1
|
Carmona JU, López C. Efficacy of Platelet-Rich Plasma in the Treatment of Equine Tendon and Ligament Injuries: A Systematic Review of Clinical and Experimental Studies. Vet Sci 2025; 12:382. [PMID: 40284884 PMCID: PMC12031177 DOI: 10.3390/vetsci12040382] [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: 03/10/2025] [Revised: 04/05/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
(1) Background: Tendon and ligament injuries are a leading cause of lameness in horses, with significant economic implications. Platelet-rich plasma (PRP) has gained attention for its regenerative potential, but its efficacy remains uncertain due to inconsistent study designs and reporting. (2) Methods: This systematic review, following the PRISMA guidelines, evaluated 22 studies (clinical and experimental) to assess the safety and efficacy of PRP in treating equine tendon and ligament injuries. The risk of bias was analyzed using the ROBINS-I and RoB 2.0 tools. (3) Results: PRP demonstrated a favorable safety profile, with no severe adverse effects reported. Clinical outcomes included improved lameness scores, ultrasonographic tissue organization, and return-to-work rates. However, variability in PRP formulations (e.g., leukocyte-rich vs. leukocyte-reduced) and activation methods (e.g., calcium chloride, thrombin) contributed to inconsistent results. Experimental studies supported PRP's role in collagen synthesis and neovascularization, but comparative trials with stem cells or other therapies (e.g., extracorporeal shockwave) showed mixed results. The methodological quality of studies varied, with only 27% achieving "good" scores for PRP reporting. (4) Conclusions: PRP is a safe and potentially effective treatment, but its clinical application is hindered by a lack of standardization. Future research should focus on large, randomized controlled trials with uniform PRP protocols, long-term (≥2 years) efficacy assessments, comparative studies with MSC combinations, and cost-effectiveness analyses.
Collapse
Affiliation(s)
- Jorge U. Carmona
- Grupo de Investigación Terapia Regenerativa, Departamento de Salud Animal, Universidad de Caldas, Calle 65 No 26-10, Manizales 170004, Colombia
| | - Catalina López
- Grupo de Investigación Patología Clínica Veterinaria, Departamento de Salud Animal, Universidad de Caldas, Calle 65 No 26-10, Manizales 170004, Colombia;
| |
Collapse
|
2
|
Veronese S, Ossanna R, Tehrani SG, Bernardi P, Khabouri S, Cannone V, Nicoletti MM, Goisis M, Sbarbati A. The Impact of Acoustic Wave Therapy on Viability and Differentiation Capacity of Human Adipose Stem Cells. J Cosmet Dermatol 2025; 24:e70142. [PMID: 40145242 PMCID: PMC11948166 DOI: 10.1111/jocd.70142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/10/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Acoustic Wave Therapy (AWT) is a particular shock wave treatment that combines focused and radial shock waves, resulting in particular efficiency in the body's superficial layers. Its application before fat grafting has the potential to enhance it, favoring a better result of the graft. AIMS The aim of this study was to analyze the effects of AWT on fat tissue. METHODS Histological analysis of fat harvested from the abdomen of 40 subjects was evaluated. Twenty subjects had been pre-treated with AWT, while 20 had not. Extraction and characterization of adipose-derived stem cells (ADSCs) were performed for all. The multilineage differentiation capacity of extracted ADSCs and exosome extracellular vesicles' production from the fat samples were analyzed in both groups of specimens. RESULTS All 40 specimens presented both preservation of the structure of the tissue and of the cells, particularly of adipocytes. The cell growth resulted higher for pre-treated samples. All ADSCs from all the samples were able to differentiate along adipogenic, chondrogenic, and osteogenic lineages. In particular, for pre-treated samples, in the adipogenic lineage, a more advanced maturation phase was observed, and in the chondrogenic lineage, a chondroid matrix was evident around chondrocyte aggregates. All the samples reacted with exosome extracellular vesicles' production to stress. CONCLUSIONS The results highlighted that AWT aligns with the in-force minimal manipulation regulations, as the tissue and cells' structure and functionality were preserved. The types of differentiation observed permitted us to speculate about possible new applications in aesthetic and regenerative medicine.
Collapse
Affiliation(s)
- Sheila Veronese
- Department of Neuroscience, Biomedicine, and Movement, Section of Anatomy and HistologyUniversity of VeronaVeronaItaly
| | - Riccardo Ossanna
- Department of Neuroscience, Biomedicine, and Movement, Section of Anatomy and HistologyUniversity of VeronaVeronaItaly
| | - Sara Ghazanfar Tehrani
- Department of Neuroscience, Biomedicine, and Movement, Section of Anatomy and HistologyUniversity of VeronaVeronaItaly
| | - Paolo Bernardi
- Department of Neuroscience, Biomedicine, and Movement, Section of Anatomy and HistologyUniversity of VeronaVeronaItaly
| | | | - Velia Cannone
- Department of Neuroscience, Biomedicine, and Movement, Section of Anatomy and HistologyUniversity of VeronaVeronaItaly
| | | | | | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine, and Movement, Section of Anatomy and HistologyUniversity of VeronaVeronaItaly
| |
Collapse
|
3
|
Wu KC, Chang YH, Ding DC, Lin SZ. Mesenchymal Stromal Cells for Aging Cartilage Regeneration: A Review. Int J Mol Sci 2024; 25:12911. [PMID: 39684619 PMCID: PMC11641625 DOI: 10.3390/ijms252312911] [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: 11/06/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Cartilage degeneration is a key feature of aging and osteoarthritis, characterized by the progressive deterioration of joint function, pain, and limited mobility. Current treatments focus on symptom relief, not cartilage regeneration. Mesenchymal stromal cells (MSCs) offer a promising therapeutic option due to their capability to differentiate into chondrocytes, modulate inflammation, and promote tissue regeneration. This review explores the potential of MSCs for cartilage regeneration, examining their biological properties, action mechanisms, and applications in preclinical and clinical settings. MSCs derived from bone marrow, adipose tissue, and other sources can self-renew and differentiate into multiple cell types. In aging cartilage, they aid in tissue regeneration by secreting growth factors and cytokines that enhance repair and modulate immune responses. Recent preclinical studies show that MSCs can restore cartilage integrity, reduce inflammation, and improve joint function, although clinical translation remains challenging due to limitations such as cell viability, scalability, and regulatory concerns. Advancements in MSC delivery, including scaffold-based approaches and engineered exosomes, may improve therapeutic effectiveness. Potential risks, such as tumorigenicity and immune rejection, are also discussed, emphasizing the need for optimized treatment protocols and large-scale clinical trials to develop effective, minimally invasive therapies for cartilage regeneration.
