1
|
Jin C, Xue L, Zhang L, Yu L, Wu P, Qian H. Engineered Nanoparticles for Theranostic Applications in Kidney Repair. Adv Healthc Mater 2025; 14:e2402480. [PMID: 39617999 DOI: 10.1002/adhm.202402480] [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: 07/06/2024] [Revised: 11/05/2024] [Indexed: 01/03/2025]
Abstract
Kidney diseases are characterized by their intricate nature and complexity, posing significant challenges in their treatment and diagnosis. Nanoparticles (NPs), which can be further classified as synthetic and biomimetic NPs, have emerged as promising candidates for treating various diseases. In recent years, the development of engineered nanotherapeutics has focused on targeting damaged tissues and serving as drug delivery vehicles. Additionally, these NPs have shown superior sensitivity and specificity in diagnosis and imaging, thus providing valuable insights for the early detection of diseases. This review aims to focus on the application of engineered synthetic and biomimetic NPs in kidney diseases in the aspects of treatment, diagnosis, and imaging. Notably, the current perspectives and challenges are evaluated, which provide inspiration for future research directions, and encourage the clinical application of NPs in this field.
Collapse
Affiliation(s)
- Can Jin
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Lingling Xue
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Leilei Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Lixia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
| | - Peipei Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| |
Collapse
|
2
|
Yun YG, Yeo D, Shin SJ, Shin JS, Lee JH, Kim HW. Polydeoxyribonucleotide enhances the bioactivities of stem cells from human exfoliated deciduous teeth through Akt activation. Biochem Biophys Res Commun 2024; 739:150947. [PMID: 39550860 DOI: 10.1016/j.bbrc.2024.150947] [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: 07/16/2024] [Revised: 10/06/2024] [Accepted: 11/02/2024] [Indexed: 11/19/2024]
Abstract
Although numerous approaches have emerged to address the challenges of critical limb ischemia (CLI), their clinical trials have proven elusive. Stem cell therapy has been utilized for CLI; however, its efficacy is limited, resulting in low survival rates in patients. Here, we investigated the impact of polydeoxyribonucleotide (PDRN) on the bioactivities of stem cells derived from human exfoliated deciduous teeth (SHED) against oxidative stress. PDRN treatment increased the proliferation, migration, antioxidant properties, and mitochondrial respiration of SHED. These beneficial effects were regulated by Akt activation. Through a murine hindlimb ischemia model, PDRN treatment demonstrated augmented the survival and proliferation of transplanted SHED at ischemic injury sites, whereas the inhibition of Akt suppressed these effects. Our findings revealed that PDRN promoted the therapeutic potential of SHED via Akt phosphorylation, suggesting PDRN-primed SHED as promising candidates for the development of novel stem cell therapeutics.
Collapse
Affiliation(s)
- Yeo Gyun Yun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Donghyeon Yeo
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Seong-Jin Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Ji-Sun Shin
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea; Department of Pediatric Dentistry, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea.
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea.
| |
Collapse
|
3
|
Lee SY, Park JM, Rhim WK, Lee EH, Lee SH, Kim JY, Cha SG, Lee SH, Kim B, Hwang DY, Rho S, Ahn TK, Kim BS, Han DK. Multifunctional extracellular vesicles and edaravone-loaded scaffolds for kidney tissue regeneration by activating GDNF/RET pathway. NANO CONVERGENCE 2024; 11:43. [PMID: 39460807 PMCID: PMC11512987 DOI: 10.1186/s40580-024-00450-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
With the severity of chronic kidney disease worldwide, strategies to recover renal function via tissue regeneration provide alternatives to kidney replacement therapy. To exclude side effects from direct cell transplantation, extracellular vesicles (EVs) are great substitutes representing paracrine cell signaling. To build three-dimensional structures for implantation into the 5/6 nephrectomy model by incorporating bioactive materials, including multifunctional EVs (mEVs), porous PMEZE/mEV scaffolds were developed in combination with edaravone (EDV; E) and mEV based on PMEZ scaffolds with PLGA (P), MH-RA (M), ECM (E), ZnO-ALA (Z). The oxygen free radical scavenger EDV was incorporated to induce tubular regeneration. mEVs were engineered to serve regenerative activities with a combination of two EVs from SDF-1α overexpressed tonsil-derived mesenchymal stem cells (sEVs) and intermediate mesoderm (IM) cells during differentiation into kidney progenitor cells (dEVs). mEVs displayed beneficial effects on regeneration by facilitating migration and inducing differentiation of surrounding stem cells, and EDV improved kidney function by regulating the GDNF/RET pathway and their downstream genes. The promotion of MSC recruitment was confirmed with sEV particles number dependently, and the regulation of the GDNF/RET pathway by the effect of EDV and its enhanced effect by mEVs were elucidated using in vitro analysis. The regeneration of tubules was additionally demonstrated through the increased expression of aquaporin-1 (AQP-1) and cadherin-16 (CDH16) for proximal tubules, and calbindin and PAX2 for distal tubules in the renal defect model. With these, structural regeneration and functional recovery were achieved with kidney regeneration in the 5/6 nephrectomy mice model.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-Do, 13488, Republic of Korea
| | - Jeong Min Park
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-Do, 13488, Republic of Korea
| | - Won-Kyu Rhim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-Do, 13488, Republic of Korea
| | - Eun Hye Lee
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-Gu, Daegu, 41944, Republic of Korea
| | - Sang-Hyuk Lee
- Department of Microbiology, School of Medicine, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea
| | - Jun Yong Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-Do, 13488, Republic of Korea
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-Do, 13488, Republic of Korea
| | - Sun Hong Lee
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-Do, 13488, Republic of Korea
| | - Boram Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-Do, 13488, Republic of Korea
| | - Dong-Youn Hwang
- Department of Microbiology, School of Medicine, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea
| | - Seungsoo Rho
- Department of Ophthalmology, CHA Bundang Medical Center, CHA University, 59 Yatap-Ro, Bundang-Gu, Seongnam, Gyeonggi-Do, 13496, Republic of Korea
| | - Tae-Keun Ahn
- Department of Orthopedic Surgery CHA Bundang Medical Center, CHA University, Seonnam, Gyeonggi-Do, 13496, Republic of Korea
| | - Bum Soo Kim
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-Gu, Daegu, 41944, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-Do, 13488, Republic of Korea.
| |
Collapse
|
4
|
Liu H, Deng H, Huang H, Cao J, Wang X, Zhou Z, Zhong Z, Chen D, Peng G. Canine mesenchymal stem cell-derived exosomes attenuate renal ischemia-reperfusion injury through miR-146a-regulated macrophage polarization. Front Vet Sci 2024; 11:1456855. [PMID: 39315083 PMCID: PMC11417097 DOI: 10.3389/fvets.2024.1456855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction The most common factor leading to renal failure or death is renal IR (ischemia-reperfusion). Studies have shown that mesenchymal stem cells (MSCs) and their exosomes have potential therapeutic effects for IR injury by inhibiting M1 macrophage polarization and inflammation. In this study, the protective effect and anti-inflammatory mechanism of adipose-derived mesenchymal stem cell-derived exosomes (ADMSC-Exos) after renal IR were investigated. Method Initially, ADMSC-Exos were intravenously injected into IR experimental beagles, and the subsequent assessment focused on inflammatory damage and macrophage phenotype. Furthermore, an in vitro inflammatory model was established by inducing DH82 cells with LPS. The impact on inflammation and macrophage phenotype was then evaluated using ADMSC and regulatory miR-146a. Results Following the administration of ADMSC-Exos in IR canines, a shift from M1 to M2 macrophage polarization was observed. Similarly, in vitro experiments demonstrated that ADMSC-Exos enhanced the transformation of LPS-induced macrophages from M1 to M2 type. Notably, the promotion of macrophage polarization by ADMSC-Exos was found to be attenuated upon the inhibition of miR-146a in ADMSC-Exos. Conclusion These findings suggest that miR-146a plays a significant role in facilitating the transition of LPS-induced macrophages from M1 to M2 phenotype. As a result, the modulation of macrophage polarization by ADMSC-Exos is achieved via the encapsulation and conveyance of miR-146a, leading to diminished infiltration of inflammatory cells in renal tissue and mitigation of the inflammatory reaction following canine renal IR.
Collapse
Affiliation(s)
- HaiFeng Liu
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hongchuan Deng
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Haocheng Huang
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jiahui Cao
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xinmiao Wang
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ziyao Zhou
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhijun Zhong
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dechun Chen
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, China
| | - Guangneng Peng
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
5
|
Shi X, Hu X, Jiang N, Mao J. Regenerative endodontic therapy: From laboratory bench to clinical practice. J Adv Res 2024:S2090-1232(24)00267-4. [PMID: 38969092 DOI: 10.1016/j.jare.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/16/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Maintaining the vitality and functionality of dental pulp is paramount for tooth integrity, longevity, and homeostasis. Aiming to treat irreversible pulpitis and necrosis, there has been a paradigm shift from conventional root canal treatment towards regenerative endodontic therapy. AIM OF REVIEW This extensive and multipart review presents crucial laboratory and practical issues related to pulp-dentin complex regeneration aimed towards advancing clinical translation of regenerative endodontic therapy and enhancing human life quality. KEY SCIENTIFIC CONCEPTS OF REVIEW In this multipart review paper, we first present a panorama of emerging potential tissue engineering strategies for pulp-dentin complex regeneration from cell transplantation and cell homing perspectives, emphasizing the critical regenerative components of stem cells, biomaterials, and conducive microenvironments. Then, this review provides details about current clinically practiced pulp regenerative/reparative approaches, including direct pulp capping and root revascularization, with a specific focus on the remaining hurdles and bright prospects in developing such therapies. Next, special attention was devoted to discussing the innovative biomimetic perspectives opened in establishing functional tissues by employing exosomes and cell aggregates, which will benefit the clinical translation of dental pulp engineering protocols. Finally, we summarize careful consideration that should be given to basic research and clinical applications of regenerative endodontics. In particular, this review article highlights significant challenges associated with residual infection and inflammation and identifies future insightful directions in creating antibacterial and immunomodulatory microenvironments so that clinicians and researchers can comprehensively understand crucial clinical aspects of regenerative endodontic procedures.
Collapse
Affiliation(s)
- Xin Shi
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiaohan Hu
- Outpatient Department Office, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Nan Jiang
- Central Laboratory, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Jing Mao
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China.
| |
Collapse
|
6
|
Augustin G, Jeong JH, Kim M, Hur SS, Lee JH, Hwang Y. Stem Cell‐Based Therapies and Tissue Engineering Innovations for Tendinopathy: A Comprehensive Review of Current Strategies and Future Directions. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202300425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Indexed: 01/06/2025]
Abstract
AbstractTendon diseases commonly lead to physical disability, exerting a profound impact on the routine of affected patients. These conditions respond poorly to existing treatments, presenting a substantial challenge for orthopedic scientists. Research into clinical translational therapy has yet to yield highly versatile interventions capable of effectively addressing tendon diseases, including tendinopathy. Stem cell‐based therapies have emerged as a promising avenue for modifying the biological milieu through the secretion of regenerative and immunomodulatory factors. The current review provides an overview of the intricate tendon microenvironment, encompassing various tendon stem progenitor cells within distinct tendon sublocations, gene regulation, and pathways pertinent to tendon development, and the pathology of tendon diseases. Subsequently, the advantages of stem cell‐based therapies are discussed that utilize distinct types of autologous and allogeneic stem cells for tendon regeneration at the translational level. Moreover, this review outlines the challenges, gaps, and future innovations to propose a consolidated stem cell‐based therapy to treat tendinopathy. Finally, regenerative soluble therapies, insoluble bio‐active therapies, along with insoluble bio‐active therapies, and implantable 3D scaffolds for tendon tissue engineering are discussed, thereby presenting a pathway toward enhanced tissue regeneration and engineering.
