1
|
Ksouri R, Odabas S, Yar Sağlam AS. Exosome loaded 3D printed magnetic PLA constructs: a candidate for bone tissue engineering. PROGRESS IN ADDITIVE MANUFACTURING 2025; 10:247-260. [DOI: 10.1007/s40964-024-00619-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 04/07/2024] [Indexed: 01/05/2025]
|
2
|
Xu W, Zhang Y, Li L, Pan L, Lu L, Zhi S, Li W. Osteocyte-derived exosomes regulate the DLX2/wnt pathway to alleviate osteoarthritis by mediating cartilage repair. Autoimmunity 2024; 57:2364686. [PMID: 38946534 DOI: 10.1080/08916934.2024.2364686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 06/02/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Chondrocyte viability, apoptosis, and migration are closely related to cartilage injury in osteoarthritis (OA) joints. Exosomes are identified as potential therapeutic agents for OA. OBJECTIVE This study aimed to investigate the role of exosomes derived from osteocytes in OA, particularly focusing on their effects on cartilage repair and molecular mechanisms. METHODS An injury cell model was established by treating chondrocytes with IL-1β. Cartilage repair was evaluated using cell counting kit-8, flow cytometry, scratch test, and Western Blot. Molecular mechanisms were analyzed using quantitative real-time PCR, bioinformatic analysis, and Western Blot. An OA mouse model was established to explore the role of exosomal DLX2 in vivo. RESULTS Osteocyte-released exosomes promoted cell viability and migration, and inhibited apoptosis and extracellular matrix (ECM) deposition. Moreover, exosomes upregulated DLX2 expression, and knockdown of DLX2 activated the Wnt pathway. Additionally, exosomes attenuated OA in mice by transmitting DLX2. CONCLUSION Osteocyte-derived exosomal DLX2 alleviated IL-1β-induced cartilage repair and inactivated the Wnt pathway, thereby alleviating OA progression. The findings suggested that osteocyte-derived exosomes may hold promise as a treatment for OA.
Collapse
Affiliation(s)
- Wenjuan Xu
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Yuanyuan Zhang
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Lijuan Li
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Liyan Pan
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Li Lu
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| | - Shenshen Zhi
- Department of Blood Transfusion, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Wei Li
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Clinical Laboratory, Chongqing, China
| |
Collapse
|
3
|
Mehrvar A, Akbari M, Khosroshahi EM, Nekavand M, Mokhtari K, Baniasadi M, Aghababaian M, Karimi M, Amiri S, Moazen A, Maghsoudloo M, Alimohammadi M, Rahimzadeh P, Farahani N, Vaghar ME, Entezari M, Hashemi M. The impact of exosomes on bone health: A focus on osteoporosis. Pathol Res Pract 2024; 263:155618. [PMID: 39362132 DOI: 10.1016/j.prp.2024.155618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Osteoporosis is a widespread chronic condition. Although standard treatments are generally effective, they are frequently constrained by side effects and the risk of developing drug resistance. A promising area of research is the investigation of extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, which play a crucial role in bone metabolism. Exosomes, in particular, have shown significant potential in both the diagnosis and treatment of osteoporosis. EVs derived from osteoclasts, osteoblasts, mesenchymal stem cells, and other sources can influence bone metabolism, while exosomes from inflammatory and tumor cells may exacerbate bone loss, highlighting their dual role in osteoporosis pathology. This review offers a comprehensive overview of EV biogenesis, composition, and function in osteoporosis, focusing on their diagnostic and therapeutic potential. We examine the roles of various types of EVs and their cargo-proteins, RNAs, and lipids-in bone metabolism. Additionally, we explore the emerging applications of EVs as biomarkers and therapeutic agents, emphasizing the need for further research to address current challenges and enhance EV-based strategies for managing osteoporosis.
Collapse
Affiliation(s)
- Amir Mehrvar
- Assistant Professor, Department of Orthopedics, Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadarian Akbari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrandokht Nekavand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Midwifery, Faculty of nursing and midwifery, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mojtaba Baniasadi
- Department of Orthopedic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran; MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Aghababaian
- Department of Orthopedic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansour Karimi
- MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- MD, Assistant Professor of Orthopaedic Surgery, Shohadaye Haftom-e-Tir Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Moazen
- Department of Orthopedics, Bone and Joint Reconstruction Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohammad Eslami Vaghar
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of gynecology, Faculty of Medicine, Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
4
|
Zhang X, Liang Y, Luo D, Li P, Chen Y, Fu X, Yue Y, Hou R, Liu J, Wang X. Advantages and disadvantages of various hydrogel scaffold types: A research to improve the clinical conversion rate of loaded MSCs-Exos hydrogel scaffolds. Biomed Pharmacother 2024; 179:117386. [PMID: 39241570 DOI: 10.1016/j.biopha.2024.117386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024] Open
Abstract
Mesenchymal stem cell-derived exosomes(MSCs-Exos) offer promising therapeutic potential for a wide range of tissues and organs such as bone/cartilage, nerves, skin, fat, and endocrine organs. In comparison to the application of mesenchymal stem cells (MSCs), MSCs-Exos address critical challenges related to rejection reactions and ethical concerns, positioning themselves as a promising cell-free therapy. As exosomes are extracellular vesicles, their effective delivery necessitates the use of carriers. Consequently, the selection of hydrogel materials as scaffolds for exosome delivery has become a focal point of contemporary research. The diversity of hydrogel scaffolds, which can take various forms such as injectable types, dressings, microneedles, and capsules, leads to differing choices among researchers for treating diseases within the same domain. This variability in hydrogel materials poses challenges for the translation of findings into clinical practice. The review highlights the potential of hydrogel-loaded exosomes in different fields and introduces the advantages and disadvantages of different forms of hydrogel applications. It aims to provide a multifunctional and highly recognized hydrogel scaffold option for tissue regeneration at specific sites, improve clinical translation efficiency, and benefit the majority of patients.
Collapse
Affiliation(s)
- Xinyao Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Yi Liang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Dongmei Luo
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Peiwen Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Yurou Chen
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Xinyu Fu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Yingge Yue
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Ruxia Hou
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Junyu Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China.
| | - Xiangyu Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China.
| |
Collapse
|
5
|
Wu Y, Sun B, Tang Y, Shen A, Lin Y, Zhao X, Li J, Monteiro MJ, Gu W. Bone targeted nano-drug and nano-delivery. Bone Res 2024; 12:51. [PMID: 39231955 PMCID: PMC11375042 DOI: 10.1038/s41413-024-00356-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 09/06/2024] Open
Abstract
There are currently no targeted delivery systems to satisfactorily treat bone-related disorders. Many clinical drugs consisting of small organic molecules have a short circulation half-life and do not effectively reach the diseased tissue site. This coupled with repeatedly high dose usage that leads to severe side effects. With the advance in nanotechnology, drugs contained within a nano-delivery device or drugs aggregated into nanoparticles (nano-drugs) have shown promises in targeted drug delivery. The ability to design nanoparticles to target bone has attracted many researchers to develop new systems for treating bone related diseases and even repurposing current drug therapies. In this review, we shall summarise the latest progress in this area and present a perspective for future development in the field. We will focus on calcium-based nanoparticle systems that modulate calcium metabolism and consequently, the bone microenvironment to inhibit disease progression (including cancer). We shall also review the bone affinity drug family, bisphosphonates, as both a nano-drug and nano-delivery system for bone targeted therapy. The ability to target and release the drug in a controlled manner at the disease site represents a promising safe therapy to treat bone diseases in the future.
