus of some recent studies. In this review, we looked through currently available literature regarding MSCs' EVs or conditioned medium and their general characteristics, function, and regenerative potentials. We will also review the novel applications in regenerating bone and periodontal defects.
Collapse
Affiliation(s)
- Leila Gholami
- Department of Periodontics, Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Science, Semnan, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Shiva Shahabi
- Student Research Committee, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marzieh Jazayeri
- Student Research Committee, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rana Tarzemany
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, Canada
| | - Zohreh Afsartala
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran.
| |
Collapse
|
115
|
Potter ML, Hill WD, Isales CM, Hamrick MW, Fulzele S. MicroRNAs are critical regulators of senescence and aging in mesenchymal stem cells. Bone 2021; 142:115679. [PMID: 33022453 PMCID: PMC7901145 DOI: 10.1016/j.bone.2020.115679] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/16/2020] [Accepted: 07/28/2020] [Indexed: 01/10/2023]
Abstract
MicroRNAs (miRNAs) have recently come under scrutiny for their role in various age-related diseases. Similarly, cellular senescence has been linked to disease and aging. MicroRNAs and senescence likely play an intertwined role in driving these pathologic states. In this review, we present the connection between these two drivers of age-related disease concerning mesenchymal stem cells (MSCs). First, we summarize key miRNAs that are differentially expressed in MSCs and other musculoskeletal lineage cells during senescence and aging. Additionally, we also reviewed miRNAs that are regulated via traditional senescence-associated secretory phenotype (SASP) cytokines in MSC. Lastly, we summarize miRNAs that have been found to target components of the cell cycle arrest pathways inherently activated in senescence. This review attempts to highlight potential miRNA targets for regenerative medicine applications in age-related musculoskeletal disease.
Collapse
Affiliation(s)
- Matthew L Potter
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - William D Hill
- Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC, 29403, United States of America
| | - Carlos M Isales
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Medicine, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America
| | - Mark W Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America; Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, United States of America
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Medicine, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America; Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, United States of America.
| |
Collapse
|
116
|
Li K, Cui M, Zhang K, Wang G, Zhai S. M1 macrophages-derived extracellular vesicles elevate microRNA-185-3p to aggravate the development of atherosclerosis in ApoE -/- mice by inhibiting small mothers against decapentaplegic 7. Int Immunopharmacol 2021; 90:107138. [PMID: 33302032 DOI: 10.1016/j.intimp.2020.107138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Extracellular vesicles (EVs) are vital mediators of transferring microRNAs (miRNAs). We focused on effect of miR-185-3p that mediated by macrophages-derived EVs on atherosclerosis (AS) by targeting small mothers against decapentaplegic 7 (Smad7). METHODS EVs were extracted from M1 macrophages and identified. ApoE-/- mice were treated with EVs, EVs containing miR-185-3p inhibitor or mimic, then the pathological changes of mouse aorta were observed. The levels of blood lipid, cell adhesion molecules, oxidative stress factors, inflammatory factors, and proliferation and apoptosis of vascular endothelial cells were assessed. Expression of miR-185-3p and Smad7 was detected and the targeting relationship between miR-185-3p and Smad7 was validated. RESULTS MiR-185-3p was upregulated while Smad7 was downregulated in atherosclerotic mouse aorta. M1 macrophages-derived EVs elevated miR-185-3p to promote development of AS pathology and levels of blood lipid, endothelial cellular adhesion, oxidative stress factors and inflammatory factors, suppressed cell proliferation and promoted cell apoptosis of vascular endothelial cells in atherosclerotic mice through downregulating Smad7. Smad7 was a target gene of miR-185-3p and miR-185-3p could inhibit expression of Smad7. CONCLUSION M1 macrophages-derived EVs and upregulated miR-185-3p aggravated the development of AS in ApoE-/- mice by negatively regulating Smad7. This research may further the understanding of AS mechanism.
Collapse
Affiliation(s)
- Kun Li
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Mingzhe Cui
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Kewei Zhang
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Guoquan Wang
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China
| | - Shuiting Zhai
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, China.
| |
Collapse
|
117
|
Aging of Bone Marrow Mesenchymal Stromal Cells: Hematopoiesis Disturbances and Potential Role in the Development of Hematologic Cancers. Cancers (Basel) 2020; 13:cancers13010068. [PMID: 33383723 PMCID: PMC7794884 DOI: 10.3390/cancers13010068] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/16/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary As for many other cancers, the risk of developing hematologic malignancies increases considerably as people age. In recent years, a growing number of studies have highlighted the influence of the aging microenvironment on hematopoiesis and tumor progression. Mesenchymal stromal cells are a major player in intercellular communication inside the bone marrow microenvironment involved in hematopoiesis support. With aging, their functions may be altered, leading to hematopoiesis disturbances which can lead to hematologic cancers. A good understanding of the mechanisms involved in mesenchymal stem cell aging and the consequences on hematopoiesis and tumor progression is therefore necessary for a better comprehension of hematologic malignancies and for the development of therapeutic approaches. Abstract Aging of bone marrow is a complex process that is involved in the development of many diseases, including hematologic cancers. The results obtained in this field of research, year after year, underline the important role of cross-talk between hematopoietic stem cells and their close environment. In bone marrow, mesenchymal stromal cells (MSCs) are a major player in cell-to-cell communication, presenting a wide range of functionalities, sometimes opposite, depending on the environmental conditions. Although these cells are actively studied for their therapeutic properties, their role in tumor progression remains unclear. One of the reasons for this is that the aging of MSCs has a direct impact on their behavior and on hematopoiesis. In addition, tumor progression is accompanied by dynamic remodeling of the bone marrow niche that may interfere with MSC functions. The present review presents the main features of MSC senescence in bone marrow and their implications in hematologic cancer progression.
Collapse
|
118
|
Liu YR, Cheng YQ, Wang SB, Su YR, Liu Y, Li CY, Jin L, Wan Q, Sang X, Wang ZC. Therapeutic effects and perspective of stem cell extracellular vesicles in aging and cancer. J Cell Physiol 2020; 236:4783-4796. [PMID: 33368322 DOI: 10.1002/jcp.30212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/02/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022]
Abstract
Senescent cells can secrete a plethora of cytokines which induce senescent phenotype of neighboring cells and was called senescence-associated secretory phenotype. Previously, it was believed that cancer was caused by the infinite division and uncontrolled proliferation of cells. Based on this, anticancer treatments were all aimed at killing cancer cells. Cancer is now considered an age-related disease. Cancer cells are not exogenous, but one of the worst results of injuries which initially induce cell senescence. Therefore, reversing cell senescence can fundamentally prevent and treat cancer. Though current anticancer treatments induce the cancer cells apoptosis, they induce senescence of normal cells at the same time, thus promoting the occurrence and development of cancer and forming a vicious circle. Extracellular vesicles (EVs) are nano-sized vesicles which partially mirror their parent cells. In the tumor microenvironment, EVs of senescent cells can change the expression profile of cancer cells, contributing to their resistance to chemotherapy. There is growing evidence indicates that stem cell EVs exert effective antiaging and anticancer actions by transferring functional microRNAs and proteins. This review will summarize the therapeutic role of stem cell EVs in reversing aging and cancer, which suggests the broad clinical application perspective.
Collapse
Affiliation(s)
- Yu-Run Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ya-Qi Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shou-Bi Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ya-Ru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chao-Yang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lin Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qi Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xuan Sang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Chong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
119
|
He C, He W, Hou J, Chen K, Huang M, Yang M, Luo X, Li C. Bone and Muscle Crosstalk in Aging. Front Cell Dev Biol 2020; 8:585644. [PMID: 33363144 PMCID: PMC7758235 DOI: 10.3389/fcell.2020.585644] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis and sarcopenia are two age-related diseases that affect the quality of life in the elderly. Initially, they were thought to be two independent diseases; however, recently, increasing basic and clinical data suggest that skeletal muscle and bone are both spatially and metabolically connected. The term "osteosarcopenia" is used to define a condition of synergy of low bone mineral density with muscle atrophy and hypofunction. Bone and muscle cells secrete several factors, such as cytokines, myokines, and osteokines, into the circulation to influence the biological and pathological activities in local and distant organs and cells. Recent studies reveal that extracellular vesicles containing microRNAs derived from senescent skeletal muscle and bone cells can also be transported and aid in regulating bone-muscle crosstalk. In this review, we summarize the age-related changes in the secretome and extracellular vesicle-microRNAs secreted by the muscle and bone, and discuss their interactions between muscle and bone cells during aging.
