1
|
Rice GE, Salomon C. IFPA Joan Hunt Senior Award in Placentology lecture: Extracellular vesicle signalling and pregnancy. Placenta 2024; 157:5-13. [PMID: 38458919 DOI: 10.1016/j.placenta.2024.02.007] [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: 10/15/2023] [Revised: 02/09/2024] [Accepted: 02/21/2024] [Indexed: 03/10/2024]
Abstract
The field of extracellular vesicle (EV) signalling has the potential to transform our understanding of maternal-fetal communication and affords new opportunities for non-invasive prenatal testing and therapeutic intervention. EVs have been implicated in implantation, placentation, maternal adaptation to pregnancy and complications of pregnancy, being detectable in maternal circulation as early as 6 weeks of pregnancy. EVs of differing biogenic origin, composition and bioactivity are released by cells to maintain homoeostasis. Induction of EV signalling is associated with aberrant cellular metabolism and manifests as changes in EV concentrations and/or composition. Characterizing such changes affords opportunity to develop more informative diagnostics and efficacious interventions. To develop accurate and reliable EV-based diagnostics requires: identification of disease-associated biomarkers in specific EV subpopulations; and rapid, reproducible and scalable sample processing. Conventional isolation methods face challenges due to co-isolation of particles with similar physicochemical properties. Methods targeting specific vesicle-surface epitopes and compatible with automated platforms show promise. Effective EV therapeutics require precise targeting, achieved through genetic engineering to release EVs expressing cell-targeting ligands and carrying therapeutic payloads. Unlike cell-based therapies, this approach offers advantages including: low immunogenicity; stability; and long-term storage. Although EV diagnostics and therapeutics in reproductive biology are nascent, available technologies can enhance our understanding of EV signalling between mother and fetus, its role in pregnancies and improve outcomes.
Collapse
Affiliation(s)
- Gregory E Rice
- Inoviq Limited, Notting Hill, Australia; Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia.
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia.
| |
Collapse
|
2
|
Aromolaran KA, Corbin A, Aromolaran AS. Obesity Arrhythmias: Role of IL-6 Trans-Signaling. Int J Mol Sci 2024; 25:8407. [PMID: 39125976 PMCID: PMC11313575 DOI: 10.3390/ijms25158407] [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: 07/07/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Obesity is a chronic disease that is rapidly increasing in prevalence and affects more than 600 million adults worldwide, and this figure is estimated to increase by at least double by 2030. In the United States, more than one-third of the adult population is either overweight or obese. The global obesity epidemic is a major risk factor for the development of life-threatening arrhythmias occurring in patients with long QT, particularly in conditions where multiple heart-rate-corrected QT-interval-prolonging mechanisms are simultaneously present. In obesity, excess dietary fat in adipose tissue stimulates the release of immunomodulatory cytokines such as interleukin (IL)-6, leading to a state of chronic inflammation in patients. Over the last decade, increasing evidence has been found to support IL-6 signaling as a powerful predictor of the severity of heart diseases and increased risk for ventricular arrhythmias. IL-6's pro-inflammatory effects are mediated via trans-signaling and may represent a novel arrhythmogenic risk factor in obese hearts. The first selective inhibitor of IL-6 trans-signaling, olamkicept, has shown encouraging results in phase II clinical studies for inflammatory bowel disease. Nevertheless, the connection between IL-6 trans-signaling and obesity-linked ventricular arrhythmias remains unexplored. Therefore, understanding how IL-6 trans-signaling elicits a cellular pro-arrhythmic phenotype and its use as an anti-arrhythmic target in a model of obesity remain unmet clinical needs.
Collapse
Affiliation(s)
- Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
| | - Andrea Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
3
|
Olson C, Ivanov K, Boyes D, Bengford D, Ku J, Flojo R, Zhang P, Lu B. Dual-Omics Approach Unveils Novel Perspective on the Quality Control of Genetically Engineered Exosomes. Pharmaceutics 2024; 16:824. [PMID: 38931944 PMCID: PMC11207238 DOI: 10.3390/pharmaceutics16060824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Exosomes, nanoscale vesicles derived from human cells, offer great promise for targeted drug delivery. However, their inherent diversity and genetic modifications present challenges in terms of ensuring quality in clinical use. To explore solutions, we employed advanced gene fusion and transfection techniques in human 293T cells to generate two distinct sets of genetically engineered samples. We used dual-omics analysis, combining transcriptomics and proteomics, to comprehensively assess exosome quality by comparing with controls. Transcriptomic profiling showed increased levels of engineering scaffolds in the modified groups, confirming the success of genetic manipulation. Through transcriptomic analysis, we identified 15 RNA species, including 2008 miRNAs and 13,897 mRNAs, loaded onto exosomes, with no significant differences in miRNA or mRNA levels between the control and engineered exosomes. Proteomics analysis identified changes introduced through genetic engineering and over 1330 endogenous exosome-associated proteins, indicating the complex nature of the samples. Further pathway analysis showed enrichment in a small subset of cellular signaling pathways, aiding in our understanding of the potential biological impacts on recipient cells. Detection of over 100 cow proteins highlighted the effectiveness of LC-MS for identifying potential contaminants. Our findings establish a dual-omics framework for the quality control of engineered exosome products, facilitating their clinical translation and therapeutic applications in nanomedicine.
Collapse
Affiliation(s)
- Christopher Olson
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA; (C.O.); (K.I.); (D.B.); (J.K.); (R.F.)
| | - Konstantin Ivanov
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA; (C.O.); (K.I.); (D.B.); (J.K.); (R.F.)
| | - Darin Boyes
- Department of Biology, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA;
| | - David Bengford
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA; (C.O.); (K.I.); (D.B.); (J.K.); (R.F.)
| | - Joy Ku
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA; (C.O.); (K.I.); (D.B.); (J.K.); (R.F.)
| | - Renceh Flojo
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA; (C.O.); (K.I.); (D.B.); (J.K.); (R.F.)
| | - Pengyang Zhang
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA; (C.O.); (K.I.); (D.B.); (J.K.); (R.F.)
| | - Biao Lu
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA; (C.O.); (K.I.); (D.B.); (J.K.); (R.F.)