Collapse
Affiliation(s)
- Yilun Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Bing Sun
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Ying Tang
- Science and Technology Innovation Centre, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Aining Shen
- Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Yanlin Lin
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Xiaohui Zhao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jingui Li
- School of Veterinary Medicine, Jiangsu Co-innovation Centre for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Michael J Monteiro
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
6
|
Zhu Y, Li Y, Cao Z, Xue J, Wang X, Hu T, Han B, Guo Y. Mechanically strained osteocyte-derived exosomes contained miR-3110-5p and miR-3058-3p and promoted osteoblastic differentiation. Biomed Eng Online 2024; 23:44. [PMID: 38705993 PMCID: PMC11070085 DOI: 10.1186/s12938-024-01237-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND Osteocytes are critical mechanosensory cells in bone, and mechanically stimulated osteocytes produce exosomes that can induce osteogenesis. MicroRNAs (miRNAs) are important constituents of exosomes, and some miRNAs in osteocytes regulate osteogenic differentiation; previous studies have indicated that some differentially expressed miRNAs in mechanically strained osteocytes likely influence osteoblastic differentiation. Therefore, screening and selection of miRNAs that regulate osteogenic differentiation in exosomes of mechanically stimulated osteocytes are important. RESULTS A mechanical tensile strain of 2500 με at 0.5 Hz 1 h per day for 3 days, elevated prostaglandin E2 (PGE2) and insulin-like growth factor-1 (IGF-1) levels and nitric oxide synthase (NOS) activity of MLO-Y4 osteocytes, and promoted osteogenic differentiation of MC3T3-E1 osteoblasts. Fourteen miRNAs differentially expressed only in MLO-Y4 osteocytes which were stimulated with mechanical tensile strain, were screened, and the miRNAs related to osteogenesis were identified. Four differentially expressed miRNAs (miR-1930-3p, miR-3110-5p, miR-3090-3p, and miR-3058-3p) were found only in mechanically strained osteocytes, and the four miRNAs, eight targeted mRNAs which were differentially expressed only in mechanically strained osteoblasts, were also identified. In addition, the mechanically strained osteocyte-derived exosomes promoted the osteoblastic differentiation of MC3T3-E1 cells in vitro, the exosomes were internalized by osteoblasts, and the up-regulated miR-3110-5p and miR-3058-3p in mechanically strained osteocytes, were both increased in the exosomes, which was verified via reverse transcription quantitative polymerase chain reaction (RT-qPCR). CONCLUSIONS In osteocytes, a mechanical tensile strain of 2500 με at 0.5 Hz induced the fourteen differentially expressed miRNAs which probably were in exosomes of osteocytes and involved in osteogenesis. The mechanically strained osteocyte-derived exosomes which contained increased miR-3110-5p and miR-3058-3p (two of the 14 miRNAs), promoted osteoblastic differentiation.
Collapse
Affiliation(s)
- Yingwen Zhu
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Yanan Li
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Zhen Cao
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Jindong Xue
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Xiaoyan Wang
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Tingting Hu
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Biao Han
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China.
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China.
| | - Yong Guo
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China.
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China.
| |
Collapse
|
7
|
Wang P, Shao W, Li Z, Wang B, Lv X, Huang Y, Feng Y. Non-bone-derived exosomes: a new perspective on regulators of bone homeostasis. Cell Commun Signal 2024; 22:70. [PMID: 38273356 PMCID: PMC10811851 DOI: 10.1186/s12964-023-01431-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/09/2023] [Indexed: 01/27/2024] Open
Abstract
Accumulating evidence indicates that exosomes help to regulate bone homeostasis. The roles of bone-derived exosomes have been well-described; however, recent studies have shown that some non-bone-derived exosomes have better bone targeting ability than bone-derived exosomes and that their performance as a drug delivery vehicle for regulating bone homeostasis may be better than that of bone-derived exosomes, and the sources of non-bone-derived exosomes are more extensive and can thus be better for clinical needs. Here, we sort non-bone-derived exosomes and describe their composition and biogenesis. Their roles and specific mechanisms in bone homeostasis and bone-related diseases are also discussed. Furthermore, we reveal obstacles to current research and future challenges in the practical application of exosomes, and we provide potential strategies for more effective application of exosomes for the regulation of bone homeostasis and the treatment of bone-related diseases. Video Abstract.
Collapse
Affiliation(s)
- Ping Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenkai Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Wang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiyao Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
8
|
Bratengeier C, Johansson L, Liszka A, Bakker AD, Hallbeck M, Fahlgren A. Mechanical loading intensities affect the release of extracellular vesicles from mouse bone marrow-derived hematopoietic progenitor cells and change their osteoclast-modulating effect. FASEB J 2024; 38:e23323. [PMID: 38015031 DOI: 10.1096/fj.202301520r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023]
Abstract
Low-intensity loading maintains or increases bone mass, whereas lack of mechanical loading and high-intensity loading decreases bone mass, possibly via the release of extracellular vesicles by mechanosensitive bone cells. How different loading intensities alter the biological effect of these vesicles is not fully understood. Dynamic fluid shear stress at low intensity (0.7 ± 0.3 Pa, 5 Hz) or high intensity (2.9 ± 0.2 Pa, 1 Hz) was used on mouse hematopoietic progenitor cells for 2 min in the presence or absence of chemical compounds that inhibit release or biogenesis of extracellular vesicles. We used a Receptor activator of nuclear factor kappa-Β ligand-induced osteoclastogenesis assay to evaluate the biological effect of different fractions of extracellular vesicles obtained through centrifugation of medium from hematopoietic stem cells. Osteoclast formation was reduced by microvesicles (10 000× g) obtained after low-intensity loading and induced by exosomes (100 000× g) obtained after high-intensity loading. These osteoclast-modulating effects could be diminished or eliminated by depletion of extracellular vesicles from the conditioned medium, inhibition of general extracellular vesicle release, inhibition of microvesicle biogenesis (low intensity), inhibition of ESCRT-independent exosome biogenesis (high intensity), as well as by inhibition of dynamin-dependent vesicle uptake in osteoclast progenitor cells. Taken together, the intensity of mechanical loading affects the release of extracellular vesicles and change their osteoclast-modulating effect.