Collapse
Affiliation(s)
- George Augustin
- Department of Anesthesiology and Pain Medicine Soonchunhyang University Bucheon Hospital Soonchunhyang University College of Medicine Bucheon‐Si 14584 Republic of Korea
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
- Department of Biochemistry and Biophysics Oregon State University Corvallis OR 92331 USA
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
- Department of Integrated Biomedical Science Soonchunhyang University Asan‐si, Chungnam‐Do 31538 Republic of Korea
| | - Min‐Kyu Kim
- Department of Anesthesiology and Pain Medicine Soonchunhyang University Bucheon Hospital Soonchunhyang University College of Medicine Bucheon‐Si 14584 Republic of Korea
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
| | - Sung Sik Hur
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
| | - Joon Ho Lee
- Department of Anesthesiology and Pain Medicine Soonchunhyang University Bucheon Hospital Soonchunhyang University College of Medicine Bucheon‐Si 14584 Republic of Korea
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
- Department of Integrated Biomedical Science Soonchunhyang University Asan‐si, Chungnam‐Do 31538 Republic of Korea
| |
Collapse
|
7
|
Moghassemi S, Dadashzadeh A, Sousa MJ, Vlieghe H, Yang J, León-Félix CM, Amorim CA. Extracellular vesicles in nanomedicine and regenerative medicine: A review over the last decade. Bioact Mater 2024; 36:126-156. [PMID: 38450204 PMCID: PMC10915394 DOI: 10.1016/j.bioactmat.2024.02.021] [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: 12/01/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Small extracellular vesicles (sEVs) are known to be secreted by a vast majority of cells. These sEVs, specifically exosomes, induce specific cell-to-cell interactions and can activate signaling pathways in recipient cells through fusion or interaction. These nanovesicles possess several desirable properties, making them ideal for regenerative medicine and nanomedicine applications. These properties include exceptional stability, biocompatibility, wide biodistribution, and minimal immunogenicity. However, the practical utilization of sEVs, particularly in clinical settings and at a large scale, is hindered by the expensive procedures required for their isolation, limited circulation lifetime, and suboptimal targeting capacity. Despite these challenges, sEVs have demonstrated a remarkable ability to accommodate various cargoes and have found extensive applications in the biomedical sciences. To overcome the limitations of sEVs and broaden their potential applications, researchers should strive to deepen their understanding of current isolation, loading, and characterization techniques. Additionally, acquiring fundamental knowledge about sEVs origins and employing state-of-the-art methodologies in nanomedicine and regenerative medicine can expand the sEVs research scope. This review provides a comprehensive overview of state-of-the-art exosome-based strategies in diverse nanomedicine domains, encompassing cancer therapy, immunotherapy, and biomarker applications. Furthermore, we emphasize the immense potential of exosomes in regenerative medicine.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Cecibel María León-Félix
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
8
|
Rahmati S, Khazaei M, Abpeikar Z, Soleimanizadeh A, Rezakhani L. Exosome-loaded decellularized tissue: Opening a new window for regenerative medicine. J Tissue Viability 2024; 33:332-344. [PMID: 38594147 DOI: 10.1016/j.jtv.2024.04.005] [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: 07/25/2023] [Revised: 03/29/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
Mesenchymal stem cell-derived exosomes (MSCs-EXO) have received a lot of interest recently as a potential therapeutic tool in regenerative medicine. Extracellular vesicles (EVs) known as exosomes (EXOs) are crucial for cell-cell communication throughout a variety of activities including stress response, aging, angiogenesis, and cell differentiation. Exploration of the potential use of EXOs as essential therapeutic effectors of MSCs to encourage tissue regeneration was motivated by success in the field of regenerative medicine. EXOs have been administered to target tissues using a variety of methods, including direct, intravenous, intraperitoneal injection, oral delivery, and hydrogel-based encapsulation, in various disease models. Despite the significant advances in EXO therapy, various methods are still being researched to optimize the therapeutic applications of these nanoparticles, and it is not completely clear which approach to EXO administration will have the greatest effects. Here, we will review emerging developments in the applications of EXOs loaded into decellularized tissues as therapeutic agents for use in regenerative medicine in various tissues.
Collapse
Affiliation(s)
- Shima Rahmati
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Abpeikar
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Arghavan Soleimanizadeh
- Faculty of Medicine, Graduate School 'Molecular Medicine, University of Ulm, 89081, Ulm, Germany
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
9
|
Das S, Lyon CJ, Hu T. A Panorama of Extracellular Vesicle Applications: From Biomarker Detection to Therapeutics. ACS NANO 2024; 18:9784-9797. [PMID: 38471757 PMCID: PMC11008359 DOI: 10.1021/acsnano.4c00666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Extracellular vesicles (EVs) secreted by all cell types are involved in the cell-to-cell transfer of regulatory factors that influence cell and tissue phenotypes in normal and diseased tissues. EVs are thus a rich source of biomarker targets for assays that analyze blood and urinary EVs for disease diagnosis. Sensitive biomarker detection in EVs derived from specific cell populations is a key major hurdle when analyzing complex biological samples, but innovative approaches surveyed in this Perspective can streamline EV isolation and enhance the sensitivity of EV detection procedures required for clinical application of EV-based diagnostics and therapeutics, including nanotechnology and microfluidics, to achieve EV characterizations. Finally, this Perspective also outlines opportunities and challenges remaining for clinical translation of EV-based assays.
Collapse
Affiliation(s)
- Sumita Das
- Center for Cellular and Molecular Diagnostics
and Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Christopher J. Lyon
- Center for Cellular and Molecular Diagnostics
and Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Tony Hu
- Center for Cellular and Molecular Diagnostics
and Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| |
Collapse
|
10
|
Rhim WK, Woo J, Kim JY, Lee EH, Cha SG, Kim DS, Baek SW, Park CG, Kim BS, Kwon TG, Han DK. Multiplexed PLGA scaffolds with nitric oxide-releasing zinc oxide and melatonin-modulated extracellular vesicles for severe chronic kidney disease. J Adv Res 2024:S2090-1232(24)00118-8. [PMID: 38537702 DOI: 10.1016/j.jare.2024.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION With prevalence of chronic kidney disease (CKD) in worldwide, the strategies to recover renal function via tissue regeneration could provide alternatives to kidney replacement therapies. However, due to relatively low reproducibility of renal basal cells and limited bioactivities of implanted biomaterials along with the high probability of substance-inducible inflammation and immunogenicity, kidney tissue regeneration could be challenging. OBJECTIVES To exclude various side effects from cell transplantations, in this study, we have induced extracellular vesicles (EVs) incorporated cell-free hybrid PMEZ scaffolds. METHODS Hybrid PMEZ scaffolds incorporating essential bioactive components, such as ricinoleic acid grafted Mg(OH)2 (M), extracellular matrix (E), and alpha lipoic acid-conjugated ZnO (Z) based on biodegradable porous PLGA (P) platform was successfully manufactured. Consecutively, for functional improvements, melatonin-modulated extracellular vesicles (mEVs), derived from the human umbilical cord MSCs in chemically defined media without serum impurities, were also loaded onto PMEZ scaffolds to construct the multiplexed PMEZ/mEV scaffold. RESULTS With functionalities of Mg(OH)2 and extracellular matrix-loaded PLGA scaffolds, the continuous nitric oxide-releasing property of modified ZnO and remarkably upregulated regenerative functionalities of mEVs showed significantly enhanced kidney regenerative activities. Based on these, the structural and functional restoration has been practically achieved in 5/6 nephrectomy mouse models that mimicked severe human CKD. CONCLUSION Our study has proved the combinatory bioactivities of the biodegradable PLGA-based multiplexed scaffold for kidney tissue regeneration in 5/6 nephrectomy mouse representing a severe CKD model. The optimal microenvironments for the morphogenetic formations of renal tissues and functional restorations have successfully achieved the combinatory bioactivities of remarkable components for PMEZ/mEV, which could be a promising therapeutic alternative for CKD treatment.
Collapse
Affiliation(s)
- Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jiwon Woo
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea; Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Eun Hye Lee
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea; Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Bum Soo Kim
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea; Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Tae Gyun Kwon
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea; Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea.
| |
Collapse
|
11
|
Xue K, Mi B. Engineered Extracellular Vesicles in Chronic Kidney Diseases: A Comprehensive Review. Int J Nanomedicine 2024; 19:2377-2393. [PMID: 38469058 PMCID: PMC10926925 DOI: 10.2147/ijn.s452393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/19/2024] [Indexed: 03/13/2024] Open
Abstract
Chronic kidney diseases (CKD) present a formidable global health challenge, characterized by a deficiency of effective treatment options. Extracellular vesicles (EVs), recognized as multifunctional drug delivery systems in biomedicine, have gained accumulative interest. Specifically, engineered EVs have emerged as a promising therapeutic approach for targeted drug delivery, potentially addressing the complexities of CKD management. In this review, we systematically dissect EVs, elucidating their classification, biogenesis, composition, and cargo molecules. Furthermore, we explore techniques for EV engineering and strategies for their precise renal delivery, focusing on cargo loading and transportation, providing a comprehensive perspective. Moreover, this review also discusses and summarizes the diverse therapeutic applications of engineered EVs in CKD, emphasizing their anti-inflammatory, immunomodulatory, renoprotective, and tissue-regenerating effects. It critically evaluates the challenges and limitations in translating EV therapies from laboratory settings to clinical applications, while outlining future prospects and emerging trends.
Collapse
Affiliation(s)
- Kaming Xue
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| |
Collapse
|
12
|
Park SY, Jung JH, Kim DS, Lee JK, Song BG, Shin HE, Jung JW, Baek SW, You S, Han I, Han DK. Therapeutic potential of luteolin-loaded poly(lactic-co-glycolic acid)/modified magnesium hydroxide microsphere in functional thermosensitive hydrogel for treating neuropathic pain. J Tissue Eng 2024; 15:20417314231226105. [PMID: 38333057 PMCID: PMC10851718 DOI: 10.1177/20417314231226105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 12/27/2023] [Indexed: 02/10/2024] Open
Abstract
Neuropathic pain (NP) is a debilitating condition stemming from damage to the somatosensory system frequently caused by nerve injuries or lesions. While existing treatments are widely employed, they often lead to side effects and lack specificity. This study aimed to alleviate NP by developing an innovative sustained-release thermosensitive hydrogel system. The system incorporates hyaluronic acid (HA)/Pluronic F127 injectable hydrogel and bupivacaine (Bup, B) in combination with poly(lactic-co-glycolic acid; PLGA)/modified magnesium hydroxide (MH)/luteolin (Lut; PML) microspheres (PML@B/Gel). The PML@B/Gel was designed for localized and prolonged co-delivery of Bup and Lut as an anesthetic and anti-inflammatory agent, respectively. Our studies demonstrated that PML@B/Gel had exceptional biocompatibility, anti-inflammatory, and antioxidant properties. In addition, it exhibited efficient pain relief in in vitro cellular assays. Moreover, this functional hydrogel showed substantial sustained drug release while diminishing microglial activation. Consequently, it effectively mitigated mechanical allodynia and thermal hyperalgesia in in vivo rat models of chronic constriction injury (CCI). Based on our research findings, PML@B/Gel emerges as a promising therapeutic approach for the protracted treatment of NP.