| |
Collapse
|
4
|
Stawarska A, Bamburowicz-Klimkowska M, Runden-Pran E, Dusinska M, Cimpan MR, Rios-Mondragon I, Grudzinski IP. Extracellular Vesicles as Next-Generation Diagnostics and Advanced Therapy Medicinal Products. Int J Mol Sci 2024; 25:6533. [PMID: 38928240 PMCID: PMC11204223 DOI: 10.3390/ijms25126533] [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: 04/26/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Extracellular vesicles (EVs) hold great promise for clinical application as new diagnostic and therapeutic modalities. This paper describes major GMP-based upstream and downstream manufacturing processes for EV large-scale production, also focusing on post-processing technologies such as surface bioengineering and uploading studies to yield novel EV-based diagnostics and advanced therapy medicinal products. This paper also focuses on the quality, safety, and efficacy issues of the bioengineered EV drug candidates before first-in-human studies. Because clinical trials involving extracellular vesicles are on the global rise, this paper encompasses different clinical studies registered on clinical-trial register platforms, with varying levels of advancement, highlighting the growing interest in EV-related clinical programs. Navigating the regulatory affairs of EVs poses real challenges, and obtaining marketing authorization for EV-based medicines remains complex due to the lack of specific regulatory guidelines for such novel products. This paper discusses the state-of-the-art regulatory knowledge to date on EV-based diagnostics and medicinal products, highlighting further research and global regulatory needs for the safe and reliable implementation of bioengineered EVs as diagnostic and therapeutic tools in clinical settings. Post-marketing pharmacovigilance for EV-based medicinal products is also presented, mainly addressing such topics as risk assessment and risk management.
Collapse
Affiliation(s)
- Agnieszka Stawarska
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, 02-097 Warsaw, Poland; (M.B.-K.); (I.P.G.)
| | - Magdalena Bamburowicz-Klimkowska
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, 02-097 Warsaw, Poland; (M.B.-K.); (I.P.G.)
| | - Elise Runden-Pran
- Health Effects Laboratory, Department of Environmental Chemistry, Norwegian Institute for Air Research, 2007 Kjeller, Norway; (E.R.-P.); (M.D.)
| | - Maria Dusinska
- Health Effects Laboratory, Department of Environmental Chemistry, Norwegian Institute for Air Research, 2007 Kjeller, Norway; (E.R.-P.); (M.D.)
| | - Mihaela Roxana Cimpan
- Biomaterials—Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien Str. 19, 5009 Bergen, Norway; (M.R.C.); (I.R.-M.)
| | - Ivan Rios-Mondragon
- Biomaterials—Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien Str. 19, 5009 Bergen, Norway; (M.R.C.); (I.R.-M.)
| | - Ireneusz P. Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, 02-097 Warsaw, Poland; (M.B.-K.); (I.P.G.)
| |
Collapse
|
5
|
Porcu C, Dobrowolny G, Scicchitano BM. Exploring the Role of Extracellular Vesicles in Skeletal Muscle Regeneration. Int J Mol Sci 2024; 25:5811. [PMID: 38892005 PMCID: PMC11171935 DOI: 10.3390/ijms25115811] [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: 04/30/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Skeletal muscle regeneration entails a multifaceted process marked by distinct phases, encompassing inflammation, regeneration, and remodeling. The coordination of these phases hinges upon precise intercellular communication orchestrated by diverse cell types and signaling molecules. Recent focus has turned towards extracellular vesicles (EVs), particularly small EVs, as pivotal mediators facilitating intercellular communication throughout muscle regeneration. Notably, injured muscle provokes the release of EVs originating from myofibers and various cell types, including mesenchymal stem cells, satellite cells, and immune cells such as M2 macrophages, which exhibit anti-inflammatory and promyogenic properties. EVs harbor a specific cargo comprising functional proteins, lipids, and nucleic acids, including microRNAs (miRNAs), which intricately regulate gene expression in target cells and activate downstream pathways crucial for skeletal muscle homeostasis and repair. Furthermore, EVs foster angiogenesis, muscle reinnervation, and extracellular matrix remodeling, thereby modulating the tissue microenvironment and promoting effective tissue regeneration. This review consolidates the current understanding on EVs released by cells and damaged tissues throughout various phases of muscle regeneration with a focus on EV cargo, providing new insights on potential therapeutic interventions to mitigate muscle-related pathologies.
Collapse
Affiliation(s)
- Cristiana Porcu
- DAHFMO-Unità di Istologia ed Embriologia Medica, Sapienza Università di Roma, 00161 Roma, Italy;
| | - Gabriella Dobrowolny
- DAHFMO-Unità di Istologia ed Embriologia Medica, Sapienza Università di Roma, 00161 Roma, Italy;
| | - Bianca Maria Scicchitano
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| |
Collapse
|
6
|
Cui Y, Lv B, Li Z, Ma C, Gui Z, Geng Y, Liu G, Sang L, Xu C, Min Q, Kong L, Zhang Z, Liu Y, Qi X, Fu D. Bone-Targeted Biomimetic Nanogels Re-Establish Osteoblast/Osteoclast Balance to Treat Postmenopausal Osteoporosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2303494. [PMID: 37794621 DOI: 10.1002/smll.202303494] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/22/2023] [Indexed: 10/06/2023]
Abstract
Insufficient bone formation and excessive bone resorption caused by estrogen deficiency are the major factors resulting in the incidence of postmenopausal osteoporosis (PMOP). The existing drugs usually fail to re-establish the osteoblast/osteoclast balance from both sides and generate side-effects owing to the lack of bone-targeting ability. Here, engineered cell-membrane-coated nanogels PNG@mR&C capable of scavenging receptor activator of nuclear factor-κB ligand (RANKL) and responsively releasing therapeutic PTH 1-34 in the bone microenvironment are prepared from RANK and CXCR4 overexpressed bone mesenchymal stem cell (BMSC) membrane-coated chitosan biopolymers. The CXCR4 on the coated-membranes confer bone-targeting ability, and abundant RANK effectively absorb RANKL to inhibit osteoclastogenesis. Meanwhile, the release of PTH 1-34 triggered by osteoclast-mediated acid microenvironment promote osteogenesis. In addition, the dose and frequency are greatly reduced due to the smart release property, prolonged circulation time, and bone-specific accumulation. Thus, PNG@mR&C exhibits satisfactory therapeutic effects in the ovariectomized (OVX) mouse model. This study provides a new paradigm re-establishing the bone metabolic homeostasis from multitargets and shows great promise for the treatment of PMOP.