Collapse
Affiliation(s)
- C Bratengeier
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - L Johansson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Pathology, Linköping University, Linköping, Sweden
| | - A Liszka
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - A D Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - M Hallbeck
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Pathology, Linköping University, Linköping, Sweden
| | - A Fahlgren
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
9
|
Zhu Q, Tang Y, Zhou T, Yang L, Zhang G, Meng Y, Zhang H, Gao J, Wang C, Su YX, Ye J. Exosomes derived from mesenchymal stromal cells promote bone regeneration by delivering miR-182-5p-inhibitor. Pharmacol Res 2023; 192:106798. [PMID: 37211240 DOI: 10.1016/j.phrs.2023.106798] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 05/23/2023]
Abstract
Exosomes, small extracellular vesicles that function as a key regulator of cell-to-cell communication, are emerging as a promising candidate for bone regeneration. Here, we aimed to investigate the effect of exosomes from pre-differentiated human alveolar bone-derived bone marrow mesenchymal stromal cells (AB-BMSCs) carrying specific microRNAs on bone regeneration. Exosomes secreted from AB-BMSCs pre-differentiated for 0 and 7 days were cocultured with BMSCs in vitro to investigate their effect on the differentiation of the BMSCs. MiRNAs from AB-BMSCs at different stages of osteogenic differentiation were analyzed. BMSCs seeded on poly-L-lactic acid(PLLA) scaffolds were treated with miRNA antagonist-decorated exosomes to verify their effect on new bone regeneration. Exosomes pre-differentiated for 7 days effectively promoted the differentiation of BMSCs. Bioinformatic analysis revealed that miRNAs within the exosomes were differentially expressed, including the upregulation of osteogenic miRNAs (miR-3182, miR-1468) and downregulation of anti-osteogenic miRNAs (miR-182-5p, miR-335-3p, miR-382-5p), causing activation of the PI3K/Akt signaling pathway. The treatment of BMSC-seeded scaffolds with anti-miR-182-5p decorated exosomes demonstrated enhanced osteogenic differentiation and efficient formation of new bone. In conclusion, Osteogenic exosomes secreted from pre-differentiated AB-BMSCs were identified and the gene modification of exosomes provides great potential as a bone regeneration strategy. DATA AVAILABILITY STATEMENT: Data generated or analyzed in this paper partly are available in the GEO public data repository(http://www.ncbi.nlm.nih.gov/geo).
Collapse
Affiliation(s)
- Qinghai Zhu
- Jiangsu Key Laboratory of Oral Disease, & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuting Tang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tian Zhou
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Li Yang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Gao Zhang
- Division of Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, 999077, China
| | - Ying Meng
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Huixin Zhang
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing 211166, China
| | - Jun Gao
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing 211166, China
| | - Chenxing Wang
- Jiangsu Key Laboratory of Oral Disease, & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yu-Xiong Su
- Discipline of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, 999077, SAR, China.
| | - Jinhai Ye
- Jiangsu Key Laboratory of Oral Disease, & Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
10
|
Panero AJ, Everts PA, Nakagawa H, Sussman W, Qin X. Basic Science of Allograft Orthobiologics. Phys Med Rehabil Clin N Am 2023; 34:49-61. [DOI: 10.1016/j.pmr.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
11
|
Cheung KCP, Jiao M, Xingxuan C, Wei J. Extracellular vesicles derived from host and gut microbiota as promising nanocarriers for targeted therapy in osteoporosis and osteoarthritis. Front Pharmacol 2023; 13:1051134. [PMID: 36686680 PMCID: PMC9859449 DOI: 10.3389/fphar.2022.1051134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/21/2022] [Indexed: 01/08/2023] Open
Abstract
Osteoporosis (OP), a systemic bone disease that causes structural bone loss and bone mass loss, is often associated with fragility fractures. Extracellular vesicles (EVs) generated by mammalian and gut bacteria have recently been identified as important mediators in the intercellular signaling pathway that may play a crucial role in microbiota-host communication. EVs are tiny membrane-bound vesicles, which range in size from 20 to 400 nm. They carry a variety of biologically active substances across intra- and intercellular space. These EVs have developed as a promising research area for the treatment of OP because of their nanosized architecture, enhanced biocompatibility, reduced toxicity, drug loading capacity, ease of customization, and industrialization. This review describes the latest development of EVs derived from mammals and bacteria, including their internalization, isolation, biogenesis, classifications, topologies, and compositions. Additionally, breakthroughs in chemical sciences and the distinctive biological features of bacterial extracellular vesicles (BEVs) allow for the customization of modified BEVs for the therapy of OP. In conclusion, we give a thorough and in-depth summary of the main difficulties and potential future of EVs in the treatment of OP, as well as highlight innovative uses and choices for the treatment of osteoarthritis (OA).
Collapse
Affiliation(s)
- Kenneth Chat Pan Cheung
- Hong Kong Traditional Chinese Medicine Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ma Jiao
- Hong Kong Traditional Chinese Medicine Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Chen Xingxuan
- Hong Kong Traditional Chinese Medicine Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Jia Wei
- Hong Kong Traditional Chinese Medicine Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
12
|
de Oliveira MC, Heredia JE, da Silva FRF, Macari S. Extracellular Vesicles in Bone Remodeling and Osteoporosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:155-168. [PMID: 37603279 DOI: 10.1007/978-981-99-1443-2_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Osteoporosis is a systemic disorder characterized by bone mass loss, leading to fractures due to weak and brittle bones. The bone tissue deterioration process is related to an impairment of bone remodeling orchestrated mainly by resident bone cells, including osteoblasts, osteoclasts, osteocytes, and their progenitors. Extracellular vesicles (EVs) are nanoparticles emerging as regulatory molecules and potential biomarkers for bone loss. Although the progress in studies relating to EVs and bone loss has increased in the last years, research on bone cells, animal models, and mainly patients is still limited. Here, we aim to review the recent advances in this field, summarizing the effect of EV components such as proteins and miRNAs in regulating bone remodeling and, consequently, osteoporosis progress and treatment. Also, we discuss the potential application of EVs in clinical practice as a biomarker and bone loss therapy, demonstrating that this rising field still needs to be further explored.
Collapse
Affiliation(s)
- Marina Chaves de Oliveira
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Joyce Elisa Heredia
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Soraia Macari
- Department of Restorative Dentistry, Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
13
|
Ren J, Yu R, Xue J, Tang Y, Su S, Liao C, Guo Q, Guo W, Zheng J. How Do Extracellular Vesicles Play a Key Role in the Maintenance of Bone Homeostasis and Regeneration? A Comprehensive Review of Literature. Int J Nanomedicine 2022; 17:5375-5389. [PMID: 36419718 PMCID: PMC9677931 DOI: 10.2147/ijn.s377598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/31/2022] [Indexed: 11/26/2023] Open
Abstract
The maintenance of bone homeostasis includes both bone resorption by osteoclasts and bone formation by osteoblasts. These two processes are in dynamic balance to maintain a constant amount of bone for accomplishing its critical functions in daily life. Multiple cell type communications are involved in these two complex and continuous processes. In recent decades, an increasing number of studies have shown that osteogenic and osteoclastic extracellular vesicles play crucial roles in regulating bone homeostasis through paracrine, autosecretory and endocrine signaling. Elucidating the functional roles of extracellular vesicles in the maintenance of bone homeostasis may contribute to the design of new strategies for bone regeneration. Hence, we review the recent understandings of the classification, production process, extraction methods, structure, contents, functions and applications of extracellular vesicles in bone homeostasis. We highlight the contents of various bone-derived extracellular vesicles and their interactions with different cells in the bone microenvironment during bone homeostasis. We also summarize the recent advances in EV-loaded biomaterial scaffolds for bone regeneration and repair.