Collapse
Affiliation(s)
- So-Yeon Park
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, Korea
| | - Joon Hyuk Jung
- Department of Life Science, CHA University School of Medicine, Seongnam-si, Gyeonggi-do, Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, USA
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Byeong Gwan Song
- Department of Life Science, CHA University School of Medicine, Seongnam-si, Gyeonggi-do, Korea
| | - Hae Eun Shin
- Department of Life Science, CHA University School of Medicine, Seongnam-si, Gyeonggi-do, Korea
| | - Ji-Won Jung
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Seungkwon You
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| |
Collapse
|
13
|
Meng S, Hu H, Qiao Y, Wang F, Zhang BN, Sun D, Zhou L, Zhao L, Xie L, Zhang H, Zhou Q. A Versatile Hydrogel with Antibacterial and Sequential Drug-Releasing Capability for the Programmable Healing of Infectious Keratitis. ACS NANO 2023; 17:24055-24069. [PMID: 38044579 DOI: 10.1021/acsnano.3c09034] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Hydrogels have attracted tremendous attention as favorable corneal substitutes for treating severe infectious keratitis (IK). However, current hydrogel-based corneal substitutes were majorly designed to promote the single stage of corneal regeneration, which falls short in meeting the clinical management needs of severe IK including the multiple phases of corneal wound healing. Herein, we introduce a versatile hybrid hydrogel (SQPV) composed of silk fibroin and chitosan, which exhibits spatiotemporal properties for drug release. The SQPV is fabricated by incorporating verteporfin-loaded poly(lactic-co-glycolic)-polyethylene glycol-o-nitrobenzene micelles into a hydrogel network, which is formed from methacrylate silk fibroin and glycidyl methacrylate functionalized quaternized chitosan containing polydeoxyribonucleotide. This double network approach results in a material with exceptional anti-inflammatory, antibacterial, and proliferative stimulation and tissue remodeling regulation capabilities. Furthermore, SQPV showcases mechanical strength and transparency akin to those of native cornea. Extensive in vitro and in vivo studies validate SQPV's ability to effectively eliminate residual bacteria, mitigate inflammation, foster regeneration of corneal epithelium and stroma, prevent corneal scarring, and ultimately expedite wound healing. In summary, the SF/CS-based hybrid hydrogel may represent a promising substitute for comprehensive corneal repair and regeneration in severe IK.
Collapse
Affiliation(s)
- Shuqin Meng
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Hao Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, P.R. China
| | - Yujie Qiao
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Fuyan Wang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Bi Ning Zhang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Difang Sun
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Longfang Zhou
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Long Zhao
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Lixin Xie
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Hengrui Zhang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| |
Collapse
|
14
|
Rhim WK, Kim JY, Lee SY, Cha SG, Park JM, Park HJ, Park CG, Han DK. Recent advances in extracellular vesicle engineering and its applications to regenerative medicine. Biomater Res 2023; 27:130. [PMID: 38082304 PMCID: PMC10712135 DOI: 10.1186/s40824-023-00468-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 11/24/2023] [Indexed: 01/02/2025] Open
Abstract
Extracellular vesicles (EVs) are nanosized particles that are released from cells and reflect the characteristics of the mother cell. Recently, the EVs have been used in several types of studies across many different fields. In the field of EV research, multiple cell culture and EV isolation techniques have been highlighted in importance. Various strategies, including exclusive component culture media, three-dimensional (3D) cultures, and hypoxic conditions, have been proposed for the cell culture to control function of the EVs. Ultracentrifugation, ultrafiltration, precipitation, and tangential flow filtration (TFF) have been utilized for EV isolation. Although isolated EVs have their own functionalities, several researchers are trying to functionalize EVs by applying various engineering approaches. Gene editing, exogenous, endogenous, and hybridization methods are the four well-known types of EV functionalization strategies. EV engineered through these processes has been applied in the field of regenerative medicine, including kidney diseases, osteoarthritis, rheumatoid arthritis, nervous system-related diseases, and others. In this review, it was focused on engineering approaches for EV functionalization and their applications in regenerative medicine.
Collapse
Affiliation(s)
| | - Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
- Department of Biomedical Engineering, 2066 Seobu-ro Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Seung Yeon Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Jeong Min Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hyeon Jeong Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, 2066 Seobu-ro Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
15
|
Cha SG, Rhim WK, Kim JY, Lee EH, Lee SY, Park JM, Lee JE, Yoon H, Park CG, Kim BS, Kwon TG, Lee Y, Lee DR, Han DK. Kidney tissue regeneration using bioactive scaffolds incorporated with differentiating extracellular vesicles and intermediate mesoderm cells. Biomater Res 2023; 27:126. [PMID: 38049879 PMCID: PMC10696796 DOI: 10.1186/s40824-023-00471-x] [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: 10/06/2023] [Accepted: 11/24/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND To overcome the limitations of current alternative therapies for chronic kidney disease (CKD), tissue engineering-mediated regeneration strategies have demonstrated the possibilities for complete kidney tissue regeneration. Given the challenges associated with the reproducibility of renal basal cells, the incorporation of intermediate mesoderm (IM) cells and bioactive materials to control bioactivities of cells with supported scaffolds should be considered as a viable approach to enable the regeneration of the complex kidney structure via renal differentiation. METHODS We developed PMEZ scaffolds by combining crucial bioactive components, such as ricinoleic acid-grafted Mg(OH)2 (M), extracellular matrix (E), and alpha lipoic acid-conjugated ZnO (Z) integrated into biodegradable porous PLGA (P) platform. Additionally, we utilized differentiating extracellular vesicles (dEV) isolated during intermediate mesoderm differentiation into kidney progenitor cells, and IM cells were serially incorporated to facilitate kidney tissue regeneration through their differentiation into kidney progenitor cells in the 3/4 nephrectomy mouse model. RESULTS The use of differentiating extracellular vesicles facilitated IM differentiation into kidney progenitor cells without additional differentiation factors. This led to improvements in various regeneration-related bioactivities including tubule and podocyte regeneration, anti-fibrosis, angiogenesis, and anti-inflammation. Finally, implanting PMEZ/dEV/IM scaffolds in mouse injury model resulted in the restoration of kidney function. CONCLUSIONS Our study has demonstrated that utilizing biodegradable PLGA-based scaffolds, which include multipotent cells capable of differentiating into various kidney progenitor cells along with supporting components, can facilitate kidney tissue regeneration in the mouse model that simulates CKD through 3/4 nephrectomy.
Collapse
Affiliation(s)
- Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea
| | - Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea
| | - Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Eun Hye Lee
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu, 41944, Republic of Korea
| | - Seung Yeon Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea
| | - Jeong Min Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea
| | - Jeoung Eun Lee
- Bundang Medical Center, CHA Advanced Research Institute, CHA University, Sungnam- si, 13488, Gyeonggi-do, Republic of Korea
| | - Hyeji Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seodaemun-gu, Seoul, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Bum Soo Kim
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu, 41944, Republic of Korea
| | - Tae Gyun Kwon
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu, 41944, Republic of Korea
| | - Youngmi Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, Seodaemun-gu, Seoul, Republic of Korea
| | - Dong Ryul Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea
- Bundang Medical Center, CHA Advanced Research Institute, CHA University, Sungnam- si, 13488, Gyeonggi-do, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
16
|
Cho YW, Park JH, Kang MJ, Lee JH, Kim YK, Luo Z, Kim TH. Electrochemical Detection of Dopamine Release from Living Neurons Using Graphene Oxide-Incorporated Polypyrrole/Gold Nanocluster Hybrid Nanopattern Arrays. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2304271. [PMID: 37649209 DOI: 10.1002/smll.202304271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/17/2023] [Indexed: 09/01/2023]
Abstract
Stem-cell-based therapeutics have shown immense potential in treating various diseases that are currently incurable. In particular, partial recovery of Parkinson's disease, which occurs due to massive loss or abnormal functionality of dopaminergic (DAnergic) neurons, through the engraftment of stem-cell-derived neurons ex vivo is reported. However, precise assessment of the functionality and maturity of DAnergic neurons is still challenging for their enhanced clinical efficacy. Here, a novel conductive cell cultivation platform, a graphene oxide (GO)-incorporated metallic polymer nanopillar array (GOMPON), that can electrochemically detect dopamine (DA) exocytosis from living DAnergic neurons, is reported. In the cell-free configuration, the linear range is 0.5-100 µm, with a limit of detection of 33.4 nm. Owing to its excellent biocompatibility, a model DAnergic neuron (SH-SY5Y cell) can be cultivated and differentiated on the platform while their DA release can be quantitatively measured in a real-time and nondestructive manner. Finally, it is showed that the functionality of the DAnergic neurons derived from stem cells can be precisely assessed via electrochemical detection of their DA exocytosis. The developed GOMPON is highly promising for a wide range of applications, including real-time monitoring of stem cell differentiation into neuronal lineages, evaluating differentiation protocols, and finding practical stem cell therapies.
Collapse
Affiliation(s)
- Yeon-Woo Cho
- School of Integrative Engineering, Chung-Ang University, 06974, Seoul, Dongjak-gu, 84 Heukseuk-ro, Republic of Korea
| | - Joon-Ha Park
- School of Integrative Engineering, Chung-Ang University, 06974, Seoul, Dongjak-gu, 84 Heukseuk-ro, Republic of Korea
| | - Min-Ji Kang
- School of Integrative Engineering, Chung-Ang University, 06974, Seoul, Dongjak-gu, 84 Heukseuk-ro, Republic of Korea
| | - Jung-Hyeon Lee
- School of Integrative Engineering, Chung-Ang University, 06974, Seoul, Dongjak-gu, 84 Heukseuk-ro, Republic of Korea
| | - Yong Kyun Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, St. Vincent's Hospital, Suwon, 16247, Republic of Korea
| | - Zhengtang Luo
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, 999077, Hong Kong, Kowloon, Clear Water Bay, China
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 06974, Seoul, Dongjak-gu, 84 Heukseuk-ro, Republic of Korea
| |
Collapse
|
17
|
Simon L, Lapinte V, Morille M. Exploring the role of polymers to overcome ongoing challenges in the field of extracellular vesicles. J Extracell Vesicles 2023; 12:e12386. [PMID: 38050832 PMCID: PMC10696644 DOI: 10.1002/jev2.12386] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 12/07/2023] Open
Abstract
Extracellular vesicles (EVs) are naturally occurring nanoparticles released from all eucaryotic and procaryotic cells. While their role was formerly largely underestimated, EVs are now clearly established as key mediators of intercellular communication. Therefore, these vesicles constitute an attractive topic of study for both basic and applied research with great potential, for example, as a new class of biomarkers, as cell-free therapeutics or as drug delivery systems. However, the complexity and biological origin of EVs sometimes complicate their identification and therapeutic use. Thus, this rapidly expanding research field requires new methods and tools for the production, enrichment, detection, and therapeutic application of EVs. In this review, we have sought to explain how polymer materials actively contributed to overcome some of the limitations associated to EVs. Indeed, thanks to their infinite diversity of composition and properties, polymers can act through a variety of strategies and at different stages of EVs development. Overall, we would like to emphasize the importance of multidisciplinary research involving polymers to address persistent limitations in the field of EVs.
Collapse
Affiliation(s)
| | | | - Marie Morille
- ICGM, Univ Montpellier, CNRS, ENSCMMontpellierFrance
- Institut universitaire de France (IUF)ParisFrance
| |
Collapse
|
18
|
Xu P, Kankala RK, Wang S, Chen A. Decellularized extracellular matrix-based composite scaffolds for tissue engineering and regenerative medicine. Regen Biomater 2023; 11:rbad107. [PMID: 38173774 PMCID: PMC10761212 DOI: 10.1093/rb/rbad107] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/17/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Despite the considerable advancements in fabricating polymeric-based scaffolds for tissue engineering, the clinical transformation of these scaffolds remained a big challenge because of the difficulty of simulating native organs/tissues' microenvironment. As a kind of natural tissue-derived biomaterials, decellularized extracellular matrix (dECM)-based scaffolds have gained attention due to their unique biomimetic properties, providing a specific microenvironment suitable for promoting cell proliferation, migration, attachment and regulating differentiation. The medical applications of dECM-based scaffolds have addressed critical challenges, including poor mechanical strength and insufficient stability. For promoting the reconstruction of damaged tissues or organs, different types of dECM-based composite platforms have been designed to mimic tissue microenvironment, including by integrating with natural polymer or/and syntenic polymer or adding bioactive factors. In this review, we summarized the research progress of dECM-based composite scaffolds in regenerative medicine, highlighting the critical challenges and future perspectives related to the medical application of these composite materials.