Collapse
Affiliation(s)
- Yongzhi Cui
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Bin Lv
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P. R. China
| | - Zhongying Li
- Department of Rehabilitation, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China
| | - Chunming Ma
- Department of Rehabilitation, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China
| | - Zhengwei Gui
- Department of Thyroid and Breast, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Yongtao Geng
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P. R. China
| | - Guohui Liu
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P. R. China
| | - Linchao Sang
- Department of Orthopaedics, The Third Hospital, Hebei Medical University, Shijiazhuang, Hebei, 050051, P. R. China
| | - Chen Xu
- Department of Spine Surgery, Changzheng hospital, Naval Medical University, Shanghai, 200003, P. R. China
| | - Qi Min
- Department of Spine Surgery, Changzheng hospital, Naval Medical University, Shanghai, 200003, P. R. China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Yang Liu
- Department of Spine Surgery, Changzheng hospital, Naval Medical University, Shanghai, 200003, P. R. China
| | - Xiangbei Qi
- Department of Orthopaedics, The Third Hospital, Hebei Medical University, Shijiazhuang, Hebei, 050051, P. R. China
| | - Dehao Fu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| |
Collapse
|
7
|
Huang H, Chen P, Feng X, Qian Y, Peng Z, Zhang T, Wang Q. Translational studies of exosomes in sports medicine - a mini-review. Front Immunol 2024; 14:1339669. [PMID: 38259444 PMCID: PMC10800726 DOI: 10.3389/fimmu.2023.1339669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
This review in sports medicine focuses on the critical role of exosomes in managing chronic conditions and enhancing athletic performance. Exosomes, small vesicles produced by various cells, are essential for cellular communication and transporting molecules like proteins and nucleic acids. Originating from the endoplasmic reticulum, they play a vital role in modulating inflammation and tissue repair. Their significance in sports medicine is increasingly recognized, particularly in healing athletic injuries, improving articular cartilage lesions, and osteoarthritic conditions by modulating cellular behavior and aiding tissue regeneration. Investigations also highlight their potential in boosting athletic performance, especially through myocytes-derived exosomes that may enhance adaptability to physical training. Emphasizing a multidisciplinary approach, this review underlines the need to thoroughly understand exosome biology, including their pathways and classifications, to fully exploit their therapeutic potential. It outlines future directions in sports medicine, focusing on personalized treatments, clinical evaluations, and embracing technological advancements. This research represents a frontier in using exosomes to improve athletes' health and performance capabilities.
Collapse
Affiliation(s)
- Haoqiang Huang
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Peng Chen
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xinting Feng
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yinhua Qian
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Zhijian Peng
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Ting Zhang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Wang
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| |
Collapse
|
8
|
Xue Y, Riva N, Zhao L, Shieh JS, Chin YT, Gatt A, Guo JJ. Recent advances of exosomes in soft tissue injuries in sports medicine: A critical review on biological and biomaterial applications. J Control Release 2023; 364:90-108. [PMID: 37866405 DOI: 10.1016/j.jconrel.2023.10.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/08/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Sports medicine is generally associated with soft tissue injuries including muscle injuries, meniscus and ligament injuries, tendon ruptures, tendinopathy, rotator cuff tears, and tendon-bone healing during injuries. Tendon and ligament injuries are the most common sport injuries accounting for 30-40% of all injuries. Therapies for tendon injuries can be divided into surgical and non-surgical methods. Surgical methods mainly depend on the operative procedures, the surgeons and postoperative interventions. In non-surgical methods, cell therapy with stem cells and cell-free therapy with secretome of stem cell origin are current directions. Exosomes are the main paracrine factors of mesenchymal stem cells (MSCs) containing biological components such as proteins, nucleic acids and lipids. Compared with MSCs, MSC-exosomes (MSC-exos) possess the capacity to escape phagocytosis and achieve long-term circulation. In addition, the functions of exosomes from various cell sources in soft tissue injuries in sports medicine have been gradually revealed in recent years. Along with the biological and biomaterial advances in exosomes, exosomes can be designed as drug carriers with biomaterials and exosome research is providing promising contributions in cell biology. Exosomes with biomaterial have the potential of becoming one of the novel therapeutic modalities in regenerative researches. This review summarizes the derives of exosomes in soft tissue regeneration and focuses on the biological and biomaterial mechanism and advances in exosomal therapy in soft tissue injuries.
Collapse
Affiliation(s)
- Yulun Xue
- Department of Orthopaedic Surgery, Suzhou Municipal Hospital/The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou 215006, Jiangsu, PR China; Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China
| | - Nicoletta Riva
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Lingying Zhao
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health of PR China, Suzhou 215006, Jiangsu, PR China; Department of Hematology, National Clinical Research Center for Hematologic Disease, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China
| | - Ju-Sheng Shieh
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei City 11490, Taiwan
| | - Yu-Tang Chin
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei City 11490, Taiwan
| | - Alexander Gatt
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta; Department of Haematology, Mater Dei Hospital, Msida, Malta
| | - Jiong Jiong Guo
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China; Department of Hematology, National Clinical Research Center for Hematologic Disease, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, PR China.
| |
Collapse
|
9
|
Zhang X, Zhao Y, Yan W. The role of extracellular vesicles in skeletal muscle wasting. J Cachexia Sarcopenia Muscle 2023; 14:2462-2472. [PMID: 37867162 PMCID: PMC10751420 DOI: 10.1002/jcsm.13364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/01/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
Skeletal muscle wasting is a complicated metabolic syndrome accompanied by multiple diseases ranging from cancer to metabolic disorders and infectious conditions. The loss of muscle mass significantly impairs muscle function, resulting in poor quality of life and high mortality of associated diseases. The fundamental cellular and molecular mechanisms inducing muscle wasting have been well established, and those related pathways can be activated by a variety of extracellular signals, including inflammatory cytokines and catabolic stimuli. As an emerging messenger of cell-to-cell communications, extracellular vesicles (EVs) also get involved in the progression of muscle wasting by transferring bioactive cargoes including various proteins and non-coding RNAs to skeletal muscle. Like a double-edged sword, EVs play either a pro-wasting or anti-wasting role in the progression of muscle wasting, highly dependent on their parental cells as well as the specific type of cargo they encapsulate. This review aims to illustrate the current knowledge about the biological function of EVs cargoes in skeletal muscle wasting. Additionally, the potential therapeutic implications of EVs in the diagnosis and treatment of skeletal muscle wasting are also discussed. Simultaneously, several outstanding questions are included to shed light on future research.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical SciencesWuhan UniversityWuhanChina
| | - Yanxia Zhao
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei Yan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical SciencesWuhan UniversityWuhanChina
| |
Collapse
|
10
|
Cheng W, Xu C, Su Y, Shen Y, Yang Q, Zhao Y, Zhao Y, Liu Y. Engineered Extracellular Vesicles: A potential treatment for regeneration. iScience 2023; 26:108282. [PMID: 38026170 PMCID: PMC10651684 DOI: 10.1016/j.isci.2023.108282] [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] [Indexed: 12/01/2023] Open
Abstract
Extracellular vesicles (EVs) play a critical role in various physiological and pathological processes. EVs have gained recognition in regenerative medicine due to their biocompatibility and low immunogenicity. However, the practical application of EVs faces challenges such as limited targeting ability, low yield, and inadequate therapeutic effects. To overcome these limitations, engineered EVs have emerged. This review aims to comprehensively analyze the engineering methods utilized for modifying donor cells and EVs, with a focus on comparing the therapeutic potential between engineered and natural EVs. Additionally, it aims to investigate the specific cell effects that play a crucial role in promoting repair and regeneration, while also exploring the underlying mechanisms involved in the field of regenerative medicine.