Collapse
Affiliation(s)
- Chen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Wenzhen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Jing Hou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Kaixuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| |
Collapse
|
120
|
Ahmadi M, Rezaie J. Ageing and mesenchymal stem cells derived exosomes: Molecular insight and challenges. Cell Biochem Funct 2020; 39:60-66. [PMID: 33164248 DOI: 10.1002/cbf.3602] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
Ageing induces a great risk factor that participates in progressing various degenerative diseases morbidities. The main characteristic of ageing is the failure in maintaining homeostasis in the organs with a cellular senescence. Senescence is characterized by reduced cell growth, evade cellular death, and acquiring a senescence-associated secretory phenotype (SASP). Mesenchymal stem cells (MSCs) are advantageous cells in regenerative medicine, exerting pleiotropic functions by producing soluble factors, such as exosomes. MSCs and their exosomes (MSCs-Exo) kinetic are affected by ageing and other aged exosomes. Exosomes biogenesis from aged MSCs is accelerated and their exosomal cargoes, such as miRNAs, vary as compared to those of normal cells. Besides, exosomes from aged MSCs loss their regenerative potential and may negatively influence the function of recipient cells. MSCs-Exo can improve ageing and age-related diseases; however, the detailed mechanisms remain yet elusive. Although exosomes-therapy may serve as a new approach to combat ageing, the translation of preclinical results to clinic needs more extensive investigation on exosomes both on their biology and related techniques. Overall, scrutiny on the effect of ageing on MSCs and vice versa is vital for designing novel therapy using MSCs with focus on the management of older individuals.
Collapse
Affiliation(s)
- Mahdi Ahmadi
- Tuberculosis and lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
121
|
Roy E, Byrareddy SN, Reid SP. Role of MicroRNAs in Bone Pathology during Chikungunya Virus Infection. Viruses 2020; 12:E1207. [PMID: 33114216 PMCID: PMC7690852 DOI: 10.3390/v12111207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/10/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023] Open
Abstract
Chikungunya virus (CHIKV) is an alphavirus, transmitted by mosquitoes, which causes Chikungunya fever with symptoms of fever, rash, headache, and joint pain. In about 30%-40% of cases, the infection leads to polyarthritis and polyarthralgia. Presently, there are no treatment strategies or vaccine for Chikungunya fever. Moreover, the mechanism of CHIKV induced bone pathology is not fully understood. The modulation of host machinery is known to be essential in establishing viral pathogenesis. MicroRNAs (miRNAs) are small non-coding RNAs that regulate major cellular functions by modulating gene expression. Fascinatingly, recent reports have indicated the role of miRNAs in regulating bone homeostasis and altered expression of miRNAs in bone-related pathological diseases. In this review, we summarize the altered expression of miRNAs during CHIKV pathogenesis and the possible role of miRNAs during bone homeostasis in the context of CHIKV infection. A holistic understanding of the different signaling pathways targeted by miRNAs during bone remodeling and during CHIKV-induced bone pathology may lead to identification of useful biomarkers or therapeutics.
Collapse
Affiliation(s)
- Enakshi Roy
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA;
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
| | - St Patrick Reid
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA;
| |
Collapse
|
122
|
Niazi V, Parseh B, Ahani M, Karami F, Gilanchi S, Atarodi K, Soufi M, Soleimani M, Ghafouri-Fard S, Taheri M, Zali H. Communication between stromal and hematopoietic stem cell by exosomes in normal and malignant bone marrow niche. Biomed Pharmacother 2020; 132:110854. [PMID: 33059261 DOI: 10.1016/j.biopha.2020.110854] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/26/2020] [Accepted: 10/04/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) have been regarded as important tools for cell-cell communication. They act as carriers for the transfer of various molecules such as genes, proteins and miRNA. EVs shift and transfer their ingredients to target cells in an active form. These particles have prominent roles in modulation of bone marrow (BM) niche; therefore they can regulate proliferation, differentiation, and other properties of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). This review discusses the different roles of EVs on BM niche; HPCs fate regulation and downstream effects of them on HSCs. Moreover, cellular and molecular mechanisms of BM microenvironment cross-talking are explained in healthy and malignant settings.
Collapse
Affiliation(s)
- Vahid Niazi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19857-17443, Iran
| | - Benyamin Parseh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 14177-55469, Iran
| | - Milad Ahani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19857-17443, Iran
| | - Farshid Karami
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19857-17443, Iran
| | - Samira Gilanchi
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, 19716-53313, Iran
| | - Kamran Atarodi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, 14665-1157, Iran
| | - Mina Soufi
- Department of Hematology and Cell Therapy, Faculty of Medical Science, Tarbiat Modares University, Tehran, 14117-13116, Iran
| | - Masoud Soleimani
- Department of Hematology and Cell Therapy, Faculty of Medical Science, Tarbiat Modares University, Tehran, 14117-13116, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19857-17443, Iran; Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, 19716-53313, Iran.
| |
Collapse
|
123
|
Liu Y, Shen Q, Zhang L, Xiang W. Extracellular Vesicles: Recent Developments in Aging and Reproductive Diseases. Front Cell Dev Biol 2020; 8:577084. [PMID: 33043012 PMCID: PMC7527740 DOI: 10.3389/fcell.2020.577084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs), present in cell culture media and several body fluids, play a prominent role in intercellular communication under physiological and pathological conditions. We performed a systematic literature search to review evidence regarding the existence, composition, and release of different EVs, as well as the biomarkers, cargos, and separation methods. We also reviewed the potential of EVs to transport cargos and alter the function and phenotype of recipient cells associated with aging and reproductive diseases, including polycystic ovary syndrome and endometriosis. In aging, EVs promote inflammatory reactions and offsetting the occurrence of aging. In the polycystic ovary syndrome and endometriosis, EVs and their cargos are involved in the occurrence of diseases, therapeutic strategies, and perform as non-invasive biomarkers. As the study of EVs is still in the early stages, it is not surprising that most of the current literature only describes their possible roles.
Collapse
Affiliation(s)
- Yu Liu
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuzi Shen
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Zhang
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenpei Xiang
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
124
|
Epigenetic Regulation in Mesenchymal Stem Cell Aging and Differentiation and Osteoporosis. Stem Cells Int 2020; 2020:8836258. [PMID: 32963550 PMCID: PMC7501554 DOI: 10.1155/2020/8836258] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a reliable source for cell-based regenerative medicine owing to their multipotency and biological functions. However, aging-induced systemic homeostasis disorders in vivo and cell culture passaging in vitro induce a functional decline of MSCs, switching MSCs to a senescent status with impaired self-renewal capacity and biased differentiation tendency. MSC functional decline accounts for the pathogenesis of many diseases and, more importantly, limits the large-scale applications of MSCs in regenerative medicine. Growing evidence implies that epigenetic mechanisms are a critical regulator of the differentiation programs for cell fate and are subject to changes during aging. Thus, we here review epigenetic dysregulations that contribute to MSC aging and osteoporosis. Comprehending detailed epigenetic mechanisms could provide us with a novel horizon for dissecting MSC-related pathogenesis and further optimizing MSC-mediated regenerative therapies.