Collapse
Affiliation(s)
- Peiyao Xu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| | - Shibin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| |
Collapse
|
19
|
Harun-Or-Rashid M, Aktar MN, Hossain MS, Sarkar N, Islam MR, Arafat ME, Bhowmik S, Yusa SI. Recent Advances in Micro- and Nano-Drug Delivery Systems Based on Natural and Synthetic Biomaterials. Polymers (Basel) 2023; 15:4563. [PMID: 38231996 PMCID: PMC10708661 DOI: 10.3390/polym15234563] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 01/19/2024] Open
Abstract
Polymeric drug delivery technology, which allows for medicinal ingredients to enter a cell more easily, has advanced considerably in recent decades. Innovative medication delivery strategies use biodegradable and bio-reducible polymers, and progress in the field has been accelerated by future possible research applications. Natural polymers utilized in polymeric drug delivery systems include arginine, chitosan, dextrin, polysaccharides, poly(glycolic acid), poly(lactic acid), and hyaluronic acid. Additionally, poly(2-hydroxyethyl methacrylate), poly(N-isopropyl acrylamide), poly(ethylenimine), dendritic polymers, biodegradable polymers, and bioabsorbable polymers as well as biomimetic and bio-related polymeric systems and drug-free macromolecular therapies have been employed in polymeric drug delivery. Different synthetic and natural biomaterials are in the clinical phase to mitigate different diseases. Drug delivery methods using natural and synthetic polymers are becoming increasingly common in the pharmaceutical industry, with biocompatible and bio-related copolymers and dendrimers having helped cure cancer as drug delivery systems. This review discusses all the above components and how, by combining synthetic and biological approaches, micro- and nano-drug delivery systems can result in revolutionary polymeric drug and gene delivery devices.
Collapse
Affiliation(s)
- Md. Harun-Or-Rashid
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji 671-2280, Hyogo, Japan; (M.H.-O.-R.); (M.N.A.); (S.B.)
| | - Most. Nazmin Aktar
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji 671-2280, Hyogo, Japan; (M.H.-O.-R.); (M.N.A.); (S.B.)
| | - Md. Sabbir Hossain
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.S.H.); (N.S.); (M.R.I.); (M.E.A.)
| | - Nadia Sarkar
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.S.H.); (N.S.); (M.R.I.); (M.E.A.)
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.S.H.); (N.S.); (M.R.I.); (M.E.A.)
| | - Md. Easin Arafat
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.S.H.); (N.S.); (M.R.I.); (M.E.A.)
| | - Shukanta Bhowmik
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji 671-2280, Hyogo, Japan; (M.H.-O.-R.); (M.N.A.); (S.B.)
| | - Shin-ichi Yusa
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji 671-2280, Hyogo, Japan; (M.H.-O.-R.); (M.N.A.); (S.B.)
| |
Collapse
|
20
|
Deng S, Cao H, Cui X, Fan Y, Wang Q, Zhang X. Optimization of exosome-based cell-free strategies to enhance endogenous cell functions in tissue regeneration. Acta Biomater 2023; 171:68-84. [PMID: 37730080 DOI: 10.1016/j.actbio.2023.09.023] [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: 06/27/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Exosomes, nanoscale extracellular vesicles, play a crucial role in intercellular communication, owing to their biologically active cargoes such as RNAs and proteins. In recent years, they have emerged as a promising tool in the field of tissue regeneration, with the potential to initiate a new trend in cell-free therapy. However, it's worth noting that not all types of exosomes derived from cells are appropriate for tissue repair. Thus, selecting suitable cell sources is critical to ensure their efficacy in specific tissue regeneration processes. Current therapeutic applications of exosomes also encounter several limitations, including low-specific content for targeted diseases, non-tissue-specific targeting, and short retention time due to rapid clearance in vivo. Consequently, this review paper focuses on exosomes from diverse cell sources with functions specific to tissue regeneration. It also highlights the latest engineering strategies developed to overcome the functional limitations of natural exosomes. These strategies encompass the loading of specific therapeutic contents into exosomes, the endowment of tissue-specific targeting capability on the exosome surface, and the incorporation of biomaterials to extend the in vivo retention time of exosomes in a controlled-release manner. Collectively, these innovative approaches aim to synergistically enhance the therapeutic effects of natural exosomes, optimizing exosome-based cell-free strategies to boost endogenous cell functions in tissue regeneration. STATEMENT OF SIGNIFICANCE: Exosome-based cell-free therapy has recently emerged as a promising tool for tissue regeneration. This review highlights the characteristics and functions of exosomes from different sources that can facilitate tissue repair and their contributions to the regeneration process. To address the functional limitations of natural exosomes in therapeutic applications, this review provides an in-depth understanding of the latest engineering strategies. These strategies include optimizing exosomal contents, endowing tissue-specific targeting capability on the exosome surface, and incorporating biomaterials to extend the in vivo retention time of exosomes in a controlled-release manner. This review aims to explore and discuss innovative approaches that can synergistically improve endogenous cell functions in advanced exosome-based cell-free therapies for a broad range of tissue regeneration.
Collapse
Affiliation(s)
- Siyan Deng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Hongfu Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiaolin Cui
- School of medicine, the Chinese University of Hong Kong, Shenzhen, China; Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
21
|
Ibi Y, Nishinakamura R. Kidney Bioengineering for Transplantation. Transplantation 2023; 107:1883-1894. [PMID: 36717963 DOI: 10.1097/tp.0000000000004526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The kidney is an important organ for maintenance of homeostasis in the human body. As renal failure progresses, renal replacement therapy becomes necessary. However, there is a chronic shortage of kidney donors, creating a major problem for transplantation. To solve this problem, many strategies for the generation of transplantable kidneys are under investigation. Since the first reports describing that nephron progenitors could be induced from human induced pluripotent stem cells, kidney organoids have been attracting attention as tools for studying human kidney development and diseases. Because the kidney is formed through the interactions of multiple renal progenitors, current studies are investigating ways to combine these progenitors derived from human induced pluripotent stem cells for the generation of transplantable kidney organoids. Other bioengineering strategies, such as decellularization and recellularization of scaffolds, 3-dimensional bioprinting, interspecies blastocyst complementation and progenitor replacement, and xenotransplantation, also have the potential to generate whole kidneys, although each of these strategies has its own challenges. Combinations of these approaches will lead to the generation of bioengineered kidneys that are transplantable into humans.
Collapse
Affiliation(s)
- Yutaro Ibi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
22
|
Wang L, Qi C, Cao H, Zhang Y, Liu X, Qiu L, Wang H, Xu L, Wu Z, Liu J, Wang S, Kong D, Wang Y. Engineered Cytokine-Primed Extracellular Vesicles with High PD-L1 Expression Ameliorate Type 1 Diabetes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301019. [PMID: 37209021 DOI: 10.1002/smll.202301019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/23/2023] [Indexed: 05/21/2023]
Abstract
Type 1 diabetes (T1D), which is a chronic autoimmune disease, results from the destruction of insulin-producing β cells targeted by autoreactive T cells. The recent discovery that mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) function as therapeutic tools for autoimmune conditions has attracted substantial attention. However, the in vivo distribution and therapeutic effects of MSC-EVs potentiated by pro-inflammatory cytokines in the context of T1D have yet to be established. Here, it is reported that hexyl 5-aminolevulinate hydrochloride (HAL)-loaded engineered cytokine-primed MSC-EVs (H@TI-EVs) with high expression of immune checkpoint molecule programmed death-legend 1 (PD-L1) exert excellent inflammatory targeting and immunosuppressive effects for T1D imaging and therapy. The accumulated H@TI-EVs in injured pancreas not only enabled the fluorescence imaging and tracking of TI-EVs through the intermediate product protoporphyrin (PpIX) generated by HAL, but also promoted the proliferative and anti-apoptotic effects of islet β cells. Further analysis revealed that H@TI-EVs exhibited an impressive ability to reduce CD4+ T cell density and activation through the PD-L1/PD-1 axis, and induced M1-to-M2 macrophage transition to reshape the immune microenvironment, exhibiting high therapeutic efficiency in mice with T1D. This work identifies a novel strategy for the imaging and treatment of T1D with great potential for clinical application.
Collapse
Affiliation(s)
- Lanxing Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chunxiao Qi
- Department of Pharmacology, Tianjin Medical University, Tianjin, 300070, China
| | - Hongmei Cao
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Yanwen Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xing Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Lina Qiu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Hang Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Lijuan Xu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhenzhou Wu
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Shusen Wang
- Institute of Transplant Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yuebing Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Surgical Intensive Care Unit, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, China
| |
Collapse
|
23
|
Roh EJ, Kim DS, Kim JH, Lim CS, Choi H, Kwon SY, Park SY, Kim JY, Kim HM, Hwang DY, Han DK, Han I. Multimodal therapy strategy based on a bioactive hydrogel for repair of spinal cord injury. Biomaterials 2023; 299:122160. [PMID: 37209541 DOI: 10.1016/j.biomaterials.2023.122160] [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: 11/03/2022] [Revised: 03/13/2023] [Accepted: 05/10/2023] [Indexed: 05/22/2023]
Abstract
Traumatic spinal cord injury results in permanent and serious neurological impairment, but there is no effective treatment yet. Tissue engineering approaches offer great potential for the treatment of SCI, but spinal cord complexity poses great challenges. In this study, the composite scaffold consists of a hyaluronic acid-based hydrogel, decellularized brain matrix (DBM), and bioactive compounds such as polydeoxyribonucleotide (PDRN), tumor necrosis factor-α/interferon-γ primed mesenchymal stem cell-derived extracellular vesicles (TI-EVs), and human embryonic stem cell-derived neural progenitor cells (NPC). The composite scaffold showed significant effects on regenerative prosses including angiogenesis, anti-inflammation, anti-apoptosis, and neural differentiation. In addition, the composite scaffold (DBM/PDRN/TI-EV/NPC@Gel) induced an effective spinal cord regeneration in a rat spinal cord transection model. Therefore, this multimodal approach using an integrated bioactive scaffold coupled with biochemical cues from PDRN and TI-EVs could be used as an advanced tissue engineering platform for spinal cord regeneration.