Collapse
Affiliation(s)
- Wen Cheng
- Department of Orthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Chenyu Xu
- Department of Orthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Yuran Su
- Department of Orthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Youqing Shen
- Department of Orthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Qiang Yang
- Department of Orthopedics, Tianjin University Tianjin Hospital, Tianjin University, Tianjin 300211, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Yanhong Zhao
- Department of Orthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Yue Liu
- Department of Orthopedics, Tianjin University Tianjin Hospital, Tianjin University, Tianjin 300211, China
| |
Collapse
|
11
|
Gupta D, Orehek S, Turunen J, O’Donovan L, Gait MJ, El-Andaloussi S, Wood MJA. Modulation of Pro-Inflammatory IL-6 Trans-Signaling Axis by Splice Switching Oligonucleotides as a Therapeutic Modality in Inflammation. Cells 2023; 12:2285. [PMID: 37759507 PMCID: PMC10526877 DOI: 10.3390/cells12182285] [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: 07/31/2023] [Revised: 08/31/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine that plays a crucial role in maintaining normal homeostatic processes under the pathogenesis of various inflammatory and autoimmune diseases. This context-dependent effect from a cytokine is due to two distinctive forms of signaling: cis-signaling and trans-signaling. IL-6 cis-signaling involves binding IL-6 to the membrane-bound IL-6 receptor and Glycoprotein 130 (GP130) signal-transducing subunit. By contrast, in IL-6 trans-signaling, complexes of IL-6 and the soluble form of the IL-6 receptor (sIL-6R) signal via membrane-bound GP130. Various strategies have been employed in the past decade to target the pro-inflammatory effect of IL-6 in numerous inflammatory disorders. However, their development has been hindered since these approaches generally target global IL-6 signaling, also affecting the anti-inflammatory effects of IL-6 signaling too. Therefore, novel strategies explicitly targeting the pro-inflammatory IL-6 trans-signaling without affecting the IL-6 cis-signaling are required and carry immense therapeutic potential. Here, we have developed a novel approach to specifically decoy IL-6-mediated trans-signaling by modulating alternative splicing in GP130, an IL-6 signal transducer, by employing splice switching oligonucleotides (SSO), to induce the expression of truncated soluble isoforms of the protein GP130. This isoform is devoid of signaling domains but allows for specifically sequestering the IL-6/sIL-6R receptor complex with high affinity in serum and thereby suppressing inflammation. Using the state-of-the-art Pip6a cell-penetrating peptide conjugated to PMO-based SSO targeting GP130 for efficient in vivo delivery, reduced disease phenotypes in two different inflammatory mouse models of systemic and intestinal inflammation were observed. Overall, this novel gene therapy platform holds great potential as a refined therapeutic intervention for chronic inflammatory diseases.
Collapse
Affiliation(s)
- Dhanu Gupta
- Department of Paediatrics, University of Oxford, Oxford OX3 7TY, UK
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, 14151 Huddinge, Sweden
| | - Sara Orehek
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, 14151 Huddinge, Sweden
| | - Janne Turunen
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, 14151 Huddinge, Sweden
| | - Liz O’Donovan
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Michael J. Gait
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Samir El-Andaloussi
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, 14151 Huddinge, Sweden
| | | |
Collapse
|
12
|
|
13
|
Ulpiano C, da Silva CL, Monteiro GA. Bioengineered Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles as an Improved Drug Delivery System: Methods and Applications. Biomedicines 2023; 11:biomedicines11041231. [PMID: 37189850 DOI: 10.3390/biomedicines11041231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nano-sized lipid membranous structures that modulate cell-cell communication by transporting a variety of biologically active cellular components. The potential of EVs in delivering functional cargos to targeted cells, their capacity to cross biological barriers, as well as their high modification flexibility, make them promising drug delivery vehicles for cell-free therapies. Mesenchymal stromal cells (MSCs) are known for their great paracrine trophic activity, which is largely sustained by the secretion of EVs. MSC-derived EVs (MSC-EVs) retain important features of the parental cells and can be bioengineered to improve their therapeutic payload and target specificity, demonstrating increased therapeutic potential in numerous pre-clinical animal models, including in the treatment of cancer and several degenerative diseases. Here, we review the fundamentals of EV biology and the bioengineering strategies currently available to maximize the therapeutic value of EVs, focusing on their cargo and surface manipulation. Then, a comprehensive overview of the methods and applications of bioengineered MSC-EVs is presented, while discussing the technical hurdles yet to be addressed before their clinical translation as therapeutic agents.
Collapse
Affiliation(s)
- Cristiana Ulpiano
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gabriel A Monteiro
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
14
|
Haghighitalab A, Dominici M, Matin MM, Shekari F, Ebrahimi Warkiani M, Lim R, Ahmadiankia N, Mirahmadi M, Bahrami AR, Bidkhori HR. Extracellular vesicles and their cells of origin: Open issues in autoimmune diseases. Front Immunol 2023; 14:1090416. [PMID: 36969255 PMCID: PMC10031021 DOI: 10.3389/fimmu.2023.1090416] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
The conventional therapeutic approaches to treat autoimmune diseases through suppressing the immune system, such as steroidal and non-steroidal anti-inflammatory drugs, are not adequately practical. Moreover, these regimens are associated with considerable complications. Designing tolerogenic therapeutic strategies based on stem cells, immune cells, and their extracellular vesicles (EVs) seems to open a promising path to managing autoimmune diseases' vast burden. Mesenchymal stem/stromal cells (MSCs), dendritic cells, and regulatory T cells (Tregs) are the main cell types applied to restore a tolerogenic immune status; MSCs play a more beneficial role due to their amenable properties and extensive cross-talks with different immune cells. With existing concerns about the employment of cells, new cell-free therapeutic paradigms, such as EV-based therapies, are gaining attention in this field. Additionally, EVs' unique properties have made them to be known as smart immunomodulators and are considered as a potential substitute for cell therapy. This review provides an overview of the advantages and disadvantages of cell-based and EV-based methods for treating autoimmune diseases. The study also presents an outlook on the future of EVs to be implemented in clinics for autoimmune patients.