Collapse
|
125
|
Salminen A, Kaarniranta K, Kauppinen A. Exosomal vesicles enhance immunosuppression in chronic inflammation: Impact in cellular senescence and the aging process. Cell Signal 2020; 75:109771. [PMID: 32896608 DOI: 10.1016/j.cellsig.2020.109771] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022]
Abstract
Exosomes represent an evolutionarily conserved signaling pathway which can act as an alarming mechanism in responses to diverse stresses, e.g. chronic inflammation activates the budding of exosomal vesicles in both immune and non-immune cells. Exosomes can contain both pro- and anti-inflammatory cargos but in chronic inflammation, exosomes mostly carry immunosuppressive cargos, e.g. enzymes and miRNAs. The aging process is associated with chronic low-grade inflammation and the accumulation of pro-inflammatory senescent cells into tissues. There is clear evidence that aging increases the number of exosomes in both the circulation and tissues. Especially, the secretion of immunosuppressive exosomes robustly increases from senescent cells. There are observations that the exosomes from senescent cells are involved in the expansion of senescence into neighbouring cells. Interestingly, the age-related exosomes contain immune suppressive cargos which enhance the immunosuppression within recipient immune cells, i.e. tissue-resident and recruited immune cells including M2 macrophages, myeloid-derived suppressor cells (MDSC), and regulatory T cells (Treg). It seems that increased immunosuppression with aging impairs the clearance of senescent cells and their accumulation within tissues augments the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029 Kuopio, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
126
|
Saheera S, Potnuri AG, Krishnamurthy P. Nano-Vesicle (Mis)Communication in Senescence-Related Pathologies. Cells 2020; 9:E1974. [PMID: 32859053 PMCID: PMC7564330 DOI: 10.3390/cells9091974] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles are a heterogeneous group of cell-derived membranous structures comprising of exosomes, apoptotic bodies, and microvesicles. Of the extracellular vesicles, exosomes are the most widely sorted and extensively explored for their contents and function. The size of the nanovesicular structures (exosomes) range from 30 to 140 nm and are present in various biological fluids such as saliva, plasma, urine etc. These cargo-laden extracellular vesicles arise from endosome-derived multivesicular bodies and are known to carry proteins and nucleic acids. Exosomes are involved in multiple physiological and pathological processes, including cellular senescence. Exosomes mediate signaling crosstalk and play a critical role in cell-cell communications. Exosomes have evolved as potential biomarkers for aging-related diseases. Aging, a physiological process, involves a progressive decline of function of organs with a loss of homeostasis and increasing probability of illness and death. The review focuses on the classic view of exosome biogenesis, biology, and age-associated changes. Owing to their ability to transport biological information among cells, the review also discusses the interplay of senescent cell-derived exosomes with the aging process, including the susceptibility of the aging population to COVID-19 infections.
Collapse
Affiliation(s)
- Sherin Saheera
- Department of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA;
| | - Ajay Godwin Potnuri
- Department of Animal Physiology, Indian Council for Medical Research—National Animal Resource Facility for Biomedical Research, Genome Valley, Shamirpet, Hyderabad, Telangana 500078, India;
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham, 1675 University Blvd, Volker Hall G094, Birmingham, AL 35294, USA
| |
Collapse
|
127
|
Gao Y, Patil S, Qian A. The Role of MicroRNAs in Bone Metabolism and Disease. Int J Mol Sci 2020; 21:ijms21176081. [PMID: 32846921 PMCID: PMC7503277 DOI: 10.3390/ijms21176081] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Bone metabolism is an intricate process involving various bone cells, signaling pathways, cytokines, hormones, growth factors, etc., and the slightest deviation can result in various bone disorders including osteoporosis, arthropathy, and avascular necrosis of femoral head. Osteoporosis is one of the most prevalent disorders affecting the skeleton, which is characterized by low bone mass and bone mineral density caused by the disruption in the balanced process of bone formation and bone resorption. The current pharmaceutical treatments such as bisphosphonates, selective estrogen receptor modulator, calcitonin, teriparatide, etc., could decrease the risk of fractures but have side-effects that have limited their long term applications. MicroRNAs (miRNAs) are one of many non-coding RNAs. These are single-stranded with a length of 19–25 nucleotides and can influence various cellular processes and play an important role in various diseases. Therefore, in this article, we review the different functions of different miRNA in bone metabolism and osteoporosis to understand their mechanism of action for the development of possible therapeutics.
Collapse
Affiliation(s)
- Yongguang Gao
- Laboratory for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (Y.G.); (S.P.)
- Department of Chemistry, Tangshan Normal University, Tangshan 063000, China
| | - Suryaji Patil
- Laboratory for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (Y.G.); (S.P.)
| | - Airong Qian
- Laboratory for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (Y.G.); (S.P.)
- Correspondence: ; Tel.: +86-135-7210-8260
| |
Collapse
|
128
|
Yan HC, Yu TT, Li J, Qiao YQ, Wang LC, Zhang T, Li Q, Zhou YH, Liu DW. The Delivery of Extracellular Vesicles Loaded in Biomaterial Scaffolds for Bone Regeneration. Front Bioeng Biotechnol 2020; 8:1015. [PMID: 32974327 PMCID: PMC7466762 DOI: 10.3389/fbioe.2020.01015] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous nanoparticles actively released by cells that comprise highly conserved and efficient systems of intercellular communication. In recent years, numerous studies have proven that EVs play an important role in the field of bone tissue engineering (BTE) due to several advantages, such as good biosafety, stability and efficient delivery. However, the application of EVs therapies in bone regeneration has not been widely used. One of the major challenges for the application of EVs is the lack of sufficient scaffolds to load and control the release of EVs. Thus, in this review, we describe the most advanced current strategies for delivering EVs with various biomaterials for the use in bone regeneration, the role of EVs in bone regeneration, the distribution of EVs mediated by biomaterials and common methods of promoting EVs delivery efficacy with a focus on biomaterial properties.
Collapse
Affiliation(s)
- Hui-Chun Yan
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Ting-Ting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Jing Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yi-Qiang Qiao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin-Chuan Wang
- Eastman Institute for Oral Health, University of Rochester, Rochester, NY, United States
| | - Ting Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Qian Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yan-Heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Da-Wei Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
129
|
Kim M, Jo H, Kwon Y, Kim Y, Jung HS, Jeoung D. Homoharringtonine Inhibits Allergic Inflammations by Regulating NF-κB-miR-183-5p-BTG1 Axis. Front Pharmacol 2020; 11:1032. [PMID: 32733254 PMCID: PMC7358642 DOI: 10.3389/fphar.2020.01032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/25/2020] [Indexed: 12/28/2022] Open
Abstract
Homoharringtonine (HHT) is a drug for treatment of chronic myeloid leukemia. However, the role of HHT in allergic inflammations remains unknown. Mouse model of atopic dermatitis (AD) induced by 2, 4,-dinitroflurobenzene (DNFB) and anaphylaxis employing 2,4-dinitropheny-human serum albumin (DNP-HSA) were used to examine the role of HHT in allergic inflammations. HHT inhibited in vitro allergic reactions and attenuated clinical symptoms associated with AD. DNFB induced features of allergic reactions in rat basophilic leukemia (RBL2H3) cells. HHT suppressed effect of AD on the expression of Th1/Th2 cytokines. HHT inhibited passive cutaneous anaphylaxis and passive systemic anaphylaxis. MiR-183-5p, increased by antigen stimulation, was downregulated by HHT in RBL2H3 cells. MiR-183-5p inhibitor suppressed anaphylaxis and AD. B cell translocation gene 1 (BTG1) was shown to be a direct target of miR-183-5p. BTG1 prevented antigen from inducing molecular features of in vitro allergic reactions. AD increased the expression of NF-κB, and NF-κB showed binding to the promoter sequences of miR-183-5p. NF-κB and miR-183 formed positive feedback to mediate in vitro allergic reactions. Thus, HHT can be an anti-allergy drug. We present evidence that NF-κB-miR-183-5p-BTG1 axis can serve as target for development of anti-allergy drug.
Collapse
Affiliation(s)
- Misun Kim
- Department of Biochemistry, Kangwon National University, Chunchon, South Korea
| | - Hyein Jo
- Department of Biochemistry, Kangwon National University, Chunchon, South Korea
| | - Yoojung Kwon
- Department of Biochemistry, Kangwon National University, Chunchon, South Korea
| | - Youngmi Kim
- College of Medicine, Institute of New Frontier Research, Hallym University, Chunchon, South Korea
| | - Hyun Suk Jung
- Department of Biochemistry, Kangwon National University, Chunchon, South Korea
| | - Dooil Jeoung
- Department of Biochemistry, Kangwon National University, Chunchon, South Korea
| |
Collapse
|
130
|
Tu C, He J, Chen R, Li Z. The Emerging Role of Exosomal Non-coding RNAs in Musculoskeletal Diseases. Curr Pharm Des 2020; 25:4523-4535. [PMID: 31724510 DOI: 10.2174/1381612825666191113104946] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023]
Abstract
Exosomes are phospholipid bilayer-enclosed membrane vesicles derived and constitutively secreted by various metabolically active cells. They are capable of mediating hetero- and homotypic intercellular communication by transferring multiple cargos from donor cells to recipient cells. Nowadays, non-coding RNAs (ncRNAs) have emerged as novel potential biomarkers or disease-targeting agents in a variety of diseases. However, the lack of effective delivery systems may impair their clinical application. Recently, accumulating evidence demonstrated that ncRNAs could be efficiently delivered to recipient cells using exosomes as a carrier, and therefore can exert a critical role in musculoskeletal diseases including osteoarthritis, rheumatoid arthritis, osteoporosis, muscular dystrophies, osteosarcoma and other diseases. Herein, we present an extensive review of biogenesis, physiological relevance and clinical implication of exosome-derived ncRNAs in musculoskeletal diseases.