Collapse
Affiliation(s)
- Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
- Institute of Medical Sciences, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
4
|
Stoian A, Adil A, Biniazan F, Haykal S. Two Decades of Advances and Limitations in Organ Recellularization. Curr Issues Mol Biol 2024; 46:9179-9214. [PMID: 39194760 DOI: 10.3390/cimb46080543] [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: 07/31/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
The recellularization of tissues after decellularization is a relatively new technology in the field of tissue engineering (TE). Decellularization involves removing cells from a tissue or organ, leaving only the extracellular matrix (ECM). This can then be recellularized with new cells to create functional tissues or organs. The first significant mention of recellularization in decellularized tissues can be traced to research conducted in the early 2000s. One of the landmark studies in this field was published in 2008 by Ott, where researchers demonstrated the recellularization of a decellularized rat heart with cardiac cells, resulting in a functional organ capable of contraction. Since then, other important studies have been published. These studies paved the way for the widespread application of recellularization in TE, demonstrating the potential of decellularized ECM to serve as a scaffold for regenerating functional tissues. Thus, although the concept of recellularization was initially explored in previous decades, these studies from the 2000s marked a major turning point in the development and practical application of the technology for the recellularization of decellularized tissues. The article reviews the historical advances and limitations in organ recellularization in TE over the last two decades.
Collapse
Affiliation(s)
- Alina Stoian
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Aisha Adil
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Felor Biniazan
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Siba Haykal
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
- Reconstructive Oncology, Division of Plastic and Reconstructive Surgery, Smilow Cancer Hospital, Yale, New Haven, CT 06519, USA
| |
Collapse
|
5
|
Arbade G, Jose JV, Gulbake A, Kadam S, Kashte SB. From stem cells to extracellular vesicles: a new horizon in tissue engineering and regenerative medicine. Cytotechnology 2024; 76:363-401. [PMID: 38933869 PMCID: PMC11196501 DOI: 10.1007/s10616-024-00631-4] [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: 07/27/2023] [Accepted: 04/07/2024] [Indexed: 06/28/2024] Open
Abstract
In the fields of tissue engineering and regenerative medicine, extracellular vesicles (EVs) have become viable therapeutic tools. EVs produced from stem cells promote tissue healing by regulating the immune system, enhancing cell proliferation and aiding remodeling processes. Recently, EV has gained significant attention from researchers due to its ability to treat various diseases. Unlike stem cells, stem cell-derived EVs show lower immunogenicity, are less able to overcome biological barriers, and have a higher safety profile. This makes the use of EVs derived from cell-free stem cells a promising alternative to whole-cell therapy. This review focuses on the biogenesis, isolation, and characterization of EVs and highlights their therapeutic potential for bone fracture healing, wound healing, and neuronal tissue repair and treatment of kidney and intestinal diseases. Additionally, this review discusses the potential of EVs for the treatment of cancer, COVID-19, and HIV. In summary, the use of EVs derived from stem cells offers a new horizon for applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
| | | | - Arvind Gulbake
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, (NIPER G), Guwahati, Assam 781101 India
| | - Sachin Kadam
- Sophisticated Analytical and Technical Help Institute, Indian Institute of Technology, Delhi, New Delhi 110016 India
| | - Shivaji B. Kashte
- Department of Stem Cell and Regenerative Medicine, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Institution Deemed to be University), Kolhapur, MS 416006 India
| |
Collapse
|
6
|
Klangprapan J, Souza GR, Ferreira JN. Bioprinting salivary gland models and their regenerative applications. BDJ Open 2024; 10:39. [PMID: 38816372 PMCID: PMC11139920 DOI: 10.1038/s41405-024-00219-2] [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: 01/12/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVE Salivary gland (SG) hypofunction is a common clinical condition arising from radiotherapy to suppress head and neck cancers. The radiation often destroys the SG secretory acini, and glands are left with limited regenerative potential. Due to the complex architecture of SG acini and ducts, three-dimensional (3D) bioprinting platforms have emerged to spatially define these in vitro epithelial units and develop mini-organs or organoids for regeneration. Due to the limited body of evidence, this comprehensive review highlights the advantages and challenges of bioprinting platforms for SG regeneration. METHODS SG microtissue engineering strategies such as magnetic 3D bioassembly of cells and microfluidic coaxial 3D bioprinting of cell-laden microfibers and microtubes have been proposed to replace the damaged acinar units, avoid the use of xenogeneic matrices (like Matrigel), and restore salivary flow. RESULTS Replacing the SG damaged organ is challenging due to its complex architecture, which combines a ductal network with acinar epithelial units to facilitate a unidirectional flow of saliva. Our research group was the first to develop 3D bioassembly SG epithelial functional organoids with innervation to respond to both cholinergic and adrenergic stimulation. More recently, microtissue engineering using coaxial 3D bioprinting of hydrogel microfibers and microtubes could also supported the formation of viable epithelial units. Both bioprinting approaches could overcome the need for Matrigel by facilitating the assembly of adult stem cells, such as human dental pulp stem cells, and primary SG cells into micro-sized 3D constructs able to produce their own matrix and self-organize into micro-modular tissue clusters with lumenized areas. Furthermore, extracellular vesicle (EV) therapies from organoid-derived secretome were also designed and validated ex vivo for SG regeneration after radiation damage. CONCLUSION Magnetic 3D bioassembly and microfluidic coaxial bioprinting platforms have the potential to create SG mini-organs for regenerative applications via organoid transplantation or organoid-derived EV therapies.