Collapse
Affiliation(s)
- Azadeh Haghighitalab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Rebecca Lim
- Department of Obstetrics and Gynaecology, Monash University, Clayton VIC, Australia
| | - Naghmeh Ahmadiankia
- Cancer Prevention Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mahdi Mirahmadi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Reza Bidkhori
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| |
Collapse
|
15
|
Delisle BP, Aromolaran AS. New Insights into Cardiac Ion Channel Regulation 2.0. Int J Mol Sci 2023; 24:4999. [PMID: 36902430 PMCID: PMC10002907 DOI: 10.3390/ijms24054999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/08/2023] Open
Abstract
Sudden cardiac death (SCD) and arrhythmias represent a global public health problem, accounting for 15-20% of all deaths [...].
Collapse
Affiliation(s)
- Brian P. Delisle
- Department of Physiology, 741 S Limestone Street BBSRB B353, Lexington, KY 40536, USA
| | - Ademuyiwa S. Aromolaran
- Department of Surgery, Division of Cardiothoracic Surgery, Nora Eccles Harrison Cardiovascular Research and Training Institute, Salt Lake City, UT 84112, USA
- Department of Biomedical Engineering, Nutrition and Integrative Physiology, Biochemistry and Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
16
|
Kim JW, Jeong MH, Yu HT, Park YJ, Kim HS, Chung KH. Fibrinogen on extracellular vesicles derived from polyhexamethylene guanidine phosphate-exposed mice induces inflammatory effects via integrin β. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114600. [PMID: 36736230 DOI: 10.1016/j.ecoenv.2023.114600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Polyhexamethylene guanidine phosphate (PHMG-p), used as a humidifier disinfectant, causes interstitial lung disease, obliterative bronchiolitis, and lung fibrosis; however, little is known about its effect on intercellular interactions. Extracellular vesicles (EVs), which carry diverse compounds including proteins, RNA, and DNA to mediate cell-to-cell communication through their paracrine effects, have been highlighted as novel factors in lung fibrogenesis. This study aimed to identify the effect of proteins on small EVs (sEVs) from bronchoalveolar lavage fluid (BALF) of the recipient cells after PHMG-p exposure. A week after intratracheal administration of PHMG-p, sEVs were isolated from BALF of tissue showing overexpressed inflammatory and fibrosis markers. To investigate the role of sEVs in inflammation, naïve macrophages were cultured with sEVs, which induced their activation. To identify sEV proteins that are associated with these responses, proteomics analysis was performed. In the gene ontology analysis, coagulation, fibrinolysis, and hemostasis were associated with the upregulated proteins in sEVs. The highest increase was observed in fibrinogen levels, which was also related to those gene ontologies. We validated role of exosomal fibrinogen in inflammation using recombinant fibrinogen and an inhibitor of the integrin, which is the binding receptor for fibrinogen. Overall, we elucidated that increased fibrinogen levels in the early sEVs-PHMG activated inflammatory response during early fibrosis. These results suggest that sEVs from the BALF of PHMG-p-exposed mice could aggravate fibrogenesis by activating naïve macrophages via various proteins in the sEVs, Furthermore, this finding will be broadening the spectrum of communicating mediators.
Collapse
Affiliation(s)
- Jun Woo Kim
- Sungkyunkwan University, School of Pharmacy, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Mi Ho Jeong
- Massachusetts General Hospital, Center for Systems Biology, Boston, MA 02114, USA
| | - Hyeong Tae Yu
- Sungkyunkwan University, School of Pharmacy, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Yong Joo Park
- Kyungsung University, College of Pharmacy, Busan 48434, Republic of Korea
| | - Hyung Sik Kim
- Sungkyunkwan University, School of Pharmacy, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Kyu Hyuck Chung
- Sungkyunkwan University, School of Pharmacy, Suwon, Gyeonggi-do 16419, Republic of Korea.
| |
Collapse
|
17
|
Abstract
Skeletal muscle plays a paramount role in physical activity, metabolism, and energy balance, while its homeostasis is being challenged by multiple unfavorable factors such as injury, aging, or obesity. Exosomes, a subset of extracellular vesicles, are now recognized as essential mediators of intercellular communication, holding great clinical potential in the treatment of skeletal muscle diseases. Herein, we outline the recent research progress in exosomal isolation, characterization, and mechanism of action, and emphatically discuss current advances in exosomes derived from multiple organs and tissues, and engineered exosomes regarding the regulation of physiological and pathological development of skeletal muscle. These remarkable advances expand our understanding of myogenesis and muscle diseases. Meanwhile, the engineered exosome, as an endogenous nanocarrier combined with advanced design methodologies of biomolecules, will help to open up innovative therapeutic perspectives for the treatment of muscle diseases.
Collapse
|
18
|
Alptekin A, Parvin M, Chowdhury HI, Rashid MH, Arbab AS. Engineered exosomes for studies in tumor immunology. Immunol Rev 2022; 312:76-102. [PMID: 35808839 DOI: 10.1111/imr.13107] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
Abstract
Exosomes are a type of extracellular vesicle (EV) with diameters of 30-150 nm secreted by most of the cells into the extracellular spaces and can alter the microenvironment through cell-to-cell interactions by fusion with the plasma membrane and subsequent endocytosis and release of the cargo. Because of their biocompatibility, low toxicity and immunogenicity, permeability (even through the blood-brain barrier (BBB)), stability in biological fluids, and ability to accumulate in the lesions with higher specificity, investigators have started making designer's exosomes or engineered exosomes to carry biologically active protein on the surface or inside the exosomes as well as using exosomes to carry drugs, micro RNA, and other products to the site of interest. In this review, we have discussed biogenesis, markers, and contents of various exosomes including exosomes of immune cells. We have also discussed the current methods of making engineered and designer's exosomes as well as the use of engineered exosomes targeting different immune cells in the tumors, stroke, as well as at peripheral blood. Genetic engineering and customizing exosomes create an unlimited opportunity to use in diagnosis and treatment. Very little use has been discovered, and we are far away to reach its limits.