Collapse
Affiliation(s)
- Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Jieyu He
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Ruiqi Chen
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
131
|
Periyasamy-Thandavan S, Burke J, Mendhe B, Kondrikova G, Kolhe R, Hunter M, Isales CM, Hamrick MW, Hill WD, Fulzele S. MicroRNA-141-3p Negatively Modulates SDF-1 Expression in Age-Dependent Pathophysiology of Human and Murine Bone Marrow Stromal Cells. J Gerontol A Biol Sci Med Sci 2020; 74:1368-1374. [PMID: 31505568 DOI: 10.1093/gerona/gly186] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Indexed: 12/11/2022] Open
Abstract
Stromal cell-derived factor-1 (SDF-1 or CXCL12) is a cytokine secreted by cells including bone marrow stromal cells (BMSCs). SDF-1 plays a vital role in BMSC migration, survival, and differentiation. Our group previously reported the role of SDF-1 in osteogenic differentiation in vitro and bone formation in vivo; however, our understanding of the post-transcriptional regulatory mechanism of SDF-1 remains poor. MicroRNAs are small noncoding RNAs that post-transcriptionally regulate the messenger RNAs (mRNAs) of protein-coding genes. In this study, we aimed to investigate the impact of miR-141-3p on SDF-1 expression in BMSCs and its importance in the aging bone marrow (BM) microenvironment. Our data demonstrated that murine and human BMSCs expressed miR-141-3p that repressed SDF-1 gene expression at the functional level (luciferase reporter assay) by targeting the 3'-untranslated region of mRNA. We also found that transfection of miR-141-3p decreased osteogenic markers in human BMSCs. Our results demonstrate that miR-141-3p expression increases with age, while SDF-1 decreases in both the human and mouse BM niche. Taken together, these results support that miR-141-3p is a novel regulator of SDF-1 in bone cells and plays an important role in the age-dependent pathophysiology of murine and human BM niche.
Collapse
Affiliation(s)
| | - John Burke
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia
| | - Bharati Mendhe
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia
| | - Galina Kondrikova
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, Georgia
| | - Monte Hunter
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia
| | - Carlos M Isales
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Georgia
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia.,Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Georgia
| | - William D Hill
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia.,Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Georgia.,Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Sadanand Fulzele
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Georgia.,Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Georgia.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Georgia
| |
Collapse
|
132
|
Ha DH, Kim HK, Lee J, Kwon HH, Park GH, Yang SH, Jung JY, Choi H, Lee JH, Sung S, Yi YW, Cho BS. Mesenchymal Stem/Stromal Cell-Derived Exosomes for Immunomodulatory Therapeutics and Skin Regeneration. Cells 2020; 9:E1157. [PMID: 32392899 PMCID: PMC7290908 DOI: 10.3390/cells9051157] [Citation(s) in RCA: 304] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/25/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022] Open
Abstract
Exosomes are nano-sized vesicles that serve as mediators for cell-to-cell communication. With their unique nucleic acids, proteins, and lipids cargo compositions that reflect the characteristics of producer cells, exosomes can be utilized as cell-free therapeutics. Among exosomes derived from various cellular origins, mesenchymal stem cell-derived exosomes (MSC-exosomes) have gained great attention due to their immunomodulatory and regenerative functions. Indeed, many studies have shown anti-inflammatory, anti-aging and wound healing effects of MSC-exosomes in various in vitro and in vivo models. In addition, recent advances in the field of exosome biology have enabled development of specific guidelines and quality control methods, which will ultimately lead to clinical application of exosomes. This review highlights recent studies that investigate therapeutic potential of MSC-exosomes and relevant mode of actions for skin diseases, as well as quality control measures required for development of exosome-derived therapeutics.
Collapse
Affiliation(s)
- Dae Hyun Ha
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Hyun-keun Kim
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Joon Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea;
| | | | - Gyeong-Hun Park
- Department of Dermatology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwasweong-si, Gyeonggi-do 18450, Korea;
| | | | | | | | - Jun Ho Lee
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Sumi Sung
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Yong Weon Yi
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| | - Byong Seung Cho
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul 08594, Korea; (D.H.H.); (H.-k.K.); (J.H.L.); (S.S.)
| |
Collapse
|
133
|
Jia Y, Qiu S, Xu J, Kang Q, Chai Y. Exosomes Secreted by Young Mesenchymal Stem Cells Promote New Bone Formation During Distraction Osteogenesis in Older Rats. Calcif Tissue Int 2020; 106:509-517. [PMID: 32103287 DOI: 10.1007/s00223-019-00656-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023]
Abstract
Distraction osteogenesis (DO) is a clinically effective procedure to regenerate large bone defects. However, the treatment duration is undesirably lengthy, especially in elderly patients. Exosomes derived from mesenchymal stem cells (MSC-Exos) could exert the beneficial effects while avoiding the possible complications of stem cell transplantation. This study aimed to evaluate the effects of MSC-Exos on bone regeneration during DO in older rats. Exosomes were isolated from the supernatants of young bone marrow mesenchymal stem cells (BMSCs) through ultra-centrifugation, and characterized using transmission electron microscopy, western blot, and tunable resistive pulse sensing analysis. The effects of MSC-Exos on the proliferation and differentiation of older BMSCs were evaluated using CCK-8 assay, ALP and ARS staining, and qRT-PCR. Unilateral tibial DO model was established on older Sprague-Dawley rats and MSC-Exos or phosphate buffer saline was locally injected into the distraction gaps after distraction weekly. Bone regeneration were evaluated using X-ray, Micro-CT, mechanical test, and histological staining. The MSC-Exos were round or cup-shaped vesicles ranging from 60 to 130 nm in diameter and expressed markers including CD9, CD63, and TSG101. The in vitro results indicated that MSC-Exos could enhance the proliferation and osteogenic differentiation of older BMSCs. Bone regeneration was markedly accelerated in rats treated with MSC-Exos according to the results of X-ray, micro-CT, and histological analysis. The distracted tibias from the MSC-Exos group also demonstrated better mechanical properties. These results suggest that MSC-Exos promote DO-mediated bone regeneration in older rats through enhancing the proliferation and osteogenic capacity of BMSCs.
Collapse
Affiliation(s)
- Yachao Jia
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China
| | - Shuo Qiu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China
| | - Qinglin Kang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China.
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China.