Collapse
Affiliation(s)
- Eun Ji Roh
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea; Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea; School of Integrative Engineering Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Chang Su Lim
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Su Yeon Kwon
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - So-Yeon Park
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea; Division of Biotechnology College of Life Sciences and Biotechnology Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jun Yong Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Hyun-Mun Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Dong-Youn Hwang
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea.
| | - Inbo Han
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea.
| |
Collapse
|
24
|
Zhang Y, Yu L, Qiu R, Cao L, Ye G, Lin R, Wang Y, Wang G, Hu B, Hou H. 3D hypoxia-mimicking and anti-synechia hydrogel enabling promoted neovascularization for renal injury repair and regeneration. Mater Today Bio 2023; 21:100694. [PMID: 37346780 PMCID: PMC10279555 DOI: 10.1016/j.mtbio.2023.100694] [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: 02/15/2023] [Revised: 05/02/2023] [Accepted: 06/06/2023] [Indexed: 06/23/2023] Open
Abstract
In-situ renal tissue engineering is promising yet challenging for renal injury repair and regeneration due to the highly vascularized structure of renal tissue and complex high-oxidative stress and ischemic microenvironment. Herein, a novel biocompatible 3D porous hydrogel (DFO-gel) with sustained release capacity of hypoxia mimicking micromolecule drug deferoxamine (DFO) was developed for in-situ renal injury repair. In vitro and in vivo experimental results demonstrated that the developed DFO-gels can exert the synchronous benefit of scavenging excess reactive oxygen species (ROS) regulating inflammatory microenvironment and promoting angiogenesis for effective renal injury repair by up-regulating hypoxia-inducible factor-1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF). The in-situ neogenesis of neonatal glomerular- and tubular-like structures in the implanted areas in the partially nephrectomized rats also suggested the potential for promoting renal injury repair and regeneration. This multifunctional hydrogel can not only exhibit the sustained release and promoted bio-uptake capacity for DFO, but also improve the renal injured microenvironment by alleviating oxidative and inflammatory stress, accelerating neovascularization, and promoting efficient anti-synechia. We believe this work offers a promising strategy for renal injury repair and regeneration.
Collapse
Affiliation(s)
- Yuehang Zhang
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Division of Nephrology, The Second Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Lei Yu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Renjie Qiu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Lisha Cao
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Genlan Ye
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Rurong Lin
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Yongqin Wang
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Guobao Wang
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Bianxiang Hu
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| |
Collapse
|
25
|
Du S, Guan Y, Xie A, Yan Z, Gao S, Li W, Rao L, Chen X, Chen T. Extracellular vesicles: a rising star for therapeutics and drug delivery. J Nanobiotechnology 2023; 21:231. [PMID: 37475025 PMCID: PMC10360328 DOI: 10.1186/s12951-023-01973-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023] Open
Abstract
Extracellular vesicles (EVs) are nano-sized, natural, cell-derived vesicles that contain the same nucleic acids, proteins, and lipids as their source cells. Thus, they can serve as natural carriers for therapeutic agents and drugs, and have many advantages over conventional nanocarriers, including their low immunogenicity, good biocompatibility, natural blood-brain barrier penetration, and capacity for gene delivery. This review first introduces the classification of EVs and then discusses several currently popular methods for isolating and purifying EVs, EVs-mediated drug delivery, and the functionalization of EVs as carriers. Thereby, it provides new avenues for the development of EVs-based therapeutic strategies in different fields of medicine. Finally, it highlights some challenges and future perspectives with regard to the clinical application of EVs.
Collapse
Affiliation(s)
- Shuang Du
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Yucheng Guan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Aihua Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Zhao Yan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Sijia Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Room 6007, N22, Taipa, 999078, Macau SAR, China
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Room 6007, N22, Taipa, 999078, Macau SAR, China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China.
| |
Collapse
|
26
|
Zhu Y, Liao ZF, Mo MH, Xiong XD. Mesenchymal Stromal Cell-Derived Extracellular Vesicles for Vasculopathies and Angiogenesis: Therapeutic Applications and Optimization. Biomolecules 2023; 13:1109. [PMID: 37509145 PMCID: PMC10377109 DOI: 10.3390/biom13071109] [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: 06/08/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Extracellular vesicles (EVs), as part of the cellular secretome, have emerged as essential cell-cell communication regulators in multiple physiological and pathological processes. Previous studies have widely reported that mesenchymal stromal cell-derived EVs (MSC-EVs) have potential therapeutic applications in ischemic diseases or regenerative medicine by accelerating angiogenesis. MSC-EVs also exert beneficial effects on other vasculopathies, including atherosclerosis, aneurysm, vascular restenosis, vascular calcification, vascular leakage, pulmonary hypertension, and diabetic retinopathy. Consequently, the potential of MSC-EVs in regulating vascular homeostasis is attracting increasing interest. In addition to native or naked MSC-EVs, modified MSC-EVs and appropriate biomaterials for delivering MSC-EVs can be introduced to this area to further promote their therapeutic applications. Herein, we outline the functional roles of MSC-EVs in different vasculopathies and angiogenesis to elucidate how MSC-EVs contribute to maintaining vascular system homeostasis. We also discuss the current strategies to optimize their therapeutic effects, which depend on the superior bioactivity, high yield, efficient delivery, and controlled release of MSC-EVs to the desired regions, as well as the challenges that need to be overcome to allow their broad clinical translation.
Collapse
Affiliation(s)
- Ying Zhu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Zhao-Fu Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Miao-Hua Mo
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Xing-Dong Xiong
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
27
|
Kim N, Lee H, Han G, Kang M, Park S, Kim DE, Lee M, Kim M, Na Y, Oh S, Bang S, Jang T, Kim H, Park J, Shin SR, Jung H. 3D-Printed Functional Hydrogel by DNA-Induced Biomineralization for Accelerated Diabetic Wound Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300816. [PMID: 37076933 PMCID: PMC10265106 DOI: 10.1002/advs.202300816] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/26/2023] [Indexed: 05/03/2023]
Abstract
Chronic wounds in diabetic patients are challenging because their prolonged inflammation makes healing difficult, thus burdening patients, society, and health care systems. Customized dressing materials are needed to effectively treat such wounds that vary in shape and depth. The continuous development of 3D-printing technology along with artificial intelligence has increased the precision, versatility, and compatibility of various materials, thus providing the considerable potential to meet the abovementioned needs. Herein, functional 3D-printing inks comprising DNA from salmon sperm and DNA-induced biosilica inspired by marine sponges, are developed for the machine learning-based 3D-printing of wound dressings. The DNA and biomineralized silica are incorporated into hydrogel inks in a fast, facile manner. The 3D-printed wound dressing thus generates provided appropriate porosity, characterized by effective exudate and blood absorption at wound sites, and mechanical tunability indicated by good shape fidelity and printability during optimized 3D printing. Moreover, the DNA and biomineralized silica act as nanotherapeutics, enhancing the biological activity of the dressings in terms of reactive oxygen species scavenging, angiogenesis, and anti-inflammation activity, thereby accelerating acute and diabetic wound healing. These bioinspired 3D-printed hydrogels produce using a DNA-induced biomineralization strategy are an excellent functional platform for clinical applications in acute and chronic wound repair.
Collapse
Affiliation(s)
- Nahyun Kim
- Department of Biomedical‐Chemical EngineeringThe Catholic University of KoreaBucheon14662Republic of Korea
- Department of BiotechnologyThe Catholic University of KoreaBucheon14662Republic of Korea
| | - Hyun Lee
- Department of Biomedical‐Chemical EngineeringThe Catholic University of KoreaBucheon14662Republic of Korea
- Department of BiotechnologyThe Catholic University of KoreaBucheon14662Republic of Korea
| | - Ginam Han
- Department of Biomedical‐Chemical EngineeringThe Catholic University of KoreaBucheon14662Republic of Korea
- Department of BiotechnologyThe Catholic University of KoreaBucheon14662Republic of Korea
| | - Minho Kang
- Department of Biomedical‐Chemical EngineeringThe Catholic University of KoreaBucheon14662Republic of Korea
- Department of BiotechnologyThe Catholic University of KoreaBucheon14662Republic of Korea
| | - Sinwoo Park
- Department of Biomedical‐Chemical EngineeringThe Catholic University of KoreaBucheon14662Republic of Korea
- Department of BiotechnologyThe Catholic University of KoreaBucheon14662Republic of Korea
| | - Dong Eung Kim
- Research Institute of Advanced Manufacturing & Materials TechnologyKorea Institute of Industrial TechnologyIncheon21999Republic of Korea
| | - Minyoung Lee
- School of Chemical and Biological Engineeringand Institute of Chemical Processes (ICP)Seoul National UniversitySeoul08826Republic of Korea
- Center for Nanoparticle ResearchInstitute of Basic Science (IBS)Seoul08826Republic of Korea
| | - Moon‐Jo Kim
- Research Institute of Advanced Manufacturing & Materials TechnologyKorea Institute of Industrial TechnologyIncheon21999Republic of Korea
| | - Yuhyun Na
- Department of Biomedical‐Chemical EngineeringThe Catholic University of KoreaBucheon14662Republic of Korea
- Department of BiotechnologyThe Catholic University of KoreaBucheon14662Republic of Korea
| | - SeKwon Oh
- Research Institute of Advanced Manufacturing & Materials TechnologyKorea Institute of Industrial TechnologyIncheon21999Republic of Korea
| | - Seo‐Jun Bang
- Department of Biomedical‐Chemical EngineeringThe Catholic University of KoreaBucheon14662Republic of Korea
- Department of BiotechnologyThe Catholic University of KoreaBucheon14662Republic of Korea
| | - Tae‐Sik Jang
- Department of Materials Science and EngineeringChosun UniversityGwangju61452Republic of Korea
| | - Hyoun‐Ee Kim
- Department of Materials Science and EngineeringSeoul National UniversitySeoul08826Republic of Korea
| | - Jungwon Park
- School of Chemical and Biological Engineeringand Institute of Chemical Processes (ICP)Seoul National UniversitySeoul08826Republic of Korea
- Center for Nanoparticle ResearchInstitute of Basic Science (IBS)Seoul08826Republic of Korea
| | - Su Ryon Shin
- Division of Engineering in MedicineDepartment of MedicineHarvard Medical Schooland Brigham and Women's HospitalCambridgeMA02139USA
| | - Hyun‐Do Jung
- Department of Biomedical‐Chemical EngineeringThe Catholic University of KoreaBucheon14662Republic of Korea
- Department of BiotechnologyThe Catholic University of KoreaBucheon14662Republic of Korea
| |
Collapse
|
28
|
Lu Y, Zhao L, Mao J, Liu W, Ma W, Zhao B. Rab27a-mediated extracellular vesicle secretion contributes to osteogenesis in periodontal ligament-bone niche communication. Sci Rep 2023; 13:8479. [PMID: 37231020 DOI: 10.1038/s41598-023-35172-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/14/2023] [Indexed: 05/27/2023] Open
Abstract
Periodontitis, an infectious and common disease worldwide, leads to the destruction of the periodontal ligament-alveolar bone complex. Within the bone metabolic niche, communication between periodontal ligament stem cells (PDLSCs) and bone marrow mesenchymal stem cells (BMMSCs) has been considered a major contributor to osteogenesis. PDLSC-derived extracellular vesicles (P-EVs) have shown great potential for bone regeneration. However, the secretion and uptake mechanisms of P-EVs remain elusive. Herein, the biogenesis of extracellular vesicles (EVs) from PDLSCs was observed using scanning and transmission electron microscopy. PDLSCs were transduced with Ras-associated protein 27a (Rab27a) siRNA (PDLSCsiRab27a) to inhibit EV secretion. The effect of P-EVs on BMMSCs was evaluated using a non-contact transwell co-culture system. We observed that Rab27a knockdown decreased EV secretion, and PDLSCsiRab27a remarkably attenuated co-culture-enhanced osteogenesis of BMMSCs. Isolated PDLSC-derived EVs enhanced osteogenic differentiation of BMMSCs in vitro and induced bone regeneration in a calvarial defect model in vivo. PDLSC-derived EVs were rapidly endocytosed by BMMSCs via the lipid raft/cholesterol endocytosis pathway and triggered the phosphorylation of extracellular signal-regulated kinase 1/2. In conclusion, PDLSCs contribute to the osteogenesis of BMMSCs through Rab27a-mediated EV secretion, thereby providing a potential cell-free approach for bone regeneration.
Collapse
Affiliation(s)
- Yun Lu
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Liru Zhao
- Department of Orthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jiaqi Mao
- Department of Orthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Wen Liu
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Wensheng Ma
- Department of Orthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Bingjiao Zhao
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China.