Collapse
Affiliation(s)
- Jutapak Klangprapan
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Pathumwan, Bangkok, 10330, Thailand
| | - Glauco R Souza
- Greiner Bio-one North America Inc., 4238 Capital Drive, Monroe, NC, 28110, USA
| | - João N Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
7
|
Liu Y, Zhang M, Wang C, Chen H, Su D, Yang C, Tao Y, Lv X, Zhou Z, Li J, Liao Y, You J, Wang Z, Cheng F, Yang R. Human Umbilical Cord Mesenchymal Stromal Cell-Derived Extracellular Vesicles Induce Fetal Wound Healing Features Revealed by Single-Cell RNA Sequencing. ACS NANO 2024; 18:13696-13713. [PMID: 38751164 DOI: 10.1021/acsnano.4c01401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The potential of human umbilical cord mesenchymal stromal cell-derived extracellular vesicles (hucMSC-EVs) in wound healing is promising, yet a comprehensive understanding of how fibroblasts and keratinocytes respond to this treatment remains limited. This study utilizes single-cell RNA sequencing (scRNA-seq) to investigate the impact of hucMSC-EVs on the cutaneous wound microenvironment in mice. Through rigorous single-cell analyses, we unveil the emergence of hucMSC-EV-induced hematopoietic fibroblasts and MMP13+ fibroblasts. Notably, MMP13+ fibroblasts exhibit fetal-like expressions of MMP13, MMP9, and HAS1, accompanied by heightened migrasome activity. Activation of MMP13+ fibroblasts is orchestrated by a distinctive PIEZO1-calcium-HIF1α-VEGF-MMP13 pathway, validated through murine models and dermal fibroblast assays. Organotypic culture assays further affirm that these activated fibroblasts induce keratinocyte migration via MMP13-LRP1 interactions. This study significantly contributes to our understanding of fibroblast heterogeneities as well as intercellular interactions in wound healing and identifies hucMSC-EV-induced hematopoietic fibroblasts as potential targets for reprogramming. The therapeutic targets presented by these fibroblasts offer exciting prospects for advancing wound healing strategies.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Medical School of Chinese People's Liberation Army, 100039 Beijing, China
- Department of Dermatology, the Seventh Medical Center of Chinese PLA General Hospital, 100010 Beijing, China
| | - Mingwang Zhang
- Department of Dermatology, Southwest Hospital, Army Medical University, 400038 Chongqing, China
| | - Chenhui Wang
- Bioinformatics Center of AMMS, Beijing 100063, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, 510275 Shenzhen, China
| | - Dandan Su
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, 510275 Shenzhen, China
| | | | - Yuandong Tao
- Department of Pediatric Urology, the Seventh Medical Center of Chinese PLA General Hospital, 100010 Beijing, China
| | - Xuexue Lv
- Department of Pediatric Urology, the Seventh Medical Center of Chinese PLA General Hospital, 100010 Beijing, China
| | - Zhe Zhou
- Bioinformatics Center of AMMS, Beijing 100063, China
| | - Jiangbo Li
- Bioinformatics Center of AMMS, Beijing 100063, China
| | - Yong Liao
- Department of Dermatology, the Seventh Medical Center of Chinese PLA General Hospital, 100010 Beijing, China
| | - Jia You
- Biomedical Treatment Center, the Seventh Medical Center of Chinese PLA General Hospital, 100010 Beijing, China
| | - Zhengxu Wang
- Biomedical Treatment Center, the Seventh Medical Center of Chinese PLA General Hospital, 100010 Beijing, China
| | - Fang Cheng
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, 510275 Shenzhen, China
| | - Rongya Yang
- Department of Dermatology, the Seventh Medical Center of Chinese PLA General Hospital, 100010 Beijing, China
| |
Collapse
|
8
|
Han P, Raveendran N, Liu C, Basu S, Jiao K, Johnson N, Moran CS, Ivanovski S. 3D bioprinted small extracellular vesicles from periodontal cells enhance mesenchymal stromal cell function. BIOMATERIALS ADVANCES 2024; 158:213770. [PMID: 38242057 DOI: 10.1016/j.bioadv.2024.213770] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 01/21/2024]
Abstract
Recent research indicates that combining 3D bioprinting and small extracellular vesicles (sEVs) offers a promising 'cell-free' regenerative medicine approach for various tissue engineering applications. Nonetheless, the majority of existing research has focused on bioprinting of sEVs sourced from cell lines. There remains a notable gap in research regarding the bioprinting of sEVs derived from primary human periodontal cells and their potential impact on ligamentous and osteogenic differentiation. Here, we investigated the effect of 3D bioprinted periodontal cell sEVs constructs on the differentiation potential of human buccal fat pad-derived mesenchymal stromal cells (hBFP-MSCs). Periodontal cell-derived sEVs were enriched by size exclusion chromatography (SEC) with particle-shaped morphology, and characterized by being smaller than 200 nm in size and CD9/CD63/CD81 positive, from primary human periodontal ligament cells (hPDLCs) and human gingival fibroblasts (hGFs). The sEVs were then 3D bioprinted in 10 % gelatin methacryloyl (GelMA) via microextrusion bioprinting. Release of sEVs from bioprinted constructs was determined by DiO-labelling and confocal imaging, and CD9 ELISA. Attachment and ligament/osteogenic/cementogenic differentiation of hBFP-MSCs was assessed on bioprinted GelMA, without and with sEVs (GelMA/hPDLCs-sEVs and GelMA/hGFs-sEVs), scaffolds. hBFP-MSCs seeded on the bioprinted sEVs constructs spread well with significantly enhanced focal adhesion, mechanotransduction associated gene expression, and ligament and osteogenesis/cementogenesis differentiation markers in GelMA/hPDLCs-sEVs, compared to GelMA/hGFs-sEVs and GelMA groups. A 2-week osteogenic and ligamentous differentiation showed enhanced ALP staining, calcium formation and toluidine blue stained cells in hBFP-MSCs on bioprinted GelMA/hPDLCs-sEVs constructs compared to the other two groups. The proof-of-concept data from this study supports the notion that 3D bioprinted GelMA/hPDLCs-sEVs scaffolds promote cell attachment, as well as ligamentous, osteogenic and cementogenic differentiation, of hBFP-MSCs in vitro.
Collapse
Affiliation(s)
- Pingping Han
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia.
| | - Nimal Raveendran
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Chun Liu
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Saraswat Basu
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Kexin Jiao
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Nigel Johnson
- The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Corey S Moran
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia
| | - Sašo Ivanovski
- The University of Queensland, School of Dentistry, Center for Orofacial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD 4006, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD 4006, Australia.
| |
Collapse
|
9
|
Liu Y, Xiong W, Li J, Feng H, Jing S, Liu Y, Zhou H, Li D, Fu D, Xu C, He Y, Ye Q. Application of dental pulp stem cells for bone regeneration. Front Med (Lausanne) 2024; 11:1339573. [PMID: 38487022 PMCID: PMC10938947 DOI: 10.3389/fmed.2024.1339573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/15/2024] [Indexed: 03/17/2024] Open
Abstract
Bone defects resulting from severe trauma, tumors, inflammation, and other factors are increasingly prevalent. Stem cell-based therapies have emerged as a promising alternative. Dental pulp stem cells (DPSCs), sourced from dental pulp, have garnered significant attention owing to their ready accessibility and minimal collection-associated risks. Ongoing investigations into DPSCs have revealed their potential to undergo osteogenic differentiation and their capacity to secrete a diverse array of ontogenetic components, such as extracellular vesicles and cell lysates. This comprehensive review article aims to provide an in-depth analysis of DPSCs and their secretory components, emphasizing extraction techniques and utilization while elucidating the intricate mechanisms governing bone regeneration. Furthermore, we explore the merits and demerits of cell and cell-free therapeutic modalities, as well as discuss the potential prospects, opportunities, and inherent challenges associated with DPSC therapy and cell-free therapies in the context of bone regeneration.