Collapse
Affiliation(s)
- Junxian Ren
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People’s Republic of China
| | - Rongcheng Yu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People’s Republic of China
| | - Jingyan Xue
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People’s Republic of China
| | - Yiqi Tang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People’s Republic of China
| | - Sihui Su
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People’s Republic of China
| | - Chenxi Liao
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People’s Republic of China
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, People’s Republic of China
| | - Weimin Guo
- Department of Orthopedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510055, People’s Republic of China
| | - Jinxuan Zheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People’s Republic of China
| |
Collapse
|
14
|
Li Z, Fang F, Long Y, Zhao Q, Wang X, Ye Z, Meng T, Gu X, Xiang W, Xiong C, Li H. The balance between NANOG and SOX17 mediated by TET proteins regulates specification of human primordial germ cell fate. Cell Biosci 2022; 12:181. [PMID: 36333732 PMCID: PMC9636699 DOI: 10.1186/s13578-022-00917-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Background Human primordial germ cells (hPGCs) initiate from the early post-implantation embryo at week 2–3 and undergo epigenetic reprogramming during development. However, the regulatory mechanism of DNA methylation during hPGC specification is still largely unknown due to the difficulties in analyzing early human embryos. Using an in vitro model of hPGC induction, we found a novel function of TET proteins and NANOG in the hPGC specification which was different from that discovered in mice. Methods Using the CRISPR–Cas9 system, we generated a set of TET1, TET2 and TET3 knockout H1 human embryonic stem cell (hESC) lines bearing a BLIMP1-2A-mKate2 reporter. We determined the global mRNA transcription and DNA methylation profiles of pluripotent cells and induced hPGC-like cells (hPGCLCs) by RNA-seq and whole-genome bisulfite sequencing (WGBS) to reveal the involved signaling pathways after TET proteins knockout. ChIP-qPCR was performed to verify the binding of TET and NANOG proteins in the SOX17 promoter. Real-time quantitative PCR, western blot and immunofluorescence were performed to measure gene expression at mRNA and protein levels. The efficiency of hPGC induction was evaluated by FACS. Results In humans, TET1, TET2 and TET3 triple-knockout (TKO) human embryonic stem cells (hESCs) impaired the NODAL signaling pathway and impeded hPGC specification in vitro, while the hyperactivated NODAL signaling pathway led to gastrulation failure when Tet proteins were inactivated in mouse. Specifically, TET proteins stimulated SOX17 through the NODAL signaling pathway and directly regulates NANOG expression at the onset of hPGCLCs induction. Notably, NANOG could bind to SOX17 promoter to regulate its expression in hPGCLCs specification. Furthermore, in TKO hESCs, DNMT3B-mediated hypermethylation of the NODAL signaling-related genes and NANOG/SOX17 promoters repressed their activation and inhibited hPGCLC induction. Knockout of DNMT3B in TKO hESCs partially restored NODAL signaling and NANOG/SOX17 expression, and rescued hPGCLC induction. Conclusion Our results show that TETs-mediated oxidation of 5-methylcytosine modulates the NODAL signaling pathway and its downstream genes, NANOG and SOX17, by promoting demethylation in opposition to DNMT3B-mediated methylation, suggesting that the epigenetic balance of DNA methylation and demethylation in key genes plays a fundamental role in early hPGC specification. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00917-0.
Collapse
|
15
|
Yao Y, Jiang Y, Song J, Wang R, Li Z, Yang L, Wu W, Zhang L, Peng Q. Exosomes as Potential Functional Nanomaterials for Tissue Engineering. Adv Healthc Mater 2022:e2201989. [PMID: 36253093 DOI: 10.1002/adhm.202201989] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/14/2022] [Indexed: 11/10/2022]
Abstract
Exosomes are cell-derived extracellular vesicles of 40-160 nm diameter, which carry numerous biomolecules and transmit information between cells. They are used as functional nanomaterials with great potential in biomedical areas, such as active agents and delivery systems for advanced drug delivery and disease therapy. In recent years, potential applications of exosomes in tissue engineering have attracted significant attention, and some critical progress has been made. This review gives a complete picture of exosomes and their applications in the regeneration of various tissues, such as the central nervous systems, kidney, bone, cartilage, heart, and endodontium. Approaches employed for modifying exosomes to equip them with excellent targeting capacity are summarized. Furthermore, current concerns and future outlook of exosomes in tissue engineering are discussed.
Collapse
Affiliation(s)
- Yang Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Yuhuan Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Jialu Song
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Ruojing Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Zhaoping Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Lei Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Weimin Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Luyue Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| |
Collapse
|
16
|
Miller G, Menzel A, Ankerst DP. Association between short-term exposure to air pollution and COVID-19 mortality in all German districts: the importance of confounders. ENVIRONMENTAL SCIENCES EUROPE 2022; 34:79. [PMID: 36062033 PMCID: PMC9418649 DOI: 10.1186/s12302-022-00657-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The focus of many studies is to estimate the effect of risk factors on outcomes, yet results may be dependent on the choice of other risk factors or potential confounders to include in a statistical model. For complex and unexplored systems, such as the COVID-19 spreading process, where a priori knowledge of potential confounders is lacking, data-driven empirical variable selection methods may be primarily utilized. Published studies often lack a sensitivity analysis as to how results depend on the choice of confounders in the model. This study showed variability in associations of short-term air pollution with COVID-19 mortality in Germany under multiple approaches accounting for confounders in statistical models. METHODS Associations between air pollution variables PM2.5, PM10, CO, NO, NO2, and O3 and cumulative COVID-19 deaths in 400 German districts were assessed via negative binomial models for two time periods, March 2020-February 2021 and March 2021-February 2022. Prevalent methods for adjustment of confounders were identified after a literature search, including change-in-estimate and information criteria approaches. The methods were compared to assess the impact on the association estimates of air pollution and COVID-19 mortality considering 37 potential confounders. RESULTS Univariate analyses showed significant negative associations with COVID-19 mortality for CO, NO, and NO2, and positive associations, at least for the first time period, for O3 and PM2.5. However, these associations became non-significant when other risk factors were accounted for in the model, in particular after adjustment for mobility, political orientation, and age. Model estimates from most selection methods were similar to models including all risk factors. CONCLUSION Results highlight the importance of adequately accounting for high-impact confounders when analyzing associations of air pollution with COVID-19 and show that it can be of help to compare multiple selection approaches. This study showed how model selection processes can be performed using different methods in the context of high-dimensional and correlated covariates, when important confounders are not known a priori. Apparent associations between air pollution and COVID-19 mortality failed to reach significance when leading selection methods were used. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1186/s12302-022-00657-5.
Collapse
Affiliation(s)
- Gregor Miller
- Department of Mathematics, Technical University of Munich, Boltzmannstrasse 3, Garching, Germany
| | - Annette Menzel
- Department of Life Science Systems, Technical University of Munich, Freising, Germany
| | - Donna P. Ankerst
- Department of Mathematics, Technical University of Munich, Boltzmannstrasse 3, Garching, Germany
- Department of Life Science Systems, Technical University of Munich, Freising, Germany
| |
Collapse
|
17
|
Yuan W, Song C. Crosstalk between bone and other organs. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:331-348. [PMID: 37724328 PMCID: PMC10471111 DOI: 10.1515/mr-2022-0018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/06/2022] [Indexed: 09/20/2023]
Abstract
Bone has long been considered as a silent organ that provides a reservoir of calcium and phosphorus, traditionally. Recently, further study of bone has revealed additional functions as an endocrine organ connecting systemic organs of the whole body. Communication between bone and other organs participates in most physiological and pathological events and is responsible for the maintenance of homeostasis. Here, we present an overview of the crosstalk between bone and other organs. Furthermore, we describe the factors mediating the crosstalk and review the mechanisms in the development of potential associated diseases. These connections shed new light on the pathogenesis of systemic diseases and provide novel potential targets for the treatment of systemic diseases.