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China.
| |
Collapse
|
29
|
Kim JY, Rhim WK, Cha SG, Woo J, Lee JY, Park CG, Han DK. Bolstering the secretion and bioactivities of umbilical cord MSC-derived extracellular vesicles with 3D culture and priming in chemically defined media. NANO CONVERGENCE 2022; 9:57. [PMID: 36534191 PMCID: PMC9761620 DOI: 10.1186/s40580-022-00349-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/01/2022] [Indexed: 06/12/2023]
Abstract
Human mesenchymal stem cells (hMSCs)-derived extracellular vesicles (EVs) have been known to possess the features of the origin cell with nano size and have shown therapeutic potentials for regenerative medicine in recent studies as alternatives for cell-based therapies. However, extremely low production yield, unknown effects derived from serum impurities, and relatively low bioactivities on doses must be overcome for translational applications. As several reports have demonstrated the tunability of secretion and bioactivities of EVs, herein, we introduced three-dimensional (3D) culture and cell priming approaches for MSCs in serum-free chemically defined media to exclude side effects from serum-derived impurities. Aggregates (spheroids) with 3D culture dramatically enhanced secretion of EVs about 6.7 times more than cells with two-dimensional (2D) culture, and altered surface compositions. Further modulation with cell priming with the combination of TNF-α and IFN-γ (TI) facilitated the production of EVs about 1.4 times more than cells without priming (9.4 times more than cells with 2D culture without priming), and bioactivities of EVs related to tissue regenerations. Interestingly, unlike changing 2D to 3D culture, TI priming altered internal cytokines of MSC-derived EVs. Through simulating characteristics of EVs with bioinformatics analysis, the regeneration-relative properties such as angiogenesis, wound healing, anti-inflammation, anti-apoptosis, and anti-fibrosis, for three different types of EVs were comparatively analyzed using cell-based assays. The present study demonstrated that a combinatory strategy, 3D cultures and priming MSCs in chemically defined media, provided the optimum environments to maximize secretion and regeneration-related bioactivities of MSC-derived EVs without impurities for future translational applications.
Collapse
Affiliation(s)
- Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Jiwon Woo
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Joo Youn Lee
- Xcell Therapeutics, 333, Yeongdong-daero, Gangnam-gu, Seoul, 06188, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
30
|
Lacueva-Aparicio A, Lindoso RS, Mihăilă SM, Giménez I. Role of extracellular matrix components and structure in new renal models in vitro. Front Physiol 2022; 13:1048738. [PMID: 36569770 PMCID: PMC9767975 DOI: 10.3389/fphys.2022.1048738] [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: 09/19/2022] [Accepted: 10/31/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM), a complex set of fibrillar proteins and proteoglycans, supports the renal parenchyma and provides biomechanical and biochemical cues critical for spatial-temporal patterning of cell development and acquisition of specialized functions. As in vitro models progress towards biomimicry, more attention is paid to reproducing ECM-mediated stimuli. ECM's role in in vitro models of renal function and disease used to investigate kidney injury and regeneration is discussed. Availability, affordability, and lot-to-lot consistency are the main factors determining the selection of materials to recreate ECM in vitro. While simpler components can be synthesized in vitro, others must be isolated from animal or human tissues, either as single isolated components or as complex mixtures, such as Matrigel or decellularized formulations. Synthetic polymeric materials with dynamic and instructive capacities are also being explored for cell mechanical support to overcome the issues with natural products. ECM components can be used as simple 2D coatings or complex 3D scaffolds combining natural and synthetic materials. The goal is to recreate the biochemical signals provided by glycosaminoglycans and other signaling molecules, together with the stiffness, elasticity, segmentation, and dimensionality of the original kidney tissue, to support the specialized functions of glomerular, tubular, and vascular compartments. ECM mimicking also plays a central role in recent developments aiming to reproduce renal tissue in vitro or even in therapeutical strategies to regenerate renal function. Bioprinting of renal tubules, recellularization of kidney ECM scaffolds, and development of kidney organoids are examples. Future solutions will probably combine these technologies.
Collapse
Affiliation(s)
- Alodia Lacueva-Aparicio
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon’s Health Sciences Institute, Zaragoza, Spain,Tissue Microenvironment Lab (TME Lab), I3A, University of Zaragoza, Zaragoza, Spain
| | - Rafael Soares Lindoso
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Silvia M. Mihăilă
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Ignacio Giménez
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon’s Health Sciences Institute, Zaragoza, Spain,Institute for Health Research Aragon (IIS Aragon), Zaragoza, Spain,School of Medicine, University of Zaragoza, Zaragoza, Spain,*Correspondence: Ignacio Giménez,
| |
Collapse
|
31
|
The Upregulation of Regenerative Activity for Extracellular Vesicles with Melatonin Modulation in Chemically Defined Media. Int J Mol Sci 2022; 23:ijms232315089. [PMID: 36499413 PMCID: PMC9736868 DOI: 10.3390/ijms232315089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Extracellular vesicles (EVs) derived from human mesenchymal stem cells (hMSCs) have been widely known to have therapeutic effects by representing characteristics of the origin cells as an alternative for cell-based therapeutics. Major limitations of EVs for clinical applications include low production yields, unknown effects from serum impurities, and relatively low bioactivities against dose. In this study, we proposed a cell modulation method with melatonin for human umbilical cord MSCs (hUCMSCs) cultured in serum-free chemically defined media (CDM) to eliminate the effects of serum-derived impurities and promote regeneration-related activities. miRNAs highly associated with regeneration were selected and the expression levels of them were comparatively analyzed among various types of EVs depending on culture conditions. The EVs derived from melatonin-stimulated hUCMSCs in CDM (CDM mEVs) showed the highest expression levels of regeneration-related miRNAs, and 7 times more hsa-let-7b-5p, 5.6 times more hsa-miR-23a-3p, and 5.7 times more hsa-miR-100-5p than others, respectively. In addition, the upregulation of various functionalities, such as wound healing, angiogenesis, anti-inflammation, ROS scavenging, and anti-apoptosis, were proven using in vitro assays by simulating the characteristics of EVs with bioinformatics analysis. The present results suggest that the highly regenerative properties of hUCMSC-derived EVs were accomplished with melatonin stimulation in CDM and provided the potential for clinical uses of EVs.
Collapse
|
32
|
Zhang R, Hu Z, Wang Y, Qiu R, Wang G, Wang L, Hu B. A biomimetic double network hydrogel ameliorates renal fibrosis and promotes renal regeneration. J Mater Chem B 2022; 10:9424-9437. [PMID: 36378134 DOI: 10.1039/d2tb01939f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are serious global public health issues. Both interconnect closely, and AKI-CKD transition significantly increases the morbidity of CKD and inevitably progresses to end stage renal disease. However, with the current drug delivery system it is hard to achieve precise delivery and apply it to clinical practice due to the local fibrotic milieu of the AKI-CKD transition procedure. Consequently, new treatment options to halt or even reverse AKI-CKD transition are urgently needed. Curcumin and Ac-SDKP were proved to be capable of ameliorating renal injury and restoring renal biological function. However, due to the water-insolubility, poor absorption and ease of degradation features, their utilization based on traditional drug delivery systems was still confined to the laboratory. A new approach for the targeted delivery of curcumin and Ac-SDKP into kidneys is needed. Hydrogels, owing to their capability of targeted-drug delivery and bio-favorable nature, emerge as a promising resolution. Herein, we developed a bioinspired double network hydrogel scaffold loaded with curcumin and N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) to explore the feasibility of drug-loaded hydrogels for treatment of AKI-CKD transition. This double network hydrogel (GCS) was prepared based on gelatin and curcumin-zinc with polydopamine (DOPA) coating and then immobilized with Ac-SDKP on the surface. The prepared hydrogels possessed appropriate porosity, suitable mechanical properties, and excellent biocompatibility. In vitro, the GCS hydrogel was demonstrated to be pro-angiogenic, anti-oxidative and anti-fibrotic. In vivo, after the GCS hydrogel was implanted into partially nephrectomized rat kidneys, local renal fibrosis was observed to be improved significantly, and neo-blood vessels and neonatal renal tubules appeared around the implanted area. We speculated that the GCS hydrogel could ameliorate renal fibrosis and injury significantly and stimulate regeneration in situ. Taken together, this study demonstrated the promising potential of this bioinspired hydrogel scaffold for renal injury repair and renal regeneration.
Collapse
Affiliation(s)
- Rui Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Key Lab for Organ Failure Research, Ministry of Education Guangzhou, Guangdong, 510515, China. .,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Zifan Hu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yongqin Wang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Key Lab for Organ Failure Research, Ministry of Education Guangzhou, Guangdong, 510515, China.
| | - Renjie Qiu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Guobao Wang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Key Lab for Organ Failure Research, Ministry of Education Guangzhou, Guangdong, 510515, China.
| | - Leyu Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Bianxiang Hu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Key Lab for Organ Failure Research, Ministry of Education Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
33
|
Pan Z, Sun W, Chen Y, Tang H, Lin W, Chen J, Chen C. Extracellular Vesicles in Tissue Engineering: Biology and Engineered Strategy. Adv Healthc Mater 2022; 11:e2201384. [PMID: 36053562 DOI: 10.1002/adhm.202201384] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/07/2022] [Indexed: 01/28/2023]
Abstract
Extracellular vesicles (EVs), acting as an important ingredient of intercellular communication through paracrine actions, have gained tremendous attention in the field of tissue engineering (TE). Moreover, these nanosized extracellular particles (30-140 nm) can be incorporated into biomaterials according to different principles to facilitate signal delivery in various regenerative processes directly or indirectly. Bioactive biomaterials as the carrier will extend the retention time and realize the controlled release of EVs, which further enhance their therapeutic efficiency in tissue regeneration. Herein, the basic biological characteristics of EVs are first introduced, and then their outstanding performance in exerting direct impacts on target cells in tissue regeneration as well as indirect effects on promoting angiogenesis and regulating the immune environment, due to specific functional components of EVs (nucleic acid, protein, lipid, etc.), is emphasized. Furthermore, different design ideas for suitable EV-loaded biomaterials are also demonstrated. In the end, this review also highlights the engineered strategies, which aim at solving the problems related to natural EVs such as highly heterogeneous functions, inadequate tissue targeting capabilities, insufficient yield and scalability, etc., thus promoting the therapeutic pertinence and clinical potential of EV-based approaches in TE.
Collapse
Affiliation(s)
- Ziyin Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Weiyan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Yi Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Hai Tang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Weikang Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Jiafei Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| |
Collapse
|
34
|
Lv K, Wang Y, Lou P, Liu S, Zhou P, Yang L, Lu Y, Cheng J, Liu J. Extracellular vesicles as advanced therapeutics for the resolution of organ fibrosis: Current progress and future perspectives. Front Immunol 2022; 13:1042983. [PMCID: PMC9630482 DOI: 10.3389/fimmu.2022.1042983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
Organ fibrosis is a serious health challenge worldwide, and its global incidence and medical burden are increasing dramatically each year. Fibrosis can occur in nearly all major organs and ultimately lead to organ dysfunction. However, current clinical treatments cannot slow or reverse the progression of fibrosis to end-stage organ failure, and thus advanced anti-fibrotic therapeutics are urgently needed. As a type of naturally derived nanovesicle, native extracellular vesicles (EVs) from multiple cell types (e.g., stem cells, immune cells, and tissue cells) have been shown to alleviate organ fibrosis in many preclinical models through multiple effective mechanisms, such as anti-inflammation, pro-angiogenesis, inactivation of myofibroblasts, and fibrinolysis of ECM components. Moreover, the therapeutic potency of native EVs can be further enhanced by multiple engineering strategies, such as genetic modifications, preconditionings, therapeutic reagent-loadings, and combination with functional biomaterials. In this review, we briefly introduce the pathology and current clinical treatments of organ fibrosis, discuss EV biology and production strategies, and particularly focus on important studies using native or engineered EVs as interventions to attenuate tissue fibrosis. This review provides insights into the development and translation of EV-based nanotherapies into clinical applications in the future.