Collapse
Affiliation(s)
- Ye Liu
- Center of Regenerative Medicine, Department of Stomatology Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Xiong
- Center of Regenerative Medicine, Department of Stomatology Renmin Hospital of Wuhan University, Wuhan, China
| | - Junyi Li
- Center of Regenerative Medicine, Department of Stomatology Renmin Hospital of Wuhan University, Wuhan, China
| | - Huixian Feng
- Center of Regenerative Medicine, Department of Stomatology Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuili Jing
- Center of Regenerative Medicine, Department of Stomatology Renmin Hospital of Wuhan University, Wuhan, China
| | - Yonghao Liu
- Center of Regenerative Medicine, Department of Stomatology Renmin Hospital of Wuhan University, Wuhan, China
| | - Heng Zhou
- Center of Regenerative Medicine, Department of Stomatology Renmin Hospital of Wuhan University, Wuhan, China
| | - Duan Li
- Center of Regenerative Medicine, Department of Stomatology Renmin Hospital of Wuhan University, Wuhan, China
| | - Dehao Fu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun Xu
- Sydney Dental School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital of Wuhan University of Science and Technology, Wuhan, China
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Qingsong Ye
- Center of Regenerative Medicine, Department of Stomatology Renmin Hospital of Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Taninokuchi Tomassoni M, Zhou Y, Braccischi L, Modestino F, Fukuda J, Mosconi C. Trans-Arterial Stem Cell Injection (TASI): The Role of Interventional Radiology in Regenerative Medicine. J Clin Med 2024; 13:910. [PMID: 38337604 PMCID: PMC10856532 DOI: 10.3390/jcm13030910] [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: 12/13/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Regenerative medicine is taking a step forward in treating multiple diseases. The possibility of renewing damaged tissues with stem cells has become a topic of interest in recent decades. Still a relatively new research topic, many issues in this discipline are being addressed, from cell culturing to the study of different graft materials, and, moreover, cell delivery. For instance, direct intravenous injection has a big downfall regarding its lack of precision and poorly targeted treatment. Trans-arterial and direct percutaneous infusion to the aimed tissue/organ are both considered ideal for reaching the desired region but require image guidance to be performed safely and precisely. In this context, interventional radiology becomes pivotal for providing different cell delivery possibilities in every case. In this review, we analyze different basic stem cell therapy concepts and the current and future role of interventional radiology with a focus on trans-arterial delivery.
Collapse
Affiliation(s)
- Makoto Taninokuchi Tomassoni
- Department of Radiology, IRRCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (L.B.)
| | - Yinghui Zhou
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama 240-8501, Kanagawa, Japan (J.F.)
| | - Lorenzo Braccischi
- Department of Radiology, IRRCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (L.B.)
| | - Francesco Modestino
- Department of Radiology, IRRCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (L.B.)
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama 240-8501, Kanagawa, Japan (J.F.)
| | - Cristina Mosconi
- Department of Radiology, IRRCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (L.B.)
| |
Collapse
|
11
|
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
|
12
|
Teli P, Vaidya A, Kale V. Signal transduction pathways alter the molecular cargo of extracellular vesicles: implications in regenerative medicine. Regen Med 2023; 18:935-944. [PMID: 38059320 DOI: 10.2217/rme-2023-0183] [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] [Indexed: 12/08/2023] Open
Abstract
Extracellular vesicles (EVs) possess regenerative properties and are also considered as future vaccines. All types of cells secrete EVs; however, the amount of EVs secreted by the cells varies under various physiological as well as pathological states. Several articles have reviewed the molecular composition and potential therapeutic applications of EVs. Likewise, the 'sorting signals' associated with specific macromolecules have also been identified, but how the signal transduction pathways prevailing in the parent cells alter the molecular profile of the EVs or the payload they carry has not been sufficiently reviewed. Here, we have specifically discussed the implications of these alterations in the macromolecular cargo of EVs for their therapeutic applications in regenerative medicine.
Collapse
Affiliation(s)
- Prajakta Teli
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune 412115, India
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune 412115, India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune 412115, India
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune 412115, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune 412115, India
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune 412115, India
| |
Collapse
|
13
|
Liu X, Shi S, Geng X, Wang E, Meng Q, Li M, Lin F, Ma X, Han W, Zhou X. Extracellular vesicles derived from different tissues attenuate cardiac dysfunction in murine MI models. Biol Direct 2023; 18:76. [PMID: 37978390 PMCID: PMC10655353 DOI: 10.1186/s13062-023-00429-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) derived from various cell sources exert cardioprotective effects during cardiac ischemic injury. Our previous study confirmed that EVs derived from ischemic-reperfusion injured heart tissue aggravated cardiac inflammation and dysfunction. However, the role of EVs derived from normal cardiac tissue in myocardial ischemic injury remains elusive. RESULTS In the present study, normal heart-derived EVs (cEVs) and kidney-derived EVs (nEVs) were isolated and intramyocardially injected into mice after myocardial infarction (MI). We demonstrated that administration of both cEVs and nEVs significantly improved cardiac function, reduced the scar size, and alleviated inflammatory infiltration into the heart. In addition, cardiomyocyte apoptosis was inhibited, whereas angiogenesis was enhanced in the hearts receiving cEVs or nEVs treatment. Moreover, intramyocardial injection of cEVs displayed much better cardiac protective efficacy than nEVs in murine MI models. RNA-seq and protein-protein interaction (PPI) network analysis revealed the protective mRNA clusters in both cEVs and nEVs. These mRNAs were involved in multiple signaling pathways, which may synergistically orchestrate to prevent the heart from further damage post MI. CONCLUSIONS Collectively, our results indicated that EVs derived from normal heart tissue may represent a promising strategy for cardiac protection in ischemic heart diseases.