| |
Collapse
|
134
|
Chen Y, Tang L. Stem Cell Senescence: the Obstacle of the Treatment of Degenerative Disk Disease. Curr Stem Cell Res Ther 2020; 14:654-668. [PMID: 31490764 DOI: 10.2174/1574888x14666190906163253] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/05/2019] [Accepted: 06/01/2019] [Indexed: 12/14/2022]
Abstract
Intervertebral disc (IVD) has a pivotal role in the maintenance of flexible motion. IVD degeneration is one of the primary causes of low back pain and disability, which seriously influences patients' health, and increases the family and social economic burden. Recently, stem cell therapy has been proven to be more effective on IVD degeneration disease. However, stem cell senescence is the limiting factor in the IVD degeneration treatment. Senescent stem cells have a negative effect on the self-repair on IVD degeneration. In this review, we delineate that the factors such as telomerase shortening, DNA damage, oxidative stress, microenvironment and exosomes will induce stem cell aging. Recent studies tried to delay the aging of stem cells by regulating the expression of aging-related genes and proteins, changing the activity of telomerase, improving the survival microenvironment of stem cells and drug treatment. Understanding the mechanism of stem cell aging and exploring new approaches to delay or reverse stem cell aging asks for research on the repair of the degenerated disc.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering Chongqing University, Chongqing 400044, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering Chongqing University, Chongqing 400044, China
| |
Collapse
|
135
|
Hong Y, He H, Jiang G, Zhang H, Tao W, Ding Y, Yuan D, Liu J, Fan H, Lin F, Liang X, Li X, Zhang Y. miR-155-5p inhibition rejuvenates aged mesenchymal stem cells and enhances cardioprotection following infarction. Aging Cell 2020; 19:e13128. [PMID: 32196916 PMCID: PMC7189985 DOI: 10.1111/acel.13128] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 01/22/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
Aging impairs the functions of human mesenchymal stem cells (MSCs), thereby severely reducing their beneficial effects on myocardial infarction (MI). MicroRNAs (miRNAs) play crucial roles in regulating the senescence of MSCs; however, the underlying mechanisms remain unclear. Here, we investigated the significance of miR-155-5p in regulating MSC senescence and whether inhibition of miR-155-5p could rejuvenate aged MSCs (AMSCs) to enhance their therapeutic efficacy for MI. Young MSCs (YMSCs) and AMSCs were isolated from young and aged donors, respectively. The cellular senescence of MSCs was evaluated by senescence-associated β-galactosidase (SA-β-gal) staining. Compared with YMSCs, AMSCs exhibited increased cellular senescence as evidenced by increased SA-β-gal activity and decreased proliferative capacity and paracrine effects. The expression of miR-155-5p was much higher in both serum and MSCs from aged donors than young donors. Upregulation of miR-155-5p in YMSCs led to increased cellular senescence, whereas downregulation of miR-155-5p decreased AMSC senescence. Mechanistically, miR-155-5p inhibited mitochondrial fission and increased mitochondrial fusion in MSCs via the AMPK signaling pathway, thereby resulting in cellular senescence by repressing the expression of Cab39. These effects were partially reversed by treatment with AMPK activator or mitofusin2-specific siRNA (Mfn2-siRNA). By enhancing angiogenesis and promoting cell survival, transplantation of anti-miR-155-5p-AMSCs led to improved cardiac function in an aged mouse model of MI compared with transplantation of AMSCs. In summary, our study shows that miR-155-5p mediates MSC senescence by regulating the Cab39/AMPK signaling pathway and miR-155-5p is a novel target to rejuvenate AMSCs and enhance their cardioprotective effects.
Collapse
Affiliation(s)
- Yimei Hong
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesSchool of MedicineSouth China University of TechnologyGuangzhouChina
- Department of Emergency MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
- Department of Emergency and Critical Care MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Haiwei He
- Department of Emergency MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
- Department of Emergency and Critical Care MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Guojun Jiang
- Faculty of PharmacyBengbu Medical CollegeBengbuChina
| | - Hao Zhang
- Faculty of PharmacyBengbu Medical CollegeBengbuChina
| | - Wuyuan Tao
- Department of Emergency MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
- Department of Emergency and Critical Care MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Yue Ding
- Department of Organ TransplantationChangzheng HospitalSecond Military Medical UniversityShanghaiChina
| | - Dongsheng Yuan
- Clinical Translational Medical Research CenterShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jing Liu
- Clinical Translational Medical Research CenterShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Huimin Fan
- Clinical Translational Medical Research CenterShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Fang Lin
- Clinical Translational Medical Research CenterShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Xiaoting Liang
- Clinical Translational Medical Research CenterShanghai East HospitalTongji University School of MedicineShanghaiChina
- Institute of Regenerative MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Xin Li
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesSchool of MedicineSouth China University of TechnologyGuangzhouChina
- Department of Emergency MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
- Department of Emergency and Critical Care MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Yuelin Zhang
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesSchool of MedicineSouth China University of TechnologyGuangzhouChina
- Department of Emergency MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
- Department of Emergency and Critical Care MedicineGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| |
Collapse
|
136
|
The Emerging Role of Senescence in Ocular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2583601. [PMID: 32215170 PMCID: PMC7085400 DOI: 10.1155/2020/2583601] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
Cellular senescence is a state of irreversible cell cycle arrest in response to an array of cellular stresses. An important role for senescence has been shown for a number of pathophysiological conditions that include cardiovascular disease, pulmonary fibrosis, and diseases of the skin. However, whether senescence contributes to the progression of age-related macular degeneration (AMD) has not been studied in detail so far and the present review describes the recent research on this topic. We present an overview of the types of senescence, pathways of senescence, senescence-associated secretory phenotype (SASP), the role of mitochondria, and their functional implications along with antisenescent therapies. As a central mechanism, senescent cells can impact the surrounding tissue microenvironment via the secretion of a pool of bioactive molecules, termed the SASP. An updated summary of a number of new members of the ever-growing SASP family is presented. Further, we introduce the significance of mechanisms by which mitochondria may participate in the development of cellular senescence. Emerging evidence shows that extracellular vesicles (EVs) are important mediators of the effects of senescent cells on their microenvironment. Based on recent studies, there is reasonable evidence that senescence could be a modifiable factor, and hence, it may be possible to delay age-related diseases by modulating basic aging mechanisms using SASP inhibitors/senolytic drugs. Thus, antisenescent therapies in aging and age-related diseases appear to have a promising potential.
Collapse
|
137
|
Boulestreau J, Maumus M, Rozier P, Jorgensen C, Noël D. Mesenchymal Stem Cell Derived Extracellular Vesicles in Aging. Front Cell Dev Biol 2020; 8:107. [PMID: 32154253 PMCID: PMC7047768 DOI: 10.3389/fcell.2020.00107] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/10/2020] [Indexed: 12/17/2022] Open
Abstract
Aging is associated with high prevalence of chronic degenerative diseases that take a large part of the increasing burden of morbidities in a growing demographic of elderly people. Aging is a complex process that involves cell autonomous and cell non-autonomous mechanisms where senescence plays an important role. Senescence is characterized by the loss of proliferative potential, resistance to cell death by apoptosis and expression of a senescence-associated secretory phenotype (SASP). SASP includes pro-inflammatory cytokines and chemokines, tissue-damaging proteases, growth factors; all contributing to tissue microenvironment alteration and loss of tissue homeostasis. Emerging evidence suggests that the changes in the number and composition of extracellular vesicles (EVs) released by senescent cells contribute to the adverse effects of senescence in aging. In addition, age-related alterations in mesenchymal stem/stromal cells (MSCs) have been associated to dysregulated functions. The loss of functional stem cells necessary to maintain tissue homeostasis likely directly contributes to aging. In this review, we will focus on the characteristics and role of EVs isolated from senescent MSCs, the potential effect of MSC-derived EVs in aging and discuss their therapeutic potential to improve age-related diseases.
Collapse
Affiliation(s)
- Jérémy Boulestreau
- Institute of Regenerative Medicine and Biotherapies (IRMB), University of Montpellier, INSERM, Montpellier, France
| | - Marie Maumus
- Institute of Regenerative Medicine and Biotherapies (IRMB), University of Montpellier, INSERM, Montpellier, France
| | - Pauline Rozier
- Institute of Regenerative Medicine and Biotherapies (IRMB), University of Montpellier, INSERM, Montpellier, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies (IRMB), University of Montpellier, INSERM, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, CHU, Montpellier, France
| | - Danièle Noël
- Institute of Regenerative Medicine and Biotherapies (IRMB), University of Montpellier, INSERM, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, CHU, Montpellier, France
| |
Collapse
|
138
|
Topping LM, Thomas BL, Rhys HI, Tremoleda JL, Foster M, Seed M, Voisin MB, Vinci C, Law HL, Perretti M, Norling LV, Azevedo HS, Nissim A. Targeting Extracellular Vesicles to the Arthritic Joint Using a Damaged Cartilage-Specific Antibody. Front Immunol 2020; 11:10. [PMID: 32117219 PMCID: PMC7033748 DOI: 10.3389/fimmu.2020.00010] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/06/2020] [Indexed: 01/18/2023] Open
Abstract
The targeted delivery of therapies to diseased tissues offers a safe opportunity to achieve optimal efficacy while limiting systemic exposure. These considerations apply to many disease indications but are especially relevant for rheumatoid arthritis (RA), as RA is a systemic autoimmune disease which affects multiple joints. We have identified an antibody that is specific to damaged arthritic cartilage (anti-ROS-CII) that can be used to deliver treatments specifically to arthritic joints, yielding augmented efficacy in experimental arthritis. In the current study, we demonstrate that scaffolds enriched with bioactive payloads can be delivered precisely to an inflamed joint and achieve superior efficacy outcomes consistent with the pharmacological properties of these payloads. As a scaffold, we have used extracellular vesicles (EVs) prepared from human neutrophils (PMNs), which possess intrinsic anti-inflammatory properties and the ability to penetrate inflamed arthritic cartilage. EV fortified with anti-ROS-CII (EV/anti-ROS-CII) retained anti-ROS-CII specificity and bound exclusively to the damaged cartilage. Following systemic administration, EV/anti-ROS-CII (a) exhibited the ability to localize specifically in the arthritic joint in vivo and (b) was able to specifically target single (viral IL-10 or anti-TNF) or combined (viral IL-10 and anti-TNF) anti-inflammatory treatments to the arthritic joint, which accelerated attenuation of clinical and synovial inflammation. Overall, this study demonstrates the attainability of targeting a pro-resolving biological scaffold to the arthritic joint. The potential of targeting scaffolds such as EV, nanoparticles, or a combination thereof alongside combined therapeutics is paramount for designing systemically administered broad-spectrum of anti-inflammatory treatments.