Collapse
Affiliation(s)
- Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Chunli Song
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
18
|
Kang M, Huang CC, Gajendrareddy P, Lu Y, Shirazi S, Ravindran S, Cooper LF. Extracellular Vesicles From TNFα Preconditioned MSCs: Effects on Immunomodulation and Bone Regeneration. Front Immunol 2022; 13:878194. [PMID: 35585987 PMCID: PMC9108364 DOI: 10.3389/fimmu.2022.878194] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/07/2022] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells show remarkable versatility and respond to extracellular and micro environmental cues by altering their phenotype and behavior. In this regard, the MSC’s immunomodulatory properties in tissue repair are well documented. The paracrine effects of MSCs in immunomodulation are, in part, attributable to their secreted extracellular vesicles (EVs). When MSCs migrate to the wound bed, they are exposed to a myriad of inflammatory signals. To understand their response to an inflammatory environment from an EV perspective, we sought to evaluate the effects of the inflammatory cytokine TNFα on MSC EV mediated immunomodulation. Our results indicate that while the physical characteristics of the EVs remain unchanged, the TNFα preconditioned MSC EVs possess enhanced immunomodulatory properties. In vitro experiments using polarized (M1 and M2) primary mouse macrophages indicated that the preconditioned MSC EVs suppressed pro-inflammatory (M1) markers such as IL-1β and iNOS and elevated reparatory (M2) markers such as Arg1 and CD206. When evaluated in vivo in a rat calvarial defect model, the TNFα preconditioned MSC EVs reduced inflammation at 1-, 3- and 7-days post wounding resulting in the subsequent enhanced bone formation at 4- and 8-weeks post wounding possibly by modulation of oncostatin M (OSM) expression. An analysis of EV miRNA composition revealed significant changes to anti-inflammatory miRNAs in the preconditioned MSC EVs hinting at a possible role for EV derived miRNA in the enhanced immunomodulatory activity. Overall, these results indicate that MSC exposure to inflammatory signals influence the MSC EV’s immunomodulatory function in the context of tissue repair. The specific function of TNFα preconditioned MSC EV miRNAs in immunomodulatory control of bone regeneration merits further investigation.
Collapse
Affiliation(s)
- Miya Kang
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Chun-Chieh Huang
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Praveen Gajendrareddy
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Yu Lu
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Sajjad Shirazi
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Sriram Ravindran
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
- *Correspondence: Lyndon F. Cooper, ; Sriram Ravindran,
| | - Lyndon F. Cooper
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
- *Correspondence: Lyndon F. Cooper, ; Sriram Ravindran,
| |
Collapse
|
19
|
Bone Cell Exosomes and Emerging Strategies in Bone Engineering. Biomedicines 2022; 10:biomedicines10040767. [PMID: 35453517 PMCID: PMC9033129 DOI: 10.3390/biomedicines10040767] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 01/27/2023] Open
Abstract
Bone tissue remodeling is a highly regulated process balancing bone formation and resorption through complex cellular crosstalk between resident bone and microenvironment cells. This cellular communication is mediated by direct cell and cell–matrix contact, autocrine, endocrine, and paracrine receptor mediated mechanisms such as local soluble signaling molecules and extracellular vesicles including nanometer sized exosomes. An impairment in this balanced process leads to development of pathological conditions. Bone tissue engineering is an emerging interdisciplinary field with potential to address bone defects and disorders by synthesizing three-dimensional bone substitutes embedded with cells for clinical implantation. However, current cell-based therapeutic approaches have faced hurdles due to safety and ethical concerns, challenging their clinical translation. Recent studies on exosome-regulated bone homeostasis and regeneration have gained interest as prospective cell free therapy in conjugation with tissue engineered bone grafts. However, exosome research is still in its nascent stages of bone tissue engineering. In this review, we specifically describe the role of exosomes secreted by cells within bone microenvironment such as osteoblasts, osteocytes, osteoclasts, mesenchymal stem cell cells, immune cells, endothelial cells, and even tumor cells during bone homeostasis and crosstalk. We also review exosome-based osteoinductive functionalization strategies for various bone-based biomaterials such as ceramics, polymers, and metals in bone tissue engineering. We further highlight biomaterials as carrier agents for exosome delivery to bone defect sites and, finally, the influence of various biomaterials in modulation of cell exosome secretome.
Collapse
|
20
|
Huber J, Griffin MF, Longaker MT, Quarto N. Exosomes: A Tool for Bone Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:101-113. [PMID: 33297857 PMCID: PMC8892957 DOI: 10.1089/ten.teb.2020.0246] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Mesenchymal stem cells (MSCs) have been repeatedly shown to be a valuable source for cell-based therapy in regenerative medicine, including bony tissue repair. However, engraftment at the injury site is poor. Recently, it has been suggested that MSCs and other cells act through a paracrine signaling mechanism. Exosomes are nanostructures that have been implicated in this process. They carry DNA, RNA, proteins, and lipids and play an important role in cell-to-cell communication directly modulating their target cell at a transcriptional level. In a bone microenvironment, they have been shown to increase osteogenesis and osteogenic differentiation in vivo and in vitro. In the following review, we will discuss the most advanced and significant knowledge of biological functions of exosomes in bone regeneration and their clinical applications in osseous diseases. Impact statement Mesenchymal stem cells have been shown to be a promising tool in bone tissue engineering. Recently, it has been suggested that they secrete exosomes containing messenger RNA, proteins, and lipids, thus acting through paracrine signaling mechanisms. Considering that exosomes are nonteratogenic and have low immunogenic potential, they could potentially replace stem-cell based therapy and thus eradicate the risk of neoplastic transformation associated with cell transplantations in bone regeneration.
Collapse
Affiliation(s)
- Julika Huber
- Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA.,Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany.,Address correspondence to: Julika Huber, MD, Dr. med, Hagey Laboratory for Pediatric Regenerative Medicine, School of Medicine, Stanford University, 257 Campus Drive, Stanford, CA 94305-5148, USA
| | - Michelle F. Griffin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA.,Stanford Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Natalina Quarto
- Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California, USA.,Dipartimento di Scienze Biomediche Avanzate, Universita’ degli Studi di Napoli Federico II, Napoli, Italy.,Address correspondence to: Natalina Quarto, PhD, Hagey Laboratory for Pediatric Regenerative Medicine, School of Medicine, Stanford University, 257 Campus Drive, Stanford, CA 94305-5148, USA
| |
Collapse
|
21
|
Deng J, Zong Z, Su Z, Chen H, Huang J, Niu Y, Zhong H, Wei B. Recent Advances in Pharmacological Intervention of Osteoarthritis: A Biological Aspect. Front Pharmacol 2021; 12:772678. [PMID: 34887766 PMCID: PMC8649959 DOI: 10.3389/fphar.2021.772678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/04/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease in the musculoskeletal system with a relatively high incidence and disability rate in the elderly. It is characterized by the degradation of articular cartilage, inflammation of the synovial membrane, and abnormal structure in the periarticular and subchondral bones. Although progress has been made in uncovering the molecular mechanism, the etiology of OA is still complicated and unclear. Nevertheless, there is no treatment method that can effectively prevent or reverse the deterioration of cartilage and bone structure. In recent years, in the field of pharmacology, research focus has shifted to disease prevention and early treatment rather than disease modification in OA. Biologic agents become more and more attractive as their direct or indirect intervention effects on the initiation or development of OA. In this review, we will discuss a wide spectrum of biologic agents ranging from DNA, noncoding RNA, exosome, platelet-rich plasma (PRP), to protein. We searched for key words such as OA, DNA, gene, RNA, exosome, PRP, protein, and so on. From the pharmacological aspect, stem cell therapy is a very special technique, which is not included in this review. The literatures ranging from January 2016 to August 2021 were included and summarized. In this review, we aim to help readers have a complete and precise understanding of the current pharmacological research progress in the intervention of OA from the biological aspect and provide an indication for the future translational studies.