Collapse
Affiliation(s)
- Xuan Liu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shanshan Shi
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Department of Pathology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xuedi Geng
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Enhao Wang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qingshu Meng
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Mimi Li
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Fang Lin
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoxue Ma
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wei Han
- Department of Heart Failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
14
|
Forteza-Genestra MA, Antich-Rosselló M, Ramis-Munar G, Calvo J, Gayà A, Monjo M, Ramis JM. Comparative effect of platelet- and mesenchymal stromal cell-derived extracellular vesicles on human cartilage explants using an ex vivo inflammatory osteoarthritis model. Bone Joint Res 2023; 12:667-676. [PMID: 37852621 PMCID: PMC10584413 DOI: 10.1302/2046-3758.1210.bjr-2023-0109.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2023] Open
Abstract
Aims Extracellular vesicles (EVs) are nanoparticles secreted by all cells, enriched in proteins, lipids, and nucleic acids related to cell-to-cell communication and vital components of cell-based therapies. Mesenchymal stromal cell (MSC)-derived EVs have been studied as an alternative for osteoarthritis (OA) treatment. However, their clinical translation is hindered by industrial and regulatory challenges. In contrast, platelet-derived EVs might reach clinics faster since platelet concentrates, such as platelet lysates (PL), are already used in therapeutics. Hence, we aimed to test the therapeutic potential of PL-derived extracellular vesicles (pEVs) as a new treatment for OA, which is a degenerative joint disease of articular cartilage and does not have any curative or regenerative treatment, by comparing its effects to those of human umbilical cord MSC-derived EVs (cEVs) on an ex vivo OA-induced model using human cartilage explants. Methods pEVs and cEVs were isolated by size exclusion chromatography (SEC) and physically characterized by nanoparticle tracking analysis (NTA), protein content, and purity. OA conditions were induced in human cartilage explants (10 ng/ml oncostatin M and 2 ng/ml tumour necrosis factor alpha (TNFα)) and treated with 1 × 109 particles of pEVs or cEVs for 14 days. Then, DNA, glycosaminoglycans (GAG), and collagen content were quantified, and a histological study was performed. EV uptake was monitored using PKH26 labelled EVs. Results Significantly higher content of DNA and collagen was observed for the pEV-treated group compared to control and cEV groups. No differences were found in GAG quantification nor in EVs uptake within any treated group. Conclusion In conclusion, pEVs showed better performance than cEVs in our in vitro OA model. Although further studies are needed, pEVs are shown as a potential alternative to cEVs for cell-free regenerative medicine.
Collapse
Affiliation(s)
- Maria A. Forteza-Genestra
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Miquel Antich-Rosselló
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Guillem Ramis-Munar
- Microscopy Area, Serveis Cietificotècnics, University of the Balearic Islands, Palma, Spain
| | - Javier Calvo
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Fundació Banc de Sang i Teixits de les Illes Balears (FBSTIB), Palma, Spain
| | - Antoni Gayà
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Fundació Banc de Sang i Teixits de les Illes Balears (FBSTIB), Palma, Spain
| | - Marta Monjo
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Joana M. Ramis
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| |
Collapse
|
15
|
Hou Y, Wang J, Wang J. Engineered biomaterial delivery strategies are used to reduce cardiotoxicity in osteosarcoma. Front Pharmacol 2023; 14:1284406. [PMID: 37854721 PMCID: PMC10579615 DOI: 10.3389/fphar.2023.1284406] [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: 08/28/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents. Chemotherapy drugs play an integral role in OS treatment. Preoperative neoadjuvant chemotherapy and postoperative conventional adjuvant chemotherapy improve survival in patients with OS. However, the toxic side effects of chemotherapy drugs are unavoidable. Cardiotoxicity is one of the common side effects of chemotherapy drugs that cannot be ignored. Chemotherapy drugs affect the destruction of mitochondrial autophagy and mitochondria-associated proteins to cause a decrease in cardiac ejection fraction and cardiomyocyte necrosis, which in turn causes heart failure and irreversible cardiomyopathy. Biomaterials play an important role in nanomedicine. Biomaterials act as carriers to deliver chemotherapy drugs precisely around tumor cells and continuously release carriers around the tumor. It not only promotes anti-tumor effects but also reduces the cardiotoxicity of chemotherapy drugs. In this paper, we first introduce the mechanism by which chemotherapy drugs commonly used in OS cause cardiotoxicity. Subsequently, we introduce biomaterials for reducing cardiotoxicity in OS chemotherapy. Finally, we prospect biomaterial delivery strategies to reduce cardiotoxicity in OS.
Collapse
Affiliation(s)
| | | | - Jianping Wang
- Department of Cardiology, Guangyuan Central Hospital, Guangyuan, China
| |
Collapse
|
16
|
Riccio M, Bondioli E, Senesi L, Zingaretti N, Gargiulo P, De Francesco F, Parodi PC, Zavan B. Fragmented Dermo-Epidermal Units (FdeU) as an Emerging Strategy to Improve Wound Healing Process: An In Vitro Evaluation and a Pilot Clinical Study. J Clin Med 2023; 12:6165. [PMID: 37834809 PMCID: PMC10573238 DOI: 10.3390/jcm12196165] [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: 07/07/2023] [Revised: 09/06/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Innovative strategies have shown beneficial effects in healing wound management involving, however, a time-consuming and arduous process in clinical contexts. Micro-fragmented skin tissue acts as a slow-released natural scaffold and continuously delivers growth factors, and much other modulatory information, into the microenvironment surrounding damaged wounds by a paracrine function on the resident cells which supports the regenerative process. In this study, in vitro and in vivo investigations were conducted to ascertain improved effectiveness and velocity of the wound healing process with the application of fragmented dermo-epidermal units (FdeU), acquired via a novel medical device (Hy-Tissue® Micrograft Technology). MTT test; LDH test; ELISA for growth factor investigation (IL) IL-2, IL-6, IL-7 IL-8, IL-10; IGF-1; adiponectin; Fibroblast Growth Factor (FGF); Vascular Endothelial Growth Factor (VEGF); and Tumor Necrosis Factor (TNF) were assessed. Therefore, clinical evaluation in 11 patients affected by Chronic Wounds (CW) and treated with FdeU were investigated. Functional outcome was assessed pre-operatory, 2 months after treatment (T0), and 6 months after treatment (T1) using the Wound Bed Score (WBS) and Vancouver Scar Scale (VSS). In this current study, we demonstrate the potential of resident cells to proliferate from the clusters of FdeU seeded in a monolayer that efficiently propagate the chronic wound. Furthermore, in this study we report how the discharge of trophic/reparative proteins are able to mediate the in vitro paracrine function of proliferation, migration, and contraction rate in fibroblasts and keratinocytes. Our investigations recommend FdeU as a favorable tool in wound healing, displaying in vitro growth-promoting potential to enhance current therapeutic mechanisms.