Collapse
Affiliation(s)
- Ke Lv
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yizhuo Wang
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Lou
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyun Liu
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Pingya Zhou
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Yanrong Lu
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingping Liu
- National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jingping Liu,
| |
Collapse
|
35
|
Mirmoghtadaei M, Khaboushan AS, Mohammadi B, Sadr M, Farmand H, Hassannejad Z, Kajbafzadeh AM. Kidney tissue engineering in preclinical models of renal failure: a systematic review and meta-analysis. Regen Med 2022; 17:941-955. [PMID: 36154467 DOI: 10.2217/rme-2022-0084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study aims to compare the efficacy of tissue engineering for kidney reconstruction. Materials & methods: We searched MEDLINE, EMBASE (May 2021), and reference lists of review articles. Results: 19 articles matched our inclusion criteria. A range of natural, synthetic and hybrid scaffolds with or without incorporating cells/growth factors was investigated in 937 animals. More favorable results were observed with a combination of two or more biomaterials, addition of bioactive moieties, and cell seeding. Creatinine concentration, PAX2, collagen type-1, α-SMA, vimentin, IL-1, IL-6 and TNF-α gene expressions were significantly increased compared with native control. Conclusion: Tissue engineering can improve renal function and regeneration; however, further research could benefit from using hybrid scaffolds, stem cells and large animal models.
Collapse
Affiliation(s)
- Milad Mirmoghtadaei
- Pediatric Urology & Regenerative Medicine Research Center, Gene, Cell & Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Soltani Khaboushan
- Pediatric Urology & Regenerative Medicine Research Center, Gene, Cell & Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Mohammadi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Matin Sadr
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hooman Farmand
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Hassannejad
- Pediatric Urology & Regenerative Medicine Research Center, Gene, Cell & Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology & Regenerative Medicine Research Center, Gene, Cell & Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Leung KS, Shirazi S, Cooper LF, Ravindran S. Biomaterials and Extracellular Vesicle Delivery: Current Status, Applications and Challenges. Cells 2022; 11:cells11182851. [PMID: 36139426 PMCID: PMC9497093 DOI: 10.3390/cells11182851] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 12/14/2022] Open
Abstract
In this review, we will discuss the current status of extracellular vesicle (EV) delivery via biopolymeric scaffolds for therapeutic applications and the challenges associated with the development of these functionalized scaffolds. EVs are cell-derived membranous structures and are involved in many physiological processes. Naïve and engineered EVs have much therapeutic potential, but proper delivery systems are required to prevent non-specific and off-target effects. Targeted and site-specific delivery using polymeric scaffolds can address these limitations. EV delivery with scaffolds has shown improvements in tissue remodeling, wound healing, bone healing, immunomodulation, and vascular performance. Thus, EV delivery via biopolymeric scaffolds is becoming an increasingly popular approach to tissue engineering. Although there are many types of natural and synthetic biopolymers, the overarching goal for many tissue engineers is to utilize biopolymers to restore defects and function as well as support host regeneration. Functionalizing biopolymers by incorporating EVs works toward this goal. Throughout this review, we will characterize extracellular vesicles, examine various biopolymers as a vehicle for EV delivery for therapeutic purposes, potential mechanisms by which EVs exert their effects, EV delivery for tissue repair and immunomodulation, and the challenges associated with the use of EVs in scaffolds.
Collapse
Affiliation(s)
- Kasey S. Leung
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Sajjad Shirazi
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Lyndon F. Cooper
- School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Sriram Ravindran
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL 60612, USA
- Correspondence:
| |
Collapse
|
37
|
Morales RTT, Ko J. Future of Digital Assays to Resolve Clinical Heterogeneity of Single Extracellular Vesicles. ACS NANO 2022; 16:11619-11645. [PMID: 35904433 PMCID: PMC10174080 DOI: 10.1021/acsnano.2c04337] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Extracellular vesicles (EVs) are complex lipid membrane vehicles with variable expressions of molecular cargo, composed of diverse subpopulations that participate in the intercellular signaling of biological responses in disease. EV-based liquid biopsies demonstrate invaluable clinical potential for overhauling current practices of disease management. Yet, EV heterogeneity is a major needle-in-a-haystack challenge to translate their use into clinical practice. In this review, existing digital assays will be discussed to analyze EVs at a single vesicle resolution, and future opportunities to optimize the throughput, multiplexing, and sensitivity of current digital EV assays will be highlighted. Furthermore, this review will outline the challenges and opportunities that impact the clinical translation of single EV technologies for disease diagnostics and treatment monitoring.
Collapse
Affiliation(s)
- Renee-Tyler T Morales
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jina Ko
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
38
|
Tang TT, Wang B, Lv LL, Dong Z, Liu BC. Extracellular vesicles for renal therapeutics: State of the art and future perspective. J Control Release 2022; 349:32-50. [PMID: 35779658 DOI: 10.1016/j.jconrel.2022.06.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 12/21/2022]
Abstract
With the ever-increasing burden of kidney disease, the need for developing new therapeutics to manage this disease has never been greater. Extracellular vesicles (EVs) are natural membranous nanoparticles present in virtually all organisms. Given their excellent delivery capacity in the body, EVs have emerged as a frontier technology for drug delivery and have the potential to usher in a new era of nanomedicine for kidney disease. This review is focused on why EVs are such compelling drug carriers and how to release their fullest potentiality in renal therapeutics. We discuss the unique features of EVs compared to artificial nanoparticles and outline the engineering technologies and steps in developing EV-based therapeutics, with an emphasis on the emerging approaches to target renal cells and prolong kidney retention. We also explore the applications of EVs as natural therapeutics or as drug carriers in the treatment of renal disorders and present our views on the critical challenges in manufacturing EVs as next-generation renal therapeutics.
Collapse
Affiliation(s)
- Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China; Department of Pathology and Pathophysiology, Southeast University School of Medicine, Nanjing, China
| | - Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China.
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China.
| |
Collapse
|
39
|
Park SY, Kim DS, Kim HM, Lee JK, Hwang DY, Kim TH, You S, Han DK. Human Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Neural Differentiation of Neural Progenitor Cells. Int J Mol Sci 2022; 23:ijms23137047. [PMID: 35806058 PMCID: PMC9267053 DOI: 10.3390/ijms23137047] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been adopted in various preclinical and clinical studies because of their multipotency and low immunogenicity. However, numerous obstacles relating to safety issues remain. Therefore, MSC-derived extracellular vesicles (EVs) have been recently employed. EVs are nano-sized endoplasmic reticulum particles generated and released in cells that have similar biological functions to their origin cells. EVs act as cargo for bioactive molecules such as proteins and genetic materials and facilitate tissue regeneration. EVs obtained from adipose-derived MSC (ADMSC) also have neuroprotective and neurogenesis effects. On the basis of the versatile effects of EVs, we aimed to enhance the neural differentiation ability of ADMSC-derived EVs by elucidating the neurogenic-differentiation process. ADMSC-derived EVs isolated from neurogenesis conditioned media (differentiated EVs, dEVs) increased neurogenic ability by altering innate microRNA expression and cytokine composition. Consequently, dEVs promoted neuronal differentiation of neural progenitor cells in vitro, suggesting that dEVs are a prospective candidate for EV-based neurological disorder regeneration therapy.
Collapse
Affiliation(s)
- So-Yeon Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-Y.P.); (D.-S.K.); (H.-M.K.); (J.-K.L.); (D.-Y.H.)
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-Y.P.); (D.-S.K.); (H.-M.K.); (J.-K.L.); (D.-Y.H.)
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea;
| | - Hyun-Mun Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-Y.P.); (D.-S.K.); (H.-M.K.); (J.-K.L.); (D.-Y.H.)
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-Y.P.); (D.-S.K.); (H.-M.K.); (J.-K.L.); (D.-Y.H.)
| | - Dong-Youn Hwang
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-Y.P.); (D.-S.K.); (H.-M.K.); (J.-K.L.); (D.-Y.H.)
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea;
| | - Seungkwon You
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
- Correspondence: (S.Y.); (D.K.H.)
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-Y.P.); (D.-S.K.); (H.-M.K.); (J.-K.L.); (D.-Y.H.)
- Correspondence: (S.Y.); (D.K.H.)
| |
Collapse
|
40
|
Li M, Fang F, Sun M, Zhang Y, Hu M, Zhang J. Extracellular vesicles as bioactive nanotherapeutics: An emerging paradigm for regenerative medicine. Theranostics 2022; 12:4879-4903. [PMID: 35836815 PMCID: PMC9274746 DOI: 10.7150/thno.72812] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/05/2022] [Indexed: 01/12/2023] Open
Abstract
In recent decades, extracellular vesicles (EVs), as bioactive cell-secreted nanoparticles which are involved in various physiological and pathological processes including cell proliferation, immune regulation, angiogenesis and tissue repair, have emerged as one of the most attractive nanotherapeutics for regenerative medicine. Herein we provide a systematic review of the latest progress of EVs for regenerative applications. Firstly, we will briefly introduce the biogenesis, function and isolation technology of EVs. Then, the underlying therapeutic mechanisms of the native unmodified EVs and engineering strategies of the modified EVs as regenerative entities will be discussed. Subsequently, the main focus will be placed on the tissue repair and regeneration applications of EVs on various organs including brain, heart, bone and cartilage, liver and kidney, as well as skin. More importantly, current clinical trials of EVs for regenerative medicine will also be briefly highlighted. Finally, the future challenges and insightful perspectives of the currently developed EV-based nanotherapeutics in biomedicine will be discussed. In short, the bioactive EV-based nanotherapeutics have opened new horizons for biologists, chemists, nanoscientists, pharmacists, as well as clinicians, making possible powerful tools and therapies for regenerative medicine.
Collapse
Affiliation(s)
- Min Li
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Fang Fang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Meng Sun
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yinfeng Zhang
- International Medical Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, P. R. China
| | - Min Hu
- Department of Hepatobiliary Surgery, Jinan University First Affiliated Hospital, Guangzhou, 510630, P. R. China
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100081, P. R. China
| |
Collapse
|
41
|
Liu X, Hu L, Liu F. Mesenchymal stem cell-derived extracellular vesicles for cell-free therapy of ocular diseases. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:102-117. [PMID: 39698446 PMCID: PMC11648472 DOI: 10.20517/evcna.2022.08] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/31/2022] [Accepted: 04/18/2022] [Indexed: 12/20/2024]
Abstract
Mesenchymal stem cells-derived extracellular vesicles (MSC-EVs) have noticeably attracted clinicians' attention in treating ocular diseases. As the paracrine factor of MSCs and an alternative for cell-free therapies, MSC-EVs can be conveniently dropped over the ocular surface or diffused through the retina upon intravitreal injection, without increasing the risks of cellular rejection and tumor formation. For clinical translation, a standardized and scalable production, as well as reprogramming the MSC-EVs, are highly encouraged. This review aims to assess the potential approaches for EV production and functional modification, in addition to summarizing the worldwide clinical trials initiated for various physiological systems and the specific biochemical effects of MSC-EVs on the therapy of eye diseases. Recent advances in the therapy of ocular diseases based on MSC-EVs are reviewed, and the associated challenges and prospects are discussed as well.