Collapse
Affiliation(s)
- Jinxia Deng
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Zhixian Zong
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Zhanpeng Su
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Haicong Chen
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Jianping Huang
- College of Dentistry, Yonsei University, Seoul, South Korea.,Department of Stomatology, Guangdong Medical University, Zhanjiang, China
| | - Yanru Niu
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Huan Zhong
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Bo Wei
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
22
|
Liu Y, Cai G, Chen P, Jiang T, Xia Z. UBE2E3 regulates cellular senescence and osteogenic differentiation of BMSCs during aging. PeerJ 2021; 9:e12253. [PMID: 34820159 PMCID: PMC8606162 DOI: 10.7717/peerj.12253] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022] Open
Abstract
Background Osteoporosis has gradually become a public health problem in the world. However, the exact molecular mechanism of osteoporosis still remains unclear. Senescence and osteogenic differentiation inhibition of bone marrow mesenchymal stem cells (BMSCs ) are supposed to play an important part in osteoporosis. Methods We used two gene expression profiles (GSE35956 and GSE35958) associated with osteoporosis and selected the promising gene Ubiquitin-conjugating enzyme E2 E3 (UBE2E3). We then verified its function and mechanism by in vitro experiments. Results UBE2E3 was highly expressed in the bone marrow and positively associated with osteogenesis related genes. Besides, UBE2E3 expression reduced in old BMSCs compared with that in young BMSCs. In in vitro experiments, knockdown of UBE2E3 accelerated cellular senescence and inhibited osteogenic differentiation of young BMSCs. On the other hand, overexpression of UBE2E3 attenuated cellular senescence as well as enhanced osteogenic differentiation of old BMSCs. Mechanistically, UBE2E3 might regulate the nuclear factor erythroid 2-related factor (Nrf2) and control its function, thus affecting the senescence and osteogenic differentiation of BMSCs. Conclusion UBE2E3 may be potentially involved in the pathogenesis of osteoporosis by regulating cellular senescence and osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Yalin Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Guangping Cai
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Peng Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China.,Department of Orthopedic, Xiangya Hospital of Central South University, Changsha, China
| | - Tiejian Jiang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Zhuying Xia
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
23
|
Zhao L, Zhang K, He H, Yang Y, Li W, Liu T, Li J. The Relationship Between Mesenchymal Stem Cells and Tumor Dormancy. Front Cell Dev Biol 2021; 9:731393. [PMID: 34712663 PMCID: PMC8545891 DOI: 10.3389/fcell.2021.731393] [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: 06/27/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor dormancy, a state of tumor, is clinically undetectable and the outgrowth of dormant tumor cells into overt metastases is responsible for cancer-associated deaths. However, the dormancy-related molecular mechanism has not been clearly described. Some researchers have proposed that cancer stem cells (CSCs) and disseminated tumor cells (DTCs) can be seen as progenitor cells of tumor dormancy, both of which can remain dormant in a non-permissive soil/niche. Nowadays, research interest in the cancer biology field is skyrocketing as mesenchymal stem cells (MSCs) are capable of regulating tumor dormancy, which will provide a unique therapeutic window to cure cancer. Although the influence of MSCs on tumor dormancy has been investigated in previous studies, there is no thorough review on the relationship between MSCs and tumor dormancy. In this paper, the root of tumor dormancy is analyzed and dormancy-related molecular mechanisms are summarized. With an emphasis on the role of the MSCs during tumor dormancy, new therapeutic strategies to prevent metastatic disease are proposed, whose clinical application potentials are discussed, and some challenges and prospects of the studies of tumor dormancy are also described.
Collapse
Affiliation(s)
- Linxian Zhao
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Kai Zhang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Hongyu He
- Operating Theater and Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Yongping Yang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Wei Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Tongjun Liu
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
24
|
New insights into exosome mediated tumor-immune escape: Clinical perspectives and therapeutic strategies. Biochim Biophys Acta Rev Cancer 2021; 1876:188624. [PMID: 34487817 DOI: 10.1016/j.bbcan.2021.188624] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
Recent advances in extracellular vesicle biology have uncovered a substantial role in maintaining cell homeostasis in health and disease conditions by mediating intercellular communication, thus catching the scientific community's attention worldwide. Extracellular microvesicles, some called exosomes, functionally transfer biomolecules such as proteins and non-coding RNAs from one cell to another, influencing the local environment's biology. Although numerous advancements have been made in treating cancer patients with immune therapy, controlling the disease remains a challenge in the clinic due to tumor-driven interference with the immune response and inability of immune cells to clear cancer cells from the body. The present review article discusses the recent findings and knowledge gaps related to the role of exosomes derived from tumors and the tumor microenvironment cells in tumor escape from immunosurveillance. Further, we highlight examples where exosomal non-coding RNAs influence immune cells' response within the tumor microenvironment and favor tumor growth and progression. Therefore, exosomes can be used as a therapeutic target for the treatment of human cancers.
Collapse
|
25
|
Role of Exosomal MicroRNAs and Their Crosstalk with Oxidative Stress in the Pathogenesis of Osteoporosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6301433. [PMID: 34336108 PMCID: PMC8315851 DOI: 10.1155/2021/6301433] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/24/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Osteoporosis (OP) is an aging-related disease involving permanent bone tissue atrophy. Most patients with OP show high levels of oxidative stress (OS), which destroys the microstructure of bone tissue and promotes disease progression. Exosomes (exos) help in the delivery of microRNAs (miRNAs) and allow intercellular communication. In OP, exosomal miRNAs modulate several physiological processes, including the OS response. In the present review, we aim to describe how exosomal miRNAs and OS contribute to OP. We first summarize the relationship of OS with OP and then detail the features of exos along with the functions of exo-related miRNAs. Further, we explore the interplay between exosomal miRNAs and OS in OP and summarize the functional role of exos in OP. Finally, we identify the advantages of exo-based miRNA delivery in treatment strategies for OP. Our review seeks to improve the current understanding of the mechanism underlying OP pathogenesis and lay the foundation for the development of novel theranostic approaches for OP.
Collapse
|
26
|
Wang YY, Xia K, Wang ZX, Xie H, Xu R. Osteocyte exosomes accelerate benign prostatic hyperplasia development. Mol Cell Endocrinol 2021; 531:111301. [PMID: 33933560 DOI: 10.1016/j.mce.2021.111301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/05/2021] [Accepted: 04/22/2021] [Indexed: 12/22/2022]
Abstract
Benign prostatic hyperplasia (BPH) is one of the most common diseases in elderly men. BPH patients exhibit an increased risk of vertebral and hip fractures, which are most attributable to pre-existing osteoporosis. However, the relationship between BPH and osteoporosis is still unknown. Here we found that osteocytes, the most abundant bone cells, promoted BPH development by secreting exosomes. In vitro, osteocyte exosomes (OCY-Exo) directly promoted cell proliferation of a prostate epithelial cell line BPH-1 and a macrophage cell line RAW264.7, OCY-Exo also stimulated macrophage-induced proliferation of BPH-1 cells. In vivo, intramedullary injection of OCY-Exo accumulated in prostate. Intravenous administration of OCY-Exo exacerbated testosterone-induced BPH in C57BL/6J mice. Our study uncovers the role of OCY-Exo as a stimulator of BPH, suggesting a novel mechanism in bone-prostate communication.