Collapse
Affiliation(s)
- Ahmet Alptekin
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Mahrima Parvin
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | | | | | - Ali S Arbab
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
19
|
Tailored Extracellular Vesicles: Novel Tool for Tissue Regeneration. Stem Cells Int 2022; 2022:7695078. [PMID: 35915850 PMCID: PMC9338735 DOI: 10.1155/2022/7695078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/10/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022] Open
Abstract
Extracellular vesicles (EVs) play an essential part in multiple pathophysiological processes including tissue injury and regeneration because of their inherent characteristics of small size, low immunogenicity and toxicity, and capability of carrying a variety of bioactive molecules and mediating intercellular communication. Nevertheless, accumulating studies have shown that the application of EVs faces many challenges such as insufficient therapeutic efficacy, a lack of targeting capability, low yield, and rapid clearance from the body. It is known that EVs can be engineered, modified, and designed to encapsulate therapeutic cargos like proteins, peptides, nucleic acids, and drugs to improve their therapeutic efficacy. Targeted peptides, antibodies, aptamers, magnetic nanoparticles, and proteins are introduced to modify various cell-derived EVs for increasing targeting ability. In addition, extracellular vesicle mimetics (EMs) and self-assembly EV-mimicking nanocomplex are applied to improve production and simplify EV purification process. The combination of EVs with biomaterials like hydrogel, and scaffolds dressing endows EVs with long-term therapeutic efficacy and synergistically enhanced regenerative outcome. Thus, we will summarize recent developments of EV modification strategies for more extraordinary regenerative effect in various tissue injury repair. Subsequently, opportunities and challenges of promoting the clinical application of engineered EVs will be discussed.
Collapse
|
20
|
Forcina L, Franceschi C, Musarò A. The hormetic and hermetic role of IL-6. Ageing Res Rev 2022; 80:101697. [PMID: 35850167 DOI: 10.1016/j.arr.2022.101697] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/24/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
Abstract
Interleukin-6 is a pleiotropic cytokine regulating different tissues and organs in diverse and sometimes discrepant ways. The dual and sometime hermetic nature of IL-6 action has been highlighted in several contexts and can be explained by the concept of hormesis, in which beneficial or toxic effects can be induced by the same molecule depending on the intensity, persistence, and nature of the stimulation. According with hormesis, a low and/or controlled IL-6 release is associated with anti-inflammatory, antioxidant, and pro-myogenic actions, whereas increased systemic levels of IL-6 can induce pro-inflammatory, pro-oxidant and pro-fibrotic responses. However, many aspects regarding the multifaceted action of IL-6 and the complex nature of its signal transduction remains to be fully elucidated. In this review we collect mechanistic insight into the molecular networks contributing to normal or pathologic changes during advancing age and in chronic diseases. We point out the involvement of IL-6 deregulation in aging-related diseases, dissecting the hormetic action of this key mediator in different tissues, with a special focus on skeletal muscle. Since IL-6 can act as an enhancer of detrimental factor associated with both aging and pathologic conditions, such as chronic inflammation and oxidative stress, this cytokine could represent a "Gerokine", a determinant of the switch from physiologic aging to age-related diseases.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, Rome 00161, Italy.
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Scuola Superiore di Studi Avanzati Sapienza (SSAS), Via A. Scarpa, 14, Rome 00161, Italy.
| |
Collapse
|
21
|
Wu C, Xu Q, Wang H, Tu B, Zeng J, Zhao P, Shi M, Qiu H, Huang Y. Neutralization of SARS-CoV-2 pseudovirus using ACE2-engineered extracellular vesicles. Acta Pharm Sin B 2022; 12:1523-1533. [PMID: 34522576 PMCID: PMC8427979 DOI: 10.1016/j.apsb.2021.09.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/04/2021] [Accepted: 08/12/2021] [Indexed: 12/11/2022] Open
Abstract
The spread of coronavirus disease 2019 (COVID-19) throughout the world has resulted in stressful healthcare burdens and global health crises. Developing an effective measure to protect people from infection is an urgent need. The blockage of interaction between angiotensin-converting enzyme 2 (ACE2) and S protein is considered an essential target for anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) drugs. A full-length ACE2 protein could be a potential drug to block early entry of SARS-CoV-2 into host cells. In this study, a therapeutic strategy was developed by using extracellular vesicles (EVs) with decoy receptor ACE2 for neutralization of SARS-CoV-2. The EVs embedded with engineered ACE2 (EVs-ACE2) were prepared; the EVs-ACE2 were derived from an engineered cell line with stable ACE2 expression. The potential effect of the EVs-ACE2 on anti-SARS-CoV-2 was demonstrated by both in vitro and in vivo neutralization experiments using the pseudovirus with the S protein (S-pseudovirus). EVs-ACE2 can inhibit the infection of S-pseudovirus in various cells, and importantly, the mice treated with intranasal administration of EVs-ACE2 can suppress the entry of S-pseudovirus into the mucosal epithelium. Therefore, the intranasal EVs-ACE2 could be a preventive medicine to protect from SARS-CoV-2 infection. This EVs-based strategy offers a potential route to COVID-19 drug development.
Collapse
Key Words
- ACE2
- ACE2, angiotensin-converting enzyme 2
- BSA, bovine albumin
- COVID-19
- EVs, extracellular vesicles
- Extracellular vesicles
- FBS, fetal bovine serum
- Intranasal administration
- NTA, nanoparticle tracking analysis
- Neutralization
- PAGE, polyacrylamide gel electrophoresis
- Pseudovirus
- RIPA, radio immunoprecipitation assay
- RLU, relative luminescence units
- S protein, spike protein
- SARS-CoV-2
- SDS, sodium dodecyl sulfate
- Spike protein
- TEM, transmission electron microscope
- WB, western blot
Collapse
Affiliation(s)
- Canhao Wu
- Artemisinin Research Center, First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qin Xu
- Artemisinin Research Center, First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
| | - Huiyuan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bin Tu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan 528437, China
| | - Jiaxin Zeng
- Artemisinin Research Center, First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Pengfei Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Center of Clinical Pharmacology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| | - Mingjie Shi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hong Qiu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan 528437, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China
- Taizhou University, School of Advanced Study, Institute of Natural Medicine and Health Product, Taizhou 318000, China
| |
Collapse
|
22
|
Abstract
Extracellular vesicles are lipid-bilayer-enclosed nanoparticles present in the majority of biological fluids that mediate intercellular communication. EVs are able to transfer their contents (including nucleic acids, proteins, lipids, and small molecules) to recipient cells, and thus hold great promise as drug delivery vehicles. However, their therapeutic application is limited by lack of efficient cargo loading strategies, a need to improve EV tissue-targeting capabilities and a requirement to improve escape from the endolysosomal system. These challenges can be effectively addressed by modifying EVs with peptides which confer specific advantageous properties, thus enhancing their therapeutic potential. Here we provide an overview of the applications of peptide technology with respect to EV therapeutics. We focus on the utility of EV-modifying peptides for the purposes of promoting cargo loading, tissue-targeting and endosomal escape, leading to enhanced delivery of the EV cargo to desired cells/tissues and subcellular target locations. Both endogenous and exogenous methods for modifying EVs with peptides are considered.