Collapse
Affiliation(s)
- Xiaoling Liu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Liang Hu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Fei Liu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
- Wenzhou Institute, University of Chinese Academy of Science, Wenzhou 325000, Zhejiang, China
| |
Collapse
|
42
|
Ko GR, Lee JS. Engineering of Immune Microenvironment for Enhanced Tissue Remodeling. Tissue Eng Regen Med 2022; 19:221-236. [PMID: 35041181 PMCID: PMC8971302 DOI: 10.1007/s13770-021-00419-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/30/2021] [Accepted: 12/14/2021] [Indexed: 01/21/2023] Open
Abstract
The capability to restore the structure and function of tissues damaged by fatal diseases and trauma is essential for living organisms. Various tissue engineering approaches have been applied in lesions to enhance tissue regeneration after injuries and diseases in living organisms. However, unforeseen immune reactions by the treatments interfere with successful healing and reduce the therapeutic efficacy of the strategies. The immune system is known to play essential roles in the regulation of the microenvironment and recruitment of cells that directly or indirectly participate in tissue remodeling in defects. Accordingly, regenerative immune engineering has emerged as a novel approach toward efficiently inducing regeneration using engineering techniques that modulate the immune system. It is aimed at providing a favorable immune microenvironment based on the controlled balance between pro-inflammation and anti-inflammation. In this review, we introduce recent developments in immune engineering therapeutics based on various cell types and biomaterials. These developments could potentially overcome the therapeutic limitations of tissue remodeling.
Collapse
Affiliation(s)
- Ga Ryang Ko
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Jung Seung Lee
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| |
Collapse
|
43
|
Xue F, Cornelissen JJ, Yuan Q, Cao S. Delivery of MicroRNAs by plant virus-based nanoparticles to functionally alter the osteogenic differentiation of human mesenchymal stem cells. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.04.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
44
|
Baskapan B, Callanan A. Electrospinning Fabrication Methods to Incorporate Laminin in Polycaprolactone for Kidney Tissue Engineering. Tissue Eng Regen Med 2022; 19:73-82. [PMID: 34714533 PMCID: PMC8782962 DOI: 10.1007/s13770-021-00398-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/23/2021] [Accepted: 09/09/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Today's treatment options for renal diseases fall behind the need, as the number of patients has increased considerably over the last few decades. Tissue engineering (TE) is one avenue which may provide a new approach for renal disease treatment. This involves creating a niche where seeded cells can function in an intended way. One approach to TE is combining natural extracellular matrix proteins with synthetic polymers, which has been shown to have many positives, yet a little is understood in kidney. Herein, we investigate the incorporation of laminin into polycaprolactone electrospun scaffolds. METHOD The scaffolds were enriched with laminin via either direct blending with polymer solution or in a form of emulsion with a surfactant. Renal epithelial cells (RC-124) were cultured on scaffolds up to 21 days. RESULTS Mechanical characterization demonstrated that the addition of the protein changed Young's modulus of polymeric fibres. Cell viability and DNA quantification tests revealed the capability of the scaffolds to maintain cell survival up to 3 weeks in culture. Gene expression analysis indicated healthy cells via three key markers. CONCLUSION Our results show the importance of hybrid scaffolds for kidney tissue engineering.
Collapse
Affiliation(s)
- Büsra Baskapan
- grid.4305.20000 0004 1936 7988Institute for Bioengineering, School of Engineering, University of Edinburgh, Faraday Building, King’s Buildings, Colin Maclaurin Road, Edinburg, EH9 3DW UK
| | - Anthony Callanan
- grid.4305.20000 0004 1936 7988Institute for Bioengineering, School of Engineering, University of Edinburgh, Faraday Building, King’s Buildings, Colin Maclaurin Road, Edinburg, EH9 3DW UK
| |
Collapse
|
45
|
Zhao B, Chen Q, Zhao L, Mao J, Huang W, Han X, Liu Y. Periodontal Ligament Stem Cell-Derived Small Extracellular Vesicles Embedded in Matrigel Enhance Bone Repair Through the Adenosine Receptor Signaling Pathway. Int J Nanomedicine 2022; 17:519-536. [PMID: 35140462 PMCID: PMC8819539 DOI: 10.2147/ijn.s346755] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Purpose Small extracellular vesicles (sEVs) are natural biocarriers for biomolecule transfer between cells and promising therapeutic strategies for bone defect repair. In this study, human periodontal ligament stem cell (PDLSC)-derived sEVs (P-EVs) were immobilized in Matrigel to establish a topical cell-free transplantation strategy for bone repair. Methods PDLSCs were cultured and P-EVs were isolated from the culture supernatant. In a rat bilateral calvarial defect model, P-EV/Matrigel was plugged into one defect and PBS/Matrigel was applied to the other. Bone repair in vivo was assessed by micro-computed tomography, histomorphometry, and immunohistochemical staining. In vitro, we investigated the effects of P-EVs on the proliferation and migration capabilities of bone marrow mesenchymal stem cells (BMMSCs) and explored the potential mechanism of action. Results The in vivo study showed that P-EV/Matrigel accelerated bone tissue repair by increasing cell infiltration when compared with the control. In vitro, P-EVs enhanced proliferation and migration of BMMSCs via increased phosphorylation of AKT and extracellular signal-regulated kinase 1/2 (ERK1/2). The role of P-EV-induced adenosine receptor signaling in AKT and ERK1/2 phosphorylation was a key mediator during enhanced BMMSC migration. Conclusion These results are the first to demonstrate that P-EVs accelerated the repair of bone defects, partially through promoting cell proliferation and migration. P-EV/Matrigel, which combines topical EV-implantation and extracellular matrix scaffolds, provides a new cell-free strategy for bone tissue repair.
Collapse
Affiliation(s)
- Bingjiao Zhao
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, People’s Republic of China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
| | - Qingqing Chen
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
| | - Liru Zhao
- Department of Orthodontics, School of Stomatology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Jiaqi Mao
- Department of Orthodontics, School of Stomatology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Wei Huang
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
| | - Xinxin Han
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, People’s Republic of China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
- Correspondence: Yuehua Liu, Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, 356 East Beijing Road, Shanghai, 200001, People’s Republic of China, Tel +86-63298475, Fax +86-63614515, Email
| |
Collapse
|
46
|
Advances in tissue engineering technology for kidney regeneration and construction. J Artif Organs 2022; 25:191-194. [PMID: 35102521 DOI: 10.1007/s10047-022-01315-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
Abstract
Tissue engineering is a highly interdisciplinary research field aiming at repairing, replacing, and regenerating the defective tissues. Over several decades of research, a variety of methods have been developed. The technical methods can be categorized into scaffold-based and scaffold-free strategies. In this mini review, the discussion will be focused on the technical methods of tissue engineering for kidney regeneration and construction.
Collapse
|
47
|
Yoo YI, Ko KW, Cha SG, Park SY, Woo J, Han DK. Highly effective induction of cell-derived extracellular matrix by macromolecular crowding for osteogenic differentiation of mesenchymal stem cells. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2021.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
48
|
Comparison of Surface Functionalization of PLGA Composite to Immobilize Extracellular Vesicles. Polymers (Basel) 2021; 13:polym13213643. [PMID: 34771200 PMCID: PMC8587822 DOI: 10.3390/polym13213643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023] Open
Abstract
Endothelialization by materials provides a promising approach for the rapid re-endothelialization of a cardiovascular implantation. Although previous studies have focused on improving endothelialization through the immobilization of bioactive molecules onto the surface of biodegradable implants, comparative studies of effective surface modification have not yet been reported. Here, we conducted a comparative study on the surface modification of poly(lactide-co-glycolide) (PLGA)-based composites to graft mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) using three different materials, fibronectin (FN), polyethylenimine (PEI), and polydopamine (PDA), which have different bond strengths of ligand–receptor interaction, ionic bond, and covalent bond, respectively. Further in vitro analysis exhibited that MSC-EVs released from all modified films sustainably, but the MSC-EVs grafted onto the surface coated with PEI are more effective than other groups in increasing angiogenesis and reducing the inflammatory responses in endothelial cells. Therefore, the overall results demonstrated that PEI is a desirable coating reagent for the immobilization of MSC-EVs on the surface of biodegradable implants.
Collapse
|
49
|
Guo Y, Wu D, Zhang X, Zhang K, Luo Y. Biomolecules in cell-derived extracellular vesicle chariots as warriors to repair damaged tissues. NANOSCALE 2021; 13:16017-16033. [PMID: 34570853 DOI: 10.1039/d1nr04999b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In this review, we highlight the innovative applications of biomolecules from parent cell-derived extracellular vesicles (EVs) for tissue repair that have been developed in recent years. We evaluate the underlying mechanisms and therapeutic efficacy of each therapy. In previous literature reviews, it was most common to classify the use of EVs in tissue repair by disease type. This article reviews the role of three biomolecules in EVs in tissue repair. This review first summarizes the definitions and classifications of EVs. Then, the importance and significance of treating tissue damage with EVs are discussed. In particular, EV contents for tissue repair are three main types of biomolecules: proteins, RNAs and cell growth factors. The therapeutic and repair mechanisms of the biomolecules are discussed respectively. Finally, the development prospect and potential challenges of EV contents from highly differentiated cells as specific agents for tissue repair are summarized. When EVs are used to treat diseases such as tissue or organ damage, EVs play a role in delivery, and the real repair effect is effected by the various biomolecules carried by EVs. We believe that EV biomolecules have unparalleled advantages and clinical transformation potential for tissue repair and expect this review to inspire more intensive research work in this field.
Collapse
Affiliation(s)
- Yingshu Guo
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Di Wu
- School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Xu Zhang
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China.
| |
Collapse
|
50
|
Calcat-i-Cervera S, Sanz-Nogués C, O'Brien T. When Origin Matters: Properties of Mesenchymal Stromal Cells From Different Sources for Clinical Translation in Kidney Disease. Front Med (Lausanne) 2021; 8:728496. [PMID: 34616756 PMCID: PMC8488400 DOI: 10.3389/fmed.2021.728496] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced therapy medicinal products (ATMPs) offer new prospects to improve the treatment of conditions with unmet medical needs. Kidney diseases are a current major health concern with an increasing global prevalence. Chronic renal failure appears after many years of impairment, which opens a temporary window to apply novel therapeutic approaches to delay or halt disease progression. The immunomodulatory, anti-inflammatory, and pro-regenerative properties of mesenchymal stromal cells (MSCs) have sparked interest for their use in cell-based regenerative therapies. Currently, several early-phase clinical trials have been completed and many are ongoing to explore MSC safety and efficacy in a wide range of nephropathies. However, one of the current roadblocks to the clinical translation of MSC therapies relates to the lack of standardization and harmonization of MSC manufacturing protocols, which currently hinders inter-study comparability. Studies have shown that cell culture processing variables can have significant effects on MSC phenotype and functionality, and these are highly variable across laboratories. In addition, heterogeneity within MSC populations is another obstacle. Furthermore, MSCs may be isolated from several sources which adds another variable to the comparative assessment of outcomes. There is now a growing body of literature highlighting unique and distinctive properties of MSCs according to the tissue origin, and that characteristics such as donor, age, sex and underlying medical conditions may alter the therapeutic effect of MSCs. These variables must be taken into consideration when developing a cell therapy product. Having an optimal scale-up strategy for MSC manufacturing is critical for ensuring product quality while minimizing costs and time of production, as well as avoiding potential risks. Ideally, optimal scale-up strategies must be carefully considered and identified during the early stages of development, as making changes later in the bioprocess workflow will require re-optimization and validation, which may have a significant long-term impact on the cost of the therapy. This article provides a summary of important cell culture processing variables to consider in the scale-up of MSC manufacturing as well as giving a comprehensive review of tissue of origin-specific biological characteristics of MSCs and their use in current clinical trials in a range of renal pathologies.
Collapse
Affiliation(s)
| | | | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), CÚRAM, Biomedical Science Building, National University of Ireland, Galway, Ireland
| |
Collapse
|