Collapse
Affiliation(s)
- Yi-Yi Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kun Xia
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhen-Xing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Ran Xu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Endocrine role of bone in the regulation of energy metabolism. Bone Res 2021; 9:25. [PMID: 34016950 PMCID: PMC8137703 DOI: 10.1038/s41413-021-00142-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 12/20/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Bone mainly functions as a supportive framework for the whole body and is the major regulator of calcium homeostasis and hematopoietic function. Recently, an increasing number of studies have characterized the significance of bone as an endocrine organ, suggesting that bone-derived factors regulate local bone metabolism and metabolic functions. In addition, these factors can regulate global energy homeostasis by altering insulin sensitivity, feeding behavior, and adipocyte commitment. These findings may provide a new pathological mechanism for related metabolic diseases or be used in the diagnosis, treatment, and prevention of metabolic diseases such as osteoporosis, obesity, and diabetes mellitus. In this review, we summarize the regulatory effect of bone and bone-derived factors on energy metabolism and discuss directions for future research.
Collapse
|
28
|
Patil KC, Soekmadji C. Extracellular Vesicle-Mediated Bone Remodeling and Bone Metastasis: Implications in Prostate Cancer. Subcell Biochem 2021; 97:297-361. [PMID: 33779922 DOI: 10.1007/978-3-030-67171-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone metastasis is the tendency of certain primary tumors to spawn and dictate secondary neoplasia in the bone. The process of bone metastasis is regulated by the dynamic crosstalk between metastatic cancer cells, cellular components of the bone marrow microenvironment (osteoblasts, osteoclasts, and osteocytes), and the bone matrix. The feed-forward loop mechanisms governs the co-option of homeostatic bone remodeling by cancer cells in bone. Recent developments have highlighted the discovery of extracellular vesicles (EVs) and their diverse roles in distant outgrowths. Several studies have implicated EV-mediated interactions between cancer cells and the bone microenvironment in synergistically promoting pathological skeletal metabolism in the metastatic site. Nevertheless, the potential role that EVs serve in arbitrating intricate sequences of coordinated events within the bone microenvironment remains an emerging field. In this chapter, we review the role of cellular participants and molecular mechanisms in regulating normal bone physiology and explore the progress of current research into bone-derived EVs in directly triggering and coordinating the processes of physiological bone remodeling. In view of the emerging role of EVs in interorgan crosstalk, this review also highlights the multiple systemic pathophysiological processes orchestrated by the EVs to direct organotropism in bone in prostate cancer. Given the deleterious consequences of bone metastasis and its clinical importance, in-depth knowledge of the multifarious role of EVs in distant organ metastasis is expected to open new possibilities for prognostic evaluation and therapeutic intervention for advanced bone metastatic prostate cancer.
Collapse
Affiliation(s)
- Kalyani C Patil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia. .,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
29
|
Gao Y, Chen Y, Wang L, Li C, Ge W. Serum-derived extracellular vesicles inhibit osteoclastogenesis in active-phase patients with SAPHO syndrome. Ther Adv Musculoskelet Dis 2021; 13:1759720X211006966. [PMID: 33948126 PMCID: PMC8053764 DOI: 10.1177/1759720x211006966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/10/2021] [Indexed: 12/02/2022] Open
Abstract
Objective: Synovitis, acne, pustulosis, hyperostosis, and osteitis (SAPHO) syndrome is a rare chronic inflammatory disorder and the underlying pathogenesis is unclear. In this study, 88 SAPHO patients and 118 healthy controls were recruited to investigate the role of serum-derived extracellular vesicles (SEVs) in SAPHO syndrome. Methods: Quantitative proteomics was applied for SEVs proteome identification, and ELISA and Western blotting was performed to verify the results of mass spectrum data. In vitro osteoclastogenesis and osteogenesis assay was used to confirm the effects of SEVs on bone metabolism. Results: Tandem mass tagging-based quantitative proteomic analysis of SAPHO SEVs revealed differential expressed proteins involved in bone metabolism. Of these, serum amyloid A-1 (SAA1) and C-reactive protein (CRP) were upregulated. Higher SAA1 levels in SAPHO patients were confirmed by ELISA. In addition, SAA1 levels were positively correlated with CRP, an inflammatory marker related to the condition of patients. In vitro celluler studies confirmed that SAPHO SEVs inhibited osteoclastogenesis in patients mainly in the active phase of the disease. Further analysis demonstrated that Nucleolin was upregulated in osteoclasts of active-phase patients under SAPHO SEVs stimulation. Conclusion: In this study, we identified SAA1 as an additional inflammation marker that can potentially assist the diagnosis of SAPHO syndrome, and speculated that Nucleolin is a key regulator of osteoclastogenesis in active-phase patients.
Collapse
Affiliation(s)
- Yanpan Gao
- State Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanyu Chen
- State Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Lun Wang
- Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chen Li
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dong Dan San Tiao, Beijing 100730, China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, No. 5 Dong Dan San Tiao, Beijing, 100005, China
| |
Collapse
|
30
|
Leal DV, Ferreira A, Watson EL, Wilund KR, Viana JL. Muscle-Bone Crosstalk in Chronic Kidney Disease: The Potential Modulatory Effects of Exercise. Calcif Tissue Int 2021; 108:461-475. [PMID: 33388899 DOI: 10.1007/s00223-020-00782-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) is a prevalent worldwide public burden that increasingly compromises overall health as the disease progresses. Two of the most negatively affected tissues are bone and skeletal muscle, with CKD negatively impacting their structure, function and activity, impairing the quality of life of these patients and contributing to morbidity and mortality. Whereas skeletal health in this population has conventionally been associated with bone and mineral disorders, sarcopenia has been observed to impact skeletal muscle health in CKD. Indeed, bone and muscle tissues are linked anatomically and physiologically, and together regulate functional and metabolic mechanisms. With the initial crosstalk between the skeleton and muscle proposed to explain bone formation through muscle contraction, it is now understood that this communication occurs through the interaction of myokines and osteokines, with the skeletal muscle secretome playing a pivotal role in the regulation of bone activity. Regular exercise has been reported to be beneficial to overall health. Also, the positive regulatory effect that exercise has been proposed to have on bone and muscle anatomical, functional, and metabolic activity has led to the proposal of regular physical exercise as a therapeutic strategy for muscle and bone-related disorders. The detection of bone- and muscle-derived cytokine secretion following physical exercise has strengthened the idea of a cross communication between these organs. Hence, this review presents an overview of the impact of CKD in bone and skeletal muscle, and narrates how these tissues intrinsically communicate with each other, with focus on the potential effect of exercise in the modulation of this intercommunication.