Collapse
Affiliation(s)
- Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, University Hospital (AOU Ospedali Riuniti di Ancona), Via Conca 71, Torrette di Ancona, 60123 Ancona, Italy; (M.R.); (L.S.); (F.D.F.)
| | - Elena Bondioli
- Burn Center and Emilia Romagna Regional Skin Bank, Bufalini Hospital, AUSL della Romagna, 47521 Cesena, Italy;
| | - Letizia Senesi
- Department of Reconstructive Surgery and Hand Surgery, University Hospital (AOU Ospedali Riuniti di Ancona), Via Conca 71, Torrette di Ancona, 60123 Ancona, Italy; (M.R.); (L.S.); (F.D.F.)
| | - Nicola Zingaretti
- Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy; (N.Z.); (P.C.P.)
| | - Paolo Gargiulo
- Engineering Department, King’s College, London WC2R 2LS, UK;
- Institute for Biomedical and Neural Engineering, Reykjavík University, 101 Reykjavík, Iceland
| | - Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, University Hospital (AOU Ospedali Riuniti di Ancona), Via Conca 71, Torrette di Ancona, 60123 Ancona, Italy; (M.R.); (L.S.); (F.D.F.)
| | - Pier Camillo Parodi
- Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy; (N.Z.); (P.C.P.)
| | - Barbara Zavan
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
| |
Collapse
|
17
|
Wang Z, Knight R, Stephens P, Ongkosuwito EM, Wagener FADTG, Von den Hoff JW. Stem cells and extracellular vesicles to improve preclinical orofacial soft tissue healing. Stem Cell Res Ther 2023; 14:203. [PMID: 37580820 PMCID: PMC10426149 DOI: 10.1186/s13287-023-03423-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 07/20/2023] [Indexed: 08/16/2023] Open
Abstract
Orofacial soft tissue wounds caused by surgery for congenital defects, trauma, or disease frequently occur leading to complications affecting patients' quality of life. Scarring and fibrosis prevent proper skin, mucosa and muscle regeneration during wound repair. This may hamper maxillofacial growth and speech development. To promote the regeneration of injured orofacial soft tissue and attenuate scarring and fibrosis, intraoral and extraoral stem cells have been studied for their properties of facilitating maintenance and repair processes. In addition, the administration of stem cell-derived extracellular vesicles (EVs) may prevent fibrosis and promote the regeneration of orofacial soft tissues. Applying stem cells and EVs to treat orofacial defects forms a challenging but promising strategy to optimize treatment. This review provides an overview of the putative pitfalls, promises and the future of stem cells and EV therapy, focused on orofacial soft tissue regeneration.
Collapse
Affiliation(s)
- Zhihao Wang
- Department of Dentistry, Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Centre, 6525EX, Nijmegen, The Netherlands
| | - Rob Knight
- Stein Eye Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Phil Stephens
- Advanced Therapeutics Group, School of Dentistry, Cardiff University, Cardiff, Wales, UK
| | - E M Ongkosuwito
- Department of Dentistry, Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Centre, 6525EX, Nijmegen, The Netherlands
| | - Frank A D T G Wagener
- Department of Dentistry, Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Centre, 6525EX, Nijmegen, The Netherlands
| | - Johannes W Von den Hoff
- Department of Dentistry, Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Centre, 6525EX, Nijmegen, The Netherlands.
| |
Collapse
|
18
|
Cieślik M, Bryniarski K, Nazimek K. Biodelivery of therapeutic extracellular vesicles: should mononuclear phagocytes always be feared? Front Cell Dev Biol 2023; 11:1211833. [PMID: 37476156 PMCID: PMC10354279 DOI: 10.3389/fcell.2023.1211833] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023] Open
Abstract
At present, extracellular vesicles (EVs) are considered key candidates for cell-free therapies, including treatment of allergic and autoimmune diseases. However, their therapeutic effectiveness, dependent on proper targeting to the desired cells, is significantly limited due to the reduced bioavailability resulting from their rapid clearance by the cells of the mononuclear phagocyte system (MPS). Thus, developing strategies to avoid EV elimination is essential when applying them in clinical practice. On the other hand, malfunctioning MPS contributes to various immune-related pathologies. Therapeutic reversal of these effects with EVs would be beneficial and could be achieved, for example, by modulating the macrophage phenotype or regulating antigen presentation by dendritic cells. Additionally, intended targeting of EVs to MPS macrophages for replication and repackaging of their molecules into new vesicle subtype can allow for their specific targeting to appropriate populations of acceptor cells. Herein, we briefly discuss the under-explored aspects of the MPS-EV interactions that undoubtedly require further research in order to accelerate the therapeutic use of EVs.
Collapse
Affiliation(s)
| | | | - Katarzyna Nazimek
- Department of Immunology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
19
|
O’Brien TJ, Hollinshead F, Goodrich LR. Extracellular vesicles in the treatment and prevention of osteoarthritis: can horses help us translate this therapy to humans? EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:151-169. [PMID: 37829144 PMCID: PMC10568983 DOI: 10.20517/evcna.2023.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Osteoarthritis (OA) is a common joint disease affecting humans and horses, resulting in significant morbidity, financial expense, and loss of athletic use. While the pathogenesis is incompletely understood, inflammation is considered crucial in the development and progression of the disease. Mesenchymal stromal cells (MSCs) have received increasing scientific attention for their anti-inflammatory, immunomodulatory, and pro-regenerative effects. However, there are concerns about their ability to become a commercially available therapeutic. Extracellular vesicles (EVs) are now recognized to play a crucial role in the therapeutic efficacy observed with MSCs and offer a potentially novel cell-free therapeutic that may negate many of the concerns with MSCs. There is evidence that EVs have profound anti-inflammatory, immunomodulatory, and pro-regenerative effects equal to or greater than the MSCs they are derived from in the treatment of OA. Most of these studies are in small animal models, limiting the translation of these results to humans. However, highly translational animal models are crucial for further understanding the efficacy of potential therapeutics and for close comparisons with humans. For this reason, the horse, which experiences the same gravitational impacts on joints similar to people, is a highly relevant large animal species for testing. The equine species has well-designed and validated OA models, and additionally, therapies can be further tested in naturally occurring OA to validate preclinical model testing. Therefore, the horse is a highly suitable model to increase our knowledge of the therapeutic potential of EVs.
Collapse
Affiliation(s)
- Thomas J O’Brien
- Department of Clinical Sciences, Veterinary Teaching Hospital, Colorado State University, Fort Collins, CO 80523, USA
| | - Fiona Hollinshead
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Laurie R Goodrich
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
20
|
Ock J, Wu J, Liu FY, Fridayana FR, Niloofar L, Vo MN, Hong SS, Kang JH, Suh JK, Yin GN, Jin HR, Ryu JK. Heme-binding protein 1 delivered via pericyte-derived extracellular vesicles improves neurovascular regeneration in a mouse model of cavernous nerve injury. Int J Biol Sci 2023; 19:2663-2677. [PMID: 37324943 PMCID: PMC10266087 DOI: 10.7150/ijbs.81809] [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: 12/13/2022] [Accepted: 05/05/2023] [Indexed: 06/17/2023] Open
Abstract
As a peripheral nerve injury disease, cavernous nerve injury (CNI) caused by prostate cancer surgery and other pelvic surgery causes organic damage to cavernous blood vessels and nerves, thereby significantly attenuating the response to phosphodiesterase-5 inhibitors. Here, we investigated the role of heme-binding protein 1 (Hebp1) in erectile function using a mouse model of bilateral CNI, which is known to promote angiogenesis and improve erection in diabetic mice. We found a potent neurovascular regenerative effect of Hebp1 in CNI mice, demonstrating that exogenously delivered Hebp1 improved erectile function by promoting the survival of cavernous endothelial-mural cells and neurons. We further found that endogenous Hebp1 delivered by mouse cavernous pericyte (MCP)-derived extracellular vesicles promoted neurovascular regeneration in CNI mice. Moreover, Hebp1 achieved these effects by reducing vascular permeability through regulation of claudin family proteins. Our findings provide new insights into Hebp1 as a neurovascular regeneration factor and demonstrate its potential therapeutic application to various peripheral nerve injuries.