Collapse
Affiliation(s)
- Louise M Topping
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Centre for Bioengineering, Life Sciences, Queen Mary University of London, London, United Kingdom
| | - Bethan L Thomas
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Hefin I Rhys
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Jordi L Tremoleda
- Barts and the London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Martyn Foster
- Experimental Pathology Consultancy, London, United Kingdom
| | - Michael Seed
- School of Health Sport and Bioscience, University of East London, London, United Kingdom
| | - Mathieu-Benoit Voisin
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Chiara Vinci
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Hannah L Law
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Mauro Perretti
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Centre for Bioengineering, Life Sciences, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Lucy V Norling
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Helena S Azevedo
- Centre for Bioengineering, Life Sciences, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom.,School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary University of London, London, United Kingdom
| | - Ahuva Nissim
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Centre for Bioengineering, Life Sciences, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
139
|
Dalton S, Smith K, Singh K, Kaiser H, Kolhe R, Mondal AK, Khayrullin A, Isales CM, Hamrick MW, Hill WD, Fulzele S. Accumulation of kynurenine elevates oxidative stress and alters microRNA profile in human bone marrow stromal cells. Exp Gerontol 2020; 130:110800. [PMID: 31790802 PMCID: PMC6998036 DOI: 10.1016/j.exger.2019.110800] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 02/09/2023]
Abstract
Kynurenine, a metabolite of tryptophan breakdown, has been shown to increase with age, and plays a vital role in a number of age-related pathophysiological changes, including bone loss. Accumulation of kynurenine in bone marrow stromal cells (BMSCs) has been associated with a decrease in cell proliferation and differentiation, though the exact mechanism by which kynurenine mediates these changes is poorly understood. MiRNAs have been shown to regulate BMSC function, and accumulation of kynurenine may alter the miRNA expression profile of BMSCs. The aim of this study was to identify differentially expressed miRNAs in human BMSCs in response to treatment with kynurenine, and correlate miRNAs function in BMSCs biology through bioinformatics analysis. Human BMSCs were cultured and treated with and without kynurenine, and subsequent miRNA isolation was performed. MiRNA array was performed to identify differentially expressed miRNA. Microarray analysis identified 50 up-regulated, and 36 down-regulated miRNAs in kynurenine-treated BMSC cultures. Differentially expressed miRNA included miR-1281, miR-330-3p, let-7f-5p, and miR-493-5p, which are important for BMSC proliferation and differentiation. KEGG analysis found up-regulated miRNA targeting glutathione metabolism, a pathway critical for removing oxidative species. Our data support that the kynurenine dependent degenerative effect is partially due to changes in the miRNA profile of BMSCs.
Collapse
Affiliation(s)
- Sherwood Dalton
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - Kathryn Smith
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - Kanwar Singh
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - Helen Kaiser
- Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America
| | - Ravindra Kolhe
- Departments of Pathology, Augusta University, Augusta, GA 30912, United States of America
| | - Ashis K Mondal
- Departments of Pathology, Augusta University, Augusta, GA 30912, United States of America
| | - Andrew Khayrullin
- Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America
| | - Carlos M Isales
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Medicine, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America
| | - Mark W Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC, 29403, United States of America
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America.
| |
Collapse
|
140
|
Ullah M, Ng NN, Concepcion W, Thakor AS. Emerging role of stem cell-derived extracellular microRNAs in age-associated human diseases and in different therapies of longevity. Ageing Res Rev 2020; 57:100979. [PMID: 31704472 DOI: 10.1016/j.arr.2019.100979] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/17/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022]
Abstract
Organismal aging involves the progressive decline in organ function and increased susceptibility to age-associated diseases. This has been associated with the aging of stem cell populations within the body that decreases the capacity of stem cells to self-renew, differentiate, and regenerate damaged tissues and organs. This review aims to explore how aging is associated with the dysregulation of stem cell-derived extracellular vesicles (SCEVs) and their corresponding miRNA cargo (SCEV-miRNAs), which are short non-coding RNAs involved in post-transcriptional regulation of target genes. Recent evidence has suggested that in aging stem cells, SCEV-miRNAs may play a vital role regulating various processes that contribute to aging: cellular senescence, stem cell exhaustion, telomere length, and circadian rhythm. Hence, further clarifying the age-dependent molecular mechanisms through which SCEV-miRNAs exert their downstream effects may inform a greater understanding of the biology of aging, elucidate their role in stem cell function, and identify important targets for future regenerative therapies. Additionally, current studies evaluating therapeutic role of SCEVs and SCEV-miRNAs in treating several age-associated diseases are also discussed.
Collapse
Affiliation(s)
- Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California, 94304, USA.
| | - Nathan Norton Ng
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California, 94304, USA
| | - Waldo Concepcion
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California, 94304, USA
| |
Collapse
|
141
|
Smirlis D, Dingli F, Pescher P, Prina E, Loew D, Rachidi N, Späth GF. SILAC-based quantitative proteomics reveals pleiotropic, phenotypic modulation in primary murine macrophages infected with the protozoan pathogen Leishmania donovani. J Proteomics 2019; 213:103617. [PMID: 31846769 DOI: 10.1016/j.jprot.2019.103617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 01/06/2023]
Abstract
Leishmaniases are major vector-borne tropical diseases responsible for great human morbidity and mortality, caused by protozoan, trypanosomatid parasites of the genus Leishmania. In the mammalian host, parasites survive and multiply within mononuclear phagocytes, especially macrophages. However, the underlying mechanisms by which Leishmania spp. affect their host are not fully understood. Herein, proteomic alterations of primary, bone marrow-derived BALB/c macrophages are documented after 72 h of infection with Leishmania donovani insect-stage promastigotes, applying a SILAC-based, quantitative proteomics approach. The protocol was optimised by combining strong anion exchange and gel electrophoresis fractionation that displayed similar depth of analysis (combined total of 6189 mouse proteins). Our analyses revealed 86 differentially modulated proteins (35 showing increased and 51 decreased abundance) in response to Leishmania donovani infection. The proteomics results were validated by analysing the abundance of selected proteins. Intracellular Leishmania donovani infection led to changes in various host cell biological processes, including primary metabolism and catabolic process, with a significant enrichment in lysosomal organisation. Overall, our analysis establishes the first proteome of bona fide primary macrophages infected ex vivo with Leishmania donovani, revealing new mechanisms acting at the host/pathogen interface. SIGNIFICANCE: Little is known on proteome changes that occur in primary macrophages after Leishmania donovani infection. This study describes a SILAC-based quantitative proteomics approach to characterise changes of bone marrow-derived macrophages infected with L. donovani promastigotes for 72 h. With the application of SILAC and the use of SAX and GEL fractionation methods, we have tested new routes for proteome quantification of primary macrophages. The protocols developed here can be applicable to other diseases and pathologies. Moreover, this study sheds important new light on the "proteomic reprogramming" of infected macrophages in response to L. donovani promastigotes that affects primary metabolism, cellular catabolic processes, and lysosomal/vacuole organisation. Thus, our study reveals key molecules and processes that act at the host/pathogen interface that may inform on new immuno- or chemotherapeutic interventions to combat leishmaniasis.