Collapse
Affiliation(s)
- Diogo V Leal
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University Institute of Maia, ISMAI, Maia, Portugal
| | - Aníbal Ferreira
- Department of Nephrology, Curry Cabral Hospital, Hospital Centre of Central Lisbon, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| | - Emma L Watson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Kenneth R Wilund
- Department of Kinesiology and Community Health, University of Illinois At Urbana-Champaign, Champaign, IL, USA
| | - João L Viana
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University Institute of Maia, ISMAI, Maia, Portugal.
| |
Collapse
|
31
|
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
|
32
|
Honma M, Ikebuchi Y, Suzuki H. Mechanisms of RANKL delivery to the osteoclast precursor cell surface. J Bone Miner Metab 2021; 39:27-33. [PMID: 33047191 DOI: 10.1007/s00774-020-01157-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/14/2020] [Indexed: 01/19/2023]
Abstract
RANKL is biosynthesized as a single-pass transmembrane protein, and soluble molecular species are produced by enzymatic cleavage at the cell surface. Recent studies have revealed that the transmembrane form of RANKL is a major contributor to the induction of mature osteoclasts under physiological conditions in vivo. In osteoblasts and osteocytes, most newly synthesized RANKL forms a protein complex with OPG and is selectively sorted to lysosomes. Only the small proportion of newly synthesized RANKL that does not form a complex with OPG is transported to the cell surface. Then, the transmembrane RANKL is delivered to the surface of osteoclast precursors to stimulate RANK, and induces the activation of a downstream signaling pathway. The ability of osteocytes to support the formation of mature osteoclasts appears to depend upon the amount of RANKL molecules present on their cell surfaces. However, the way in which osteocytes, which are embedded in the bone matrix, deliver transmembrane RANKL to the cell surfaces of osteoclast precursors, which are localized in the bone marrow cavity, remains to be elucidated. Further studies are needed to clarify the mechanisms underlying this process.
Collapse
Affiliation(s)
- Masashi Honma
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yuki Ikebuchi
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
33
|
Gharbi T, Zhang Z, Yang GY. The Function of Astrocyte Mediated Extracellular Vesicles in Central Nervous System Diseases. Front Cell Dev Biol 2020; 8:568889. [PMID: 33178687 PMCID: PMC7593543 DOI: 10.3389/fcell.2020.568889] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Astrocyte activation plays an important role during disease-induced inflammatory response in the brain. Exosomes in the brain could be released from bone marrow (BM)-derived stem cells, neuro stem cells (NSC), mesenchymal stem cells (MSC), etc. We summarized that exosomes release and transport signaling to the target cells, and then produce function. Furthermore, we discussed the pathological interactions between astrocytes and other brain cells, which are related to brain diseases such as stroke, Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) disease, multiple sclerosis (MS), psychiatric, traumatic brain injury (TBI), etc. We provide up-to-date, comprehensive and valuable information on the involvement of exosomes in brain diseases, which is beneficial for basic researchers and clinical physicians.
Collapse
Affiliation(s)
- Tahereh Gharbi
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
34
|
Li S, Wang X. The potential roles of exosomal noncoding RNAs in osteosarcoma. J Cell Physiol 2020; 236:3354-3365. [PMID: 33044018 DOI: 10.1002/jcp.30101] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022]
Abstract
Clinically, it is difficult to efficaciously screen and diagnose osteosarcoma (OS) in advance due to the low sensitivity and poor specificity of the existing tumor markers. Exosomes (Exos) are nanoscale vesicles containing RNAs, lipids, and proteins with a diameter of 30-100 nm. They are multivesicular bodies formed during the invagination of lysosomal particles in cells and released extracellularly after fusing with cell membranes. Besides, Exos are important carriers of cell-to-cell communication signals and genetic materials in the tumor microenvironment. During tumorigenesis, the tumor cells interplay with immune cells, endothelial cells, and related fibroblasts through Exos and boost cancer development. After altering the surrounding microenvironment, the Exos drive tumor cells to proliferate, speed up angiogenesis, and boost cancers to develop along with body fluid transportation. Currently, Exos are becoming novel noninvasive tumor diagnostic markers with high sensitivity, exerting pivotal impacts in fundamental research and clinical applications. Here, we review the existing literature on the roles of exosomal noncoding RNAs in OS progression and their potential clinical applications as novel biomarkers and therapeutics.
Collapse
Affiliation(s)
- Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China.,School of Fundamental Sciences, Center of 3D Printing and Organ Manufacturing, China Medical University (CMU), Shenyang, China
| | - Xiaohong Wang
- School of Fundamental Sciences, Center of 3D Printing and Organ Manufacturing, China Medical University (CMU), Shenyang, China.,Department of Mechanical Engineering, Center of Organ Manufacturing, Tsinghua University, Beijing, China
| |
Collapse
|
35
|
Wu H, Xu Z, Wang Z, Ren Z, Li L, Ruan Y. Exosomes from dendritic cells with Mettl3 gene knockdown prevent immune rejection in a mouse cardiac allograft model. Immunogenetics 2020; 72:423-430. [PMID: 33009922 DOI: 10.1007/s00251-020-01180-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022]
Abstract
We have previously demonstrated that Mettl3-silencing dendritic cells (DCs) exhibited immature properties and prolonged allograft survival in a murine heart transplantation model. Exosomes derived from donor DCs (Dex) are involved in the immune rejection of organ transplantation, and blocking Dex transfer may suppress immune rejection. Herein, this study aimed to investigate whether Mettl3 knockdown inhibits the secretion and activity of donor Dex, thereby inhibiting donor Dex-mediated immune rejection. The imDex, mDex, shCtrl-mDex, and shMettl3-mDex were obtained from the culture supernatant of DCs (immature DCs, mature DCs, shCtrl-infected mature DCs, shMettl3-infected mature DCs) derived from donor BALB/c mouse bone marrow and then co-cultured with splenic T cell lymphocyte suspension from recipient C57BL/6 mice in vitro or injected into recipient C57BL/6 mice before the cardiac transplantation. Donor shMettl3-mDex expressed lower concentration of exosomes and lower expression of Mettl3, Dex markers (ICAM-1, MHC-I, MHC-II), as well as lower ability to activate T cell immune response than shCtrl-mDex. Administration of donor shMettl3-mDex attenuated immune rejection after mouse heart transplantation and prolonged the allograft survival. In summary, Mettl3 knockdown inhibits the immune rejection of Dex in a mouse cardiac allograft model.
Collapse
Affiliation(s)
- Hongbing Wu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University; Hubei Cardiovascular Medicine Clinical Research Center & Hubei Key Laboratory of Cardiology, 238# Jiefang Road, Wuchang District, Hubei Province, Wuhan, China
| | - Zhaojia Xu
- Department of Critical Care Medicine, Jin Yin-tan Hospital, 1# Yin-Tan Road, Dongxihu District, Hubei Province, Wuhan, China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University; Hubei Cardiovascular Medicine Clinical Research Center & Hubei Key Laboratory of Cardiology, 238# Jiefang Road, Wuchang District, Hubei Province, Wuhan, China.
| | - Zongli Ren
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University; Hubei Cardiovascular Medicine Clinical Research Center & Hubei Key Laboratory of Cardiology, 238# Jiefang Road, Wuchang District, Hubei Province, Wuhan, China
| | - Luocheng Li
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University; Hubei Cardiovascular Medicine Clinical Research Center & Hubei Key Laboratory of Cardiology, 238# Jiefang Road, Wuchang District, Hubei Province, Wuhan, China
| | - Yongle Ruan
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University; Hubei Cardiovascular Medicine Clinical Research Center & Hubei Key Laboratory of Cardiology, 238# Jiefang Road, Wuchang District, Hubei Province, Wuhan, China
| |
Collapse
|