Collapse
Affiliation(s)
- Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Jitao Wu
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, People's Republic of China
| | - Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Lashkari Niloofar
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Program in Biomedical Science & Engineering, Inha University, Incheon, Korea
| | - Ju-Hee Kang
- Department of Pharmacology, Medicinal Toxicology Research Center, Inha University College of Medicine, Incheon, Korea
| | - Jun-Kyu Suh
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| | - Hai-Rong Jin
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, People's Republic of China
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, 22332, Republic of Korea
| |
Collapse
|
21
|
Lopes D, Lopes J, Pereira-Silva M, Peixoto D, Rabiee N, Veiga F, Moradi O, Guo ZH, Wang XD, Conde J, Makvandi P, Paiva-Santos AC. Bioengineered exosomal-membrane-camouflaged abiotic nanocarriers: neurodegenerative diseases, tissue engineering and regenerative medicine. Mil Med Res 2023; 10:19. [PMID: 37101293 PMCID: PMC10134679 DOI: 10.1186/s40779-023-00453-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 04/07/2023] [Indexed: 04/28/2023] Open
Abstract
A bio-inspired strategy has recently been developed for camouflaging nanocarriers with biomembranes, such as natural cell membranes or subcellular structure-derived membranes. This strategy endows cloaked nanomaterials with improved interfacial properties, superior cell targeting, immune evasion potential, and prolonged duration of systemic circulation. Here, we summarize recent advances in the production and application of exosomal membrane-coated nanomaterials. The structure, properties, and manner in which exosomes communicate with cells are first reviewed. This is followed by a discussion of the types of exosomes and their fabrication methods. We then discuss the applications of biomimetic exosomes and membrane-cloaked nanocarriers in tissue engineering, regenerative medicine, imaging, and the treatment of neurodegenerative diseases. Finally, we appraise the current challenges associated with the clinical translation of biomimetic exosomal membrane-surface-engineered nanovehicles and evaluate the future of this technology.
Collapse
Affiliation(s)
- Daniela Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Joana Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, 6150, Australia
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Omid Moradi
- Department of Chemistry, Shahr-e-Qods Branch, Islamic Azad University, Tehran, 374-37515, Iran
| | - Zhan-Hu Guo
- Integrated Composites Laboratory (ICL), Department of Mechanical and Construction Engineering, Northumbria University, Newcastle Upon Tyne, NE1 8ST, UK
| | - Xiang-Dong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, 200032, China.
| | - João Conde
- Faculdade de Ciências Médicas, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
- Centre for Toxicogenomics and Human Health, Genetics, Oncology and Human Toxicology, Faculdade de Ciências Médicas, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
| | - Pooyan Makvandi
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal.
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal.
| |
Collapse
|
22
|
Sen S, Xavier J, Kumar N, Ahmad MZ, Ranjan OP. Exosomes as natural nanocarrier-based drug delivery system: recent insights and future perspectives. 3 Biotech 2023; 13:101. [PMID: 36860361 PMCID: PMC9970142 DOI: 10.1007/s13205-023-03521-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/13/2023] [Indexed: 03/03/2023] Open
Abstract
Exosomes are nanosized (size ~ 30-150 nm) natural vesicular structures released from cells by physiological processes or pathological circumstances. Exosomes are growing in popularity as a result of their many benefits over conventional nanovehicles, including their ability to escape homing in the liver or metabolic destruction and their lack of undesired accumulation before reaching their intended targets. Various therapeutic molecules, including nucleic acids, have been incorporated into exosomes by different techniques, many of which have shown satisfactory performance in various diseases. Surface-modified exosomes are a potentially effective strategy, and it increases the circulation time and produces the specific drug target vehicle. In this comprehensive review, we describe composition exosomes biogenesis and the role of exosomes in intercellular signaling and cell-cell communications, immune responses, cellular homeostasis, autophagy, and infectious diseases. In addition, we discuss the role of exosomes as diagnostic markers, and their therapeutic and clinical implications. Furthermore, we addressed the challenges and outstanding developments in exosome research and discuss future perspectives. In addition to the current status of exosomes as a therapeutic carrier, the lacuna in the clinical development lifecycles along with the possible strategies to fill the lacuna have been addressed.
Collapse
Affiliation(s)
- Srijita Sen
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101 India
| | - Joyal Xavier
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar 844102 India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar 844102 India
| | - Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, 11001 Kingdom of Saudi Arabia
| | - Om Prakash Ranjan
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101 India
| |
Collapse
|
23
|
Mansour A, Romani M, Acharya AB, Rahman B, Verron E, Badran Z. Drug Delivery Systems in Regenerative Medicine: An Updated Review. Pharmaceutics 2023; 15:pharmaceutics15020695. [PMID: 36840018 PMCID: PMC9967372 DOI: 10.3390/pharmaceutics15020695] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Modern drug discovery methods led to evolving new agents with significant therapeutic potential. However, their properties, such as solubility and administration-related challenges, may hinder their benefits. Moreover, advances in biotechnology resulted in the development of a new generation of molecules with a short half-life that necessitates frequent administration. In this context, controlled release systems are required to enhance treatment efficacy and improve patient compliance. Innovative drug delivery systems are promising tools that protect therapeutic proteins and peptides against proteolytic degradation where controlled delivery is achievable. The present review provides an overview of different approaches used for drug delivery.