Collapse
Affiliation(s)
- Despina Smirlis
- Institut Pasteur and Institut National de Santé et Recherche Médicale INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France; Hellenic Pasteur Institute, Molecular Parasitology Laboratory, Athens, Greece.
| | - Florent Dingli
- Laboratoire de Spectrométrie de Masse Protéomique, Centre de Recherche, Institut Curie, Université de recherche PSL, Paris, France
| | - Pascale Pescher
- Institut Pasteur and Institut National de Santé et Recherche Médicale INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Eric Prina
- Institut Pasteur and Institut National de Santé et Recherche Médicale INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Damarys Loew
- Laboratoire de Spectrométrie de Masse Protéomique, Centre de Recherche, Institut Curie, Université de recherche PSL, Paris, France
| | - Najma Rachidi
- Institut Pasteur and Institut National de Santé et Recherche Médicale INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Gerald F Späth
- Institut Pasteur and Institut National de Santé et Recherche Médicale INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France.
| |
Collapse
|
142
|
Abstract
PURPOSE OF REVIEW Extracellular vesicles (EV), which include exosomes and microvesicles, are membrane-bound particles shed by most cell types and are important mediators of cell-cell communication by delivering their cargo of proteins, miRNA, and mRNA to target cells and altering their function. Here, we provide an overview of what is currently known about EV composition and function in bone and muscle cells and discuss their role in mediating crosstalk between these two tissues as well as their role in musculoskeletal aging. RECENT FINDINGS Recent studies have shown that muscle and bone cells produce EV, whose protein, mRNA, and miRNA cargo reflects the differentiated state of the parental cells. These EV have functional effects within their respective tissues, but evidence is accumulating that they are also shed into the circulation and can have effects on distant tissues. Bone- and muscle-derived EV can alter the differentiation and function of bone and muscle cells. Many of these effects are mediated via small microRNAs that regulate target genes in recipient cells. EV-mediated signaling in muscle and bone is an exciting and emerging field. While considerable progress has been made, much is still to be discovered about the mechanisms regulating EV composition, release, uptake, and function in muscle and bone. A key challenge is to understand more precisely how exosomes function in truly physiological settings.
Collapse
Affiliation(s)
- Weiping Qin
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Veterans Affairs Medical Center, Bronx, New York, NY, 10468, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri Kansas City, 650 E. 25th Street, Kansas City, MO, 64108, USA.
| |
Collapse
|
143
|
Griukova A, Deryabin P, Shatrova A, Burova E, Severino V, Farina A, Nikolsky N, Borodkina A. Molecular basis of senescence transmitting in the population of human endometrial stromal cells. Aging (Albany NY) 2019; 11:9912-9931. [PMID: 31689238 PMCID: PMC6874437 DOI: 10.18632/aging.102441] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022]
Abstract
Hormone-regulated proliferation and differentiation of endometrial stromal cells (ESCs) determine overall endometrial plasticity and receptivity to embryos. Previously we revealed that ESCs may undergo premature senescence, accompanied by proliferation loss and various intracellular alterations. Here we focused on whether and how senescence may be transmitted within the ESCs population. We revealed that senescent ESCs may induce paracrine senescence in young counterparts via cell contacts, secreted factors and extracellular vesicles. According to secretome-wide profiling we identified plasminogen activator inhibitor -1 (PAI-1) to be the most prominent protein secreted by senescent ESCs (data are available via ProteomeXchange with identifier PXD015742). By applying CRISPR/Cas9 techniques we disclosed that PAI-1 secreted by senescent ESCs may serve as the master-regulator of paracrine senescence progression within the ESCs population. Unraveled molecular basis of senescence transduction in the ESCs population may be further considered in terms of altered endometrial plasticity and sensitivity to invading embryo, thus contributing to the female infertility curing.
Collapse
Affiliation(s)
- Anastasiia Griukova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Pavel Deryabin
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Alla Shatrova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Elena Burova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Valeria Severino
- Department of Medicine, University Medical Center (CMU), Faculty of Medicine, Geneva University, Geneva CH-1211, Switzerland
| | - Annarita Farina
- Department of Medicine, University Medical Center (CMU), Faculty of Medicine, Geneva University, Geneva CH-1211, Switzerland
| | - Nikolay Nikolsky
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Aleksandra Borodkina
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| |
Collapse
|
144
|
Lv L, Sheng C, Zhou Y. Extracellular vesicles as a novel therapeutic tool for cell-free regenerative medicine in oral rehabilitation. J Oral Rehabil 2019; 47 Suppl 1:29-54. [PMID: 31520537 DOI: 10.1111/joor.12885] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 07/26/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022]
Abstract
Oral maxillofacial defects may always lead to complicated hard and soft tissue loss, including bone, nerve, blood vessels, teeth and skin, which are difficult to restore and severely influence the life quality of patients. Extracellular vesicles (EVs), including exosomes, microvesicles and apoptotic bodies, are emerging as potential solutions for complex tissue regeneration through cell-free therapies. In this review, we highlight the functional roles of EVs in the regenerative medicine for oral maxillofacial rehabilitation, specifically bone, skin, blood vessels, peripheral nerve and tooth-related tissue regeneration. Publications were reviewed by two researchers independently basing on three databases (PubMed, MEDLINE and Web of Science), until 31 December 2018. Basing on current researches, we classified the origin of EVs for regenerative medicine into four categories: related cells in the regenerative niche, mesenchymal stem cells, immune cells and body fluids. The secretome of different cells are distinct, while the same cells secrete different EVs under varied conditions; therefore, the content profiles of EVs and regulatory mechanisms on target cells are compared and emphasised. By unravelling the regulatory mechanisms of EVs in tissue regeneration, modified cells and tailored EVs with specific target may be produced for precision medicine with high efficacy.
Collapse
Affiliation(s)
- Longwei Lv
- Department of Prosthodontics, National Clinical Research Center for Oral Disease, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Chunhui Sheng
- Department of Prosthodontics, National Clinical Research Center for Oral Disease, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, National Clinical Research Center for Oral Disease, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
145
|
O'Connor KC. Molecular Profiles of Cell-to-Cell Variation in the Regenerative Potential of Mesenchymal Stromal Cells. Stem Cells Int 2019; 2019:5924878. [PMID: 31636675 PMCID: PMC6766122 DOI: 10.1155/2019/5924878] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 08/20/2019] [Indexed: 12/22/2022] Open
Abstract
Cell-to-cell variation in the regenerative potential of mesenchymal stromal cells (MSCs) impedes the translation of MSC therapies into clinical practice. Cellular heterogeneity is ubiquitous across MSC cultures from different species and tissues. This review highlights advances to elucidate molecular profiles that identify cell subsets with specific regenerative properties in heterogeneous MSC cultures. Cell surface markers and global signatures are presented for proliferation and differentiation potential, as well as immunomodulation and trophic properties. Key knowledge gaps are discussed as potential areas of future research. Molecular profiles of MSC heterogeneity have the potential to enable unprecedented control over the regenerative potential of MSC therapies through the discovery of new molecular targets and as quality attributes to develop robust and reproducible biomanufacturing processes. These advances would have a positive impact on the nascent field of MSC therapeutics by accelerating the development of therapies with more consistent and effective treatment outcomes.