Collapse
|
23
|
Gupta D, Wiklander OPB, Görgens A, Conceição M, Corso G, Liang X, Seow Y, Balusu S, Feldin U, Bostancioglu B, Jawad R, Mamand DR, Lee YXF, Hean J, Mäger I, Roberts TC, Gustafsson M, Mohammad DK, Sork H, Backlund A, Lundin P, de Fougerolles A, Smith CIE, Wood MJA, Vandenbroucke RE, Nordin JZ, El-Andaloussi S. Amelioration of systemic inflammation via the display of two different decoy protein receptors on extracellular vesicles. Nat Biomed Eng 2021; 5:1084-1098. [PMID: 34616047 DOI: 10.1038/s41551-021-00792-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/07/2021] [Indexed: 02/01/2023]
Abstract
Extracellular vesicles (EVs) can be functionalized to display specific protein receptors on their surface. However, surface-display technology typically labels only a small fraction of the EV population. Here, we show that the joint display of two different therapeutically relevant protein receptors on EVs can be optimized by systematically screening EV-loading protein moieties. We used cytokine-binding domains derived from tumour necrosis factor receptor 1 (TNFR1) and interleukin-6 signal transducer (IL-6ST), which can act as decoy receptors for the pro-inflammatory cytokines tumour necrosis factor alpha (TNF-α) and IL-6, respectively. We found that the genetic engineering of EV-producing cells to express oligomerized exosomal sorting domains and the N-terminal fragment of syntenin (a cytosolic adaptor of the single transmembrane domain protein syndecan) increased the display efficiency and inhibitory activity of TNFR1 and IL-6ST and facilitated their joint display on EVs. In mouse models of systemic inflammation, neuroinflammation and intestinal inflammation, EVs displaying the cytokine decoys ameliorated the disease phenotypes with higher efficacy as compared with clinically approved biopharmaceutical agents targeting the TNF-α and IL-6 pathways.
Collapse
Affiliation(s)
- Dhanu Gupta
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Oscar P B Wiklander
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - André Görgens
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Giulia Corso
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xiuming Liang
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yiqi Seow
- Molecular Engineering Laboratory, Institute for Bioengineering and Nanotechnology, A*STAR, Singapore, Singapore
| | - Sriram Balusu
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ulrika Feldin
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Beklem Bostancioglu
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rim Jawad
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Doste R Mamand
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Biology Department, Cihan University-Erbil, Erbil, Iraq
| | - Yi Xin Fiona Lee
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Genome Institute of Singapore, Agency for Science, Technology and Research, A*STAR, Singapore, Singapore
| | | | - Imre Mäger
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Thomas C Roberts
- Department of Paediatrics, University of Oxford, Oxford, UK.,MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Manuela Gustafsson
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Dara K Mohammad
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,College of Agricultural Engineering Sciences, Salahaddin University-Erbil, Erbil, Iraq
| | - Helena Sork
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alexandra Backlund
- Cardiovascular Medicine Unit, Department of Medicine, Solna, Karolinska Institute, Stockholm, Sweden
| | | | | | - C I Edvard Smith
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Matthew J A Wood
- Department of Paediatrics, University of Oxford, Oxford, UK.,MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Joel Z Nordin
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Samir El-Andaloussi
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Massaro C, Min W, Pegtel DM, Baglio SR. Harnessing EV communication to restore antitumor immunity. Adv Drug Deliv Rev 2021; 176:113838. [PMID: 34144088 DOI: 10.1016/j.addr.2021.113838] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 02/06/2023]
Abstract
Restoring effective anti-tumor immune responses to cure cancer is a promising strategy, but challenging to achieve due to the intricate crosstalk between tumor and immune cells. While it is established that tumor cells acquire traits to escape immune recognition, the involvement of extracellular vesicles (EVs) in curbing immune cell activation is rapidly emerging. By assisting cancer cells in spreading immunomodulatory signals in the form of (glyco)proteins, lipids, nucleic acids and metabolic regulators, EVs recently emerged as versatile mediators of immune suppression. Blocking their action might reactivate immune cell function and natural antitumor immune responses. Alternatively, EV communication may be exploited to boost anti-tumor immunity. Indeed, novel insights into EV biology paved the way for efficient ex vivo production of 'rationally engineered' EVs that function as potent antitumor vaccines or carry out specific functional tasks. In this review we discuss the latest findings on immune regulation by cancer EVs and explore how EV-mediated communication can be either targeted or harnessed to restore immunity as a means for cancer therapy.
Collapse
|
25
|
Johnson J, Shojaee M, Mitchell Crow J, Khanabdali R. From Mesenchymal Stromal Cells to Engineered Extracellular Vesicles: A New Therapeutic Paradigm. Front Cell Dev Biol 2021; 9:705676. [PMID: 34409037 PMCID: PMC8366519 DOI: 10.3389/fcell.2021.705676] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells obtained from many tissues including bone marrow, adipose tissue, umbilical cord, amniotic fluid, and placenta. MSCs are the leading cell source for stem cell therapy due to their regenerative and immunomodulatory properties, their low risk of tumorigenesis and lack of ethical constraints. However, clinical applications of MSCs remain limited. MSC therapeutic development continues to pose challenges in terms of preparation, purity, consistency, efficiency, reproducibility, processing time and scalability. Additionally, there are issues with their poor engraftment and survival in sites of disease or damage that limit their capacity to directly replace damaged cells. A key recent development in MSC research, however, is the now widely accepted view that MSCs primarily exert therapeutic effects via paracrine factor secretion. One of the major paracrine effectors are extracellular vesicles (EVs). EVs represent a potential cell-free alternative to stem cell therapy but are also rapidly emerging as a novel therapeutic platform in their own right, particularly in the form of engineered EVs (EEVs) tailored to target a broad range of clinical indications. However, the development of EVs and EEVs for therapeutic application still faces a number of hurdles, including the establishment of a consistent, scalable cell source, and the development of robust GMP-compliant upstream and downstream manufacturing processes. In this review we will highlight the clinical challenges of MSC therapeutic development and discuss how EVs and EEVs can overcome the challenges faced in the clinical application of MSCs.