Collapse
Affiliation(s)
- Kim C. O'Connor
- Department of Chemical and Biomolecular Engineering, Tulane University, New Orleans, Louisiana, USA
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
146
|
D'Anca M, Fenoglio C, Serpente M, Arosio B, Cesari M, Scarpini EA, Galimberti D. Exosome Determinants of Physiological Aging and Age-Related Neurodegenerative Diseases. Front Aging Neurosci 2019; 11:232. [PMID: 31555123 PMCID: PMC6722391 DOI: 10.3389/fnagi.2019.00232] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/13/2019] [Indexed: 01/08/2023] Open
Abstract
Aging is consistently reported as the most important independent risk factor for neurodegenerative diseases. As life expectancy has significantly increased during the last decades, neurodegenerative diseases became one of the most critical public health problem in our society. The most investigated neurodegenerative diseases during aging are Alzheimer disease (AD), Frontotemporal Dementia (FTD) and Parkinson disease (PD). The search for biomarkers has been focused so far on cerebrospinal fluid (CSF) and blood. Recently, exosomes emerged as novel biological source with increasing interest for age-related neurodegenerative disease biomarkers. Exosomes are tiny Extracellular vesicles (EVs; 30-100 nm in size) released by all cell types which originate from the endosomal compartment. They constitute important vesicles for the release and transfer of multiple (signaling, toxic, and regulatory) molecules among cells. Initially considered with merely waste disposal function, instead exosomes have been recently recognized as fundamental mediators of intercellular communication. They can move from the site of release by diffusion and be retrieved in several body fluids, where they may dynamically reflect pathological changes of cells present in inaccessible sites such as the brain. Multiple evidence has implicated exosomes in age-associated neurodegenerative processes, which lead to cognitive impairment in later life. Critically, consolidated evidence indicates that pathological protein aggregates, including Aβ, tau, and α-synuclein are released from brain cells in association with exosomes. Importantly, exosomes act as vehicles between cells not only of proteins but also of nucleic acids [DNA, mRNA transcripts, miRNA, and non-coding RNAs (ncRNAs)] thus potentially influencing gene expression in target cells. In this framework, exosomes could contribute to elucidate the molecular mechanisms underneath neurodegenerative diseases and could represent a promising source of biomarkers. Despite the involvement of exosomes in age-associated neurodegeneration, the study of exosomes and their genetic cargo in physiological aging and in neurodegenerative diseases is still in its infancy. Here, we review, the current knowledge on protein and ncRNAs cargo of exosomes in normal aging and in age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Marianna D'Anca
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Faculty of Medicine and Surgery, University of Milan, Milan, Italy
| | - Chiara Fenoglio
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Faculty of Medicine and Surgery, University of Milan, Milan, Italy
| | - Maria Serpente
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Faculty of Medicine and Surgery, University of Milan, Milan, Italy
| | - Beatrice Arosio
- Department of Clinical Sciences and Community Health, Faculty of Medicine and Surgery, University of Milan, Milan, Italy
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, Faculty of Medicine and Surgery, University of Milan, Milan, Italy.,Geriatrics Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elio Angelo Scarpini
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Faculty of Medicine and Surgery, University of Milan, Milan, Italy.,Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, Faculty of Medicine and Surgery, University of Milan, Milan, Italy
| |
Collapse
|
147
|
The Role of Exosomes in Bone Remodeling: Implications for Bone Physiology and Disease. DISEASE MARKERS 2019; 2019:9417914. [PMID: 31485281 PMCID: PMC6710799 DOI: 10.1155/2019/9417914] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 12/18/2022]
Abstract
Bone remodeling represents a physiological phenomenon of continuous bone tissue renewal that requires fine orchestration of multiple cell types, which is critical for the understanding of bone disease but not yet clarified in precise detail. Exosomes, which are cell-secreted nanovesicles drawing increasing attention for their broad biosignaling functions, can shed new light on how multiple heterogeneous cells communicate for the purpose of bone remodeling. In the healthy bone, exosomes transmit signals favoring both bone synthesis and resorption, regulating the differentiation, recruitment, and activity of most cell types involved in bone remodeling and even assuming an active role in extracellular matrix mineralization. Additionally, in the ailing bone, they actively participate in pathogenic processes constituting also potential therapeutic agents and drug vectors. The present review summarizes the current knowledge on bone exosomes and bone remodeling in health and disease.
Collapse
|
148
|
Lange-Consiglio A, Lazzari B, Pizzi F, Stella A, Girani A, Quintè A, Cremonesi F, Capra E. Different Culture Times Affect MicroRNA Cargo in Equine Amniotic Mesenchymal Cells and Their Microvesicles. Tissue Eng Part C Methods 2019; 24:596-604. [PMID: 30234462 DOI: 10.1089/ten.tec.2018.0205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Conditioned medium (CM) and microvesicles (MVs) are produced using different protocols: CM is collected following 12-96 h of cell culture without renewal of tissue culture medium, while MVs are collected after overnight cell culture. For future comparative studies in regenerative medicine looking at the efficacy of CM and MVs, it is important to understand how the quality of cell secretions is affected by culture. The aim of this study was to evaluate whether the duration of culturing influences the micro-RNAs (miRNAs) cargo of equine amniotic mesenchymal cells (AMCs) and their MVs. The analysis identified 990 miRNAs. After one night, there were 347 differently expressed (DE)-miRNAs between MVs and cells, whereas after four nights there were 359. About 58.3% of the DE-miRNAs were shared between samples produced under the two conditions. The comparison between miRNA content in AMC cells cultured for one night versus four nights showed eight DE-Equus caballus (eca)-miRNAs, which target genes were involved in immune response to external stimulus, inflammatory response, and production of reactive oxygen species. Comparing MVs isolated from one or four nights, four DE-miRNAs that target genes regulating cell cycle progression and production of reactive oxygen species were found, but only eca-miR-214 was enriched in the MVs after four nights. In conclusion, after 4 days of cell culture, the profile of AMC miRNAs was altered, indicating a probable phenotypic transition versus a new cell culture environment and aging. After this time, MVs accumulated eca-miR-214, which may help cells survive or adapt to new culture conditions.
Collapse
Affiliation(s)
- Anna Lange-Consiglio
- 1 Department of Veterinary Medicine, Università Degli Studi di Milano , Milano, Italy .,2 Reproduction Unit, Centro Clinico-Veterinario e Zootecnico-Sperimentale di Ateneo , Università degli Studi di Milano, Lodi, Italy
| | - Barbara Lazzari
- 3 Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, via Einstein , Lodi, Italy
| | - Flavia Pizzi
- 3 Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, via Einstein , Lodi, Italy
| | - Alessandra Stella
- 3 Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, via Einstein , Lodi, Italy
| | - Alessia Girani
- 1 Department of Veterinary Medicine, Università Degli Studi di Milano , Milano, Italy
| | - Arianna Quintè
- 1 Department of Veterinary Medicine, Università Degli Studi di Milano , Milano, Italy
| | - Fausto Cremonesi
- 1 Department of Veterinary Medicine, Università Degli Studi di Milano , Milano, Italy .,2 Reproduction Unit, Centro Clinico-Veterinario e Zootecnico-Sperimentale di Ateneo , Università degli Studi di Milano, Lodi, Italy
| | - Emanuele Capra
- 3 Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, via Einstein , Lodi, Italy
| |
Collapse
|
149
|
Sun D, Song H, Shen Z. [Research progress in mesenchymal stem cells modified by Heme oxygenase 1]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:901-906. [PMID: 31298011 PMCID: PMC8337431 DOI: 10.7507/1002-1892.201812079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/15/2019] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To review the literature reports on research progress of Heme oxygenase 1 (HO-1) modified mesenchymal stem cells (MSCs). METHODS The significance, effects, and related mechanism of HO-1 modification of MSCs were summarized by consulting the related literatures and reports of HO-1 modification of MSCs. RESULTS HO-1 modification of MSCs has important research value. It can effectively enhance the anti-oxidative stress and anti-apoptotic properties of MSCs in complex internal environment after transplantation into vivo. It can also effectively enhance the immune regulation function of MSCs. It can improve the anti-injury, repair, and immune regulation effect of MSCs in various disease models and research fields. CONCLUSION The basic research of HO-1 modified MSCs has made remarkable progress, which is expected to be applied in clinical trials and provide theoretical basis and reference value for stem cell therapy.
Collapse
Affiliation(s)
- Dong Sun
- The First Central Clinical College, Tianjin Medical University, Tianjin, 300192, P.R.China
| | - Hongli Song
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin Key Laboratory of Organ Transplantation, Key Laboratory of Transplantation Medicine, Chinese Academy of Medical Sciences, Tianjin, 300192,
| | - Zhongyang Shen
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin Key Laboratory of Organ Transplantation, Key Laboratory of Transplantation Medicine, Chinese Academy of Medical Sciences, Tianjin, 300192, P.R.China
| |
Collapse
|
150
|
Kinser HE, Pincus Z. MicroRNAs as modulators of longevity and the aging process. Hum Genet 2019; 139:291-308. [PMID: 31297598 DOI: 10.1007/s00439-019-02046-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/05/2019] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are short, non-coding RNAs that post-transcriptionally repress translation or induce mRNA degradation of target transcripts through sequence-specific binding. miRNAs target hundreds of transcripts to regulate diverse biological pathways and processes, including aging. Many microRNAs are differentially expressed during aging, generating interest in their use as aging biomarkers and roles as regulators of the aging process. In the invertebrates Caenorhabditis elegans and Drosophila, a number of miRNAs have been found to both positive and negatively modulate longevity through canonical aging pathways. Recent studies have also shown that miRNAs regulate age-associated processes and pathologies in a diverse array of mammalian tissues, including brain, heart, bone, and muscle. The review will present an overview of these studies, highlighting the role of individual miRNAs as biomarkers of aging and regulators of longevity and tissue-specific aging processes.
Collapse
Affiliation(s)
- Holly E Kinser
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, USA
| | - Zachary Pincus
- Department of Developmental Biology and Department of Genetics, Washington University in St. Louis, St. Louis, USA.
| |
Collapse
|