Collapse
Affiliation(s)
- Jancy Johnson
- Exopharm Ltd., Melbourne, VIC, Australia.,Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, Australia
| | | | | | | |
Collapse
|
26
|
Pelosi L, Berardinelli MG, Forcina L, Ascenzi F, Rizzuto E, Sandri M, De Benedetti F, Scicchitano BM, Musarò A. Sustained Systemic Levels of IL-6 Impinge Early Muscle Growth and Induce Muscle Atrophy and Wasting in Adulthood. Cells 2021; 10:1816. [PMID: 34359985 PMCID: PMC8306542 DOI: 10.3390/cells10071816] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023] Open
Abstract
IL-6 is a pleiotropic cytokine that can exert different and opposite effects. The muscle-induced and transient expression of IL-6 can act in an autocrine or paracrine manner, stimulating anabolic pathways associated with muscle growth, myogenesis, and with regulation of energy metabolism. In contrast, under pathologic conditions, including muscular dystrophy, cancer associated cachexia, aging, chronic inflammatory diseases, and other pathologies, the plasma levels of IL-6 significantly increase, promoting muscle wasting. Nevertheless, the specific physio-pathological role exerted by IL-6 in the maintenance of differentiated phenotype remains to be addressed. The purpose of this study was to define the role of increased plasma levels of IL-6 on muscle homeostasis and the mechanisms contributing to muscle loss. Here, we reported that increased plasma levels of IL-6 promote alteration in muscle growth at early stage of postnatal life and induce muscle wasting by triggering a shift of the slow-twitch fibers toward a more sensitive fast fiber phenotype. These findings unveil a role for IL-6 as a potential biomarker of stunted growth and skeletal muscle wasting.
Collapse
Affiliation(s)
- Laura Pelosi
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161 Rome, Italy; (L.P.); (M.G.B.); (L.F.)
| | - Maria Grazia Berardinelli
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161 Rome, Italy; (L.P.); (M.G.B.); (L.F.)
| | - Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161 Rome, Italy; (L.P.); (M.G.B.); (L.F.)
| | - Francesca Ascenzi
- Department of Clinical and Molecular Medicine, Risk Management Q and A, Sant’Andrea Hospital, “Sapienza” University, 00161 Rome, Italy;
| | - Emanuele Rizzuto
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184 Rome, Italy;
| | - Marco Sandri
- Veneto Institute of Molecular Medicine, 35129 Padua, Italy;
- Department of Biomedical Sciences, University of Padova, 35121 Padua, Italy
| | - Fabrizio De Benedetti
- Division of Rheumatology and Immuno-Rheumatology Research Laboratories, Bambino Gesù Children’s Hospital, 00146 Rome, Italy;
| | - Bianca Maria Scicchitano
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Rome, Italy;
| | - Antonio Musarò
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via Antonio Scarpa, 14, 00161 Rome, Italy
- Scuola Superiore di Studi Avanzati Sapienza (SSAS), Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
27
|
Sandonà M, Di Pietro L, Esposito F, Ventura A, Silini AR, Parolini O, Saccone V. Mesenchymal Stromal Cells and Their Secretome: New Therapeutic Perspectives for Skeletal Muscle Regeneration. Front Bioeng Biotechnol 2021; 9:652970. [PMID: 34095095 PMCID: PMC8172230 DOI: 10.3389/fbioe.2021.652970] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells found in different tissues: bone marrow, peripheral blood, adipose tissues, skeletal muscle, perinatal tissues, and dental pulp. MSCs are able to self-renew and to differentiate into multiple lineages, and they have been extensively used for cell therapy mostly owing to their anti-fibrotic and immunoregulatory properties that have been suggested to be at the basis for their regenerative capability. MSCs exert their effects by releasing a variety of biologically active molecules such as growth factors, chemokines, and cytokines, either as soluble proteins or enclosed in extracellular vesicles (EVs). Analyses of MSC-derived secretome and in particular studies on EVs are attracting great attention from a medical point of view due to their ability to mimic all the therapeutic effects produced by the MSCs (i.e., endogenous tissue repair and regulation of the immune system). MSC-EVs could be advantageous compared with the parental cells because of their specific cargo containing mRNAs, miRNAs, and proteins that can be biologically transferred to recipient cells. MSC-EV storage, transfer, and production are easier; and their administration is also safer than MSC therapy. The skeletal muscle is a very adaptive tissue, but its regenerative potential is altered during acute and chronic conditions. Recent works demonstrate that both MSCs and their secretome are able to help myofiber regeneration enhancing myogenesis and, interestingly, can be manipulated as a novel strategy for therapeutic interventions in muscular diseases like muscular dystrophies or atrophy. In particular, MSC-EVs represent promising candidates for cell free-based muscle regeneration. In this review, we aim to give a complete picture of the therapeutic properties and advantages of MSCs and their products (MSC-derived EVs and secreted factors) relevant for skeletal muscle regeneration in main muscular diseases.
Collapse
Affiliation(s)
- Martina Sandonà
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy
| | - Lorena Di Pietro
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federica Esposito
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy
| | - Alessia Ventura
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca "E. Menni", Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Valentina Saccone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy.,Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
28
|
Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol 2021; 14:24. [PMID: 33579329 PMCID: PMC7880217 DOI: 10.1186/s13045-021-01037-x] [Citation(s) in RCA: 316] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stromal cells (MSCs), also known as mesenchymal stem cells, have been intensely investigated for clinical applications within the last decades. However, the majority of registered clinical trials applying MSC therapy for diverse human diseases have fallen short of expectations, despite the encouraging pre-clinical outcomes in varied animal disease models. This can be attributable to inconsistent criteria for MSCs identity across studies and their inherited heterogeneity. Nowadays, with the emergence of advanced biological techniques and substantial improvements in bio-engineered materials, strategies have been developed to overcome clinical challenges in MSC application. Here in this review, we will discuss the major challenges of MSC therapies in clinical application, the factors impacting the diversity of MSCs, the potential approaches that modify MSC products with the highest therapeutic potential, and finally the usage of MSCs for COVID-19 pandemic disease.
Collapse
|