Collapse
Affiliation(s)
- Alaa Mansour
- Periodontology Unit, College of Dental Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Maya Romani
- Department of Family Medicine, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | | | - Betul Rahman
- Periodontology Unit, College of Dental Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence:
| | - Elise Verron
- CNRS, CEISAM, UMR 6230, Nantes Université, F-44000 Nantes, France
| | - Zahi Badran
- Periodontology Unit, College of Dental Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
24
|
Cha KY, Cho W, Park S, Ahn J, Park H, Baek I, Lee M, Lee S, Arai Y, Lee SH. Generation of bioactive MSC-EVs for bone tissue regeneration by tauroursodeoxycholic acid treatment. J Control Release 2023; 354:45-56. [PMID: 36586671 DOI: 10.1016/j.jconrel.2022.12.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 01/02/2023]
Abstract
Extracellular vesicles (EVs) are nano-sized carriers that reflect the parent cell's information and are known to mediate cell-cell communication. In order to overcome the disadvantages of mesenchymal stem cells (MSCs) in cell therapy, such as unexpected differentiation leading to tumorization, immune rejection, and other side effects, EVs derived from MSCs (MSC-EVs) with the tissue regenerative function have been studied as new cell-free therapeutics. However, therapeutic applications of EVs require overcoming several challenges. First, the production efficiency of MSC-EVs should be increased at least as much as the quantity of them are required to their clinical application; second, MSC-EVs needs to show various functionality further, thereby increasing tissue regeneration efficiency. In this study, we treated tauroursodeoxycholic acid (TUDCA), a biological derivative known to regulate cholesterol, to MSCs and investigated whether TUDCA treatment would be able to increase EV production efficiency and tissue regenerative capacity of EVs. Indeed, it appears that TUDCA priming to MSC increases the yield of MSC-EVs >2 times by reducing the cellular cholesterol level in MSCs and increasing the exocytosis-related CAV1 expression. Interestingly, it was found that the EVs derived from TUDCA-primed MSCs (T-EV) contained higher amounts of anti-inflammatory cytokines (IL1RN, IL6, IL10, and IL11) and osteogenic proteins (ALP, RUNX2, BMP2, BMPR1, and BMPR2) than those in control MSC-EVs (C-EV). Besides, it was shown that T-EV not only regulated M1/M2 macrophages differentiation of monocytes, also effectively increased the osteogenic differentiation of MSCs as well as bone tissue regeneration in a bone defect rat model. Based on these results, it is concluded that TUDCA treatment to MSC as a new approach endows EV with high-yield production and functionality. Thus, we strongly believe T-EV would be a powerful therapeutic material for bone tissue regeneration and potentially could be expanded to other types of tissue regeneration for clinical applications.
Collapse
Affiliation(s)
- Kyung-Yup Cha
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Woongjin Cho
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Sunghyun Park
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Jinsung Ahn
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Hyoeun Park
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Inho Baek
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Minju Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Sunjun Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Yoshie Arai
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea.
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea.
| |
Collapse
|
25
|
Rajendran RL, Gangadaran P, Kwack MH, Oh JM, Hong CM, Jeyaraman M, Sung YK, Lee J, Ahn BC. Engineered Nanovesicles from Fibroblasts Modulate Dermal Papillae Cells In Vitro and Promote Human Hair Follicle Growth Ex Vivo. Cells 2022; 11:4066. [PMID: 36552830 PMCID: PMC9777471 DOI: 10.3390/cells11244066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/27/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Alopecia is a common medical condition affecting both sexes. Dermal papilla (DP) cells are the primary source of hair regeneration in alopecia patients. Therapeutic applications of extracellular vesicles (EVs) are restricted by low yields, high costs, and their time-consuming collection process. Thus, engineered nanovesicles (eNVs) have emerged as suitable therapeutic biomaterials in translational medicine. We isolated eNVs by the serial extrusion of fibroblasts (FBs) using polycarbonate membrane filters and serial and ultracentrifugation. We studied the internalization, proliferation, and migration of human DP cells in the presence and absence of FB-eNVs. The therapeutic potential of FB-eNVs was studied on ex vivo organ cultures of human hair follicles (HFs) from three human participants. FB-eNVs (2.5, 5, 7.5, and 10 µg/mL) significantly enhanced DP cell proliferation, with the maximum effect observed at 7.5 µg/mL. FB-eNVs (5 and 10 µg/mL) significantly enhanced the migration of DP cells at 36 h. Western blotting results suggested that FB-eNVs contain vascular endothelial growth factor (VEGF)-a. FB-eNV treatment increased the levels of PCNA, pAKT, pERK, and VEGF-receptor-2 (VEGFR2) in DP cells. Moreover, FB-eNVs increased the human HF shaft size in a short duration ex vivo. Altogether, FB-eNVs are promising therapeutic candidates for alopecia.
Collapse
Affiliation(s)
- Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Mi Hee Kwack
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Immunology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute University, Chennai 600056, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
| | - Young Kwan Sung
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Immunology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| |
Collapse
|
26
|
Birjandi AA, Sharpe P. Potential of extracellular space for tissue regeneration in dentistry. Front Physiol 2022; 13:1034603. [DOI: 10.3389/fphys.2022.1034603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/24/2022] [Indexed: 11/19/2022] Open
Abstract
With the proven relationship between oral and general health and the growing aging population, it is pivotal to provide accessible therapeutic approaches to regenerate oral tissues and restore clinical function. However, despite sharing many core concepts with medicine, dentistry has fallen behind the progress in precision medicine and regenerative treatments. Stem cell therapies are a promising avenue for tissue regeneration, however, ethical, safety and cost issues may limit their clinical use. With the significance of paracrine signalling in stem cell and tissue regeneration, extracellular space comprising of the cell secretome, and the extracellular matrix can serve as a potent source for tissue regeneration. Extravesicles are secreted and naturally occurring vesicles with biologically active cargo that can be harvested from the extracellular space. These vesicles have shown great potential as disease biomarkers and can be used in regenerative medicine. As a cell free therapy, secretome and extracellular vesicles can be stored and transferred easily and pose less ethical and safety risks in clinical application. Since there are currently many reviews on the secretome and the biogenesis, characterization and function of extracellular vesicles, here we look at the therapeutic potential of extracellular space to drive oral tissue regeneration and the current state of the field in comparison to regenerative medicine.
Collapse
|
27
|
Xia Y, Yang R, Hou Y, Wang H, Li Y, Zhu J, Fu C. Application of mesenchymal stem cell-derived exosomes from different sources in intervertebral disc degeneration. Front Bioeng Biotechnol 2022; 10:1019437. [PMID: 36277386 PMCID: PMC9585200 DOI: 10.3389/fbioe.2022.1019437] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 12/12/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is a main cause of lower back pain, leading to psychological and economic burdens to patients. Physical therapy only delays pain in patients but cannot eliminate the cause of IVDD. Surgery is required when the patient cannot tolerate pain or has severe neurological symptoms. Although surgical resection of IVD or decompression of the laminae eliminates the diseased segment, it damages adjacent normal IVD. There is also a risk of re-protrusion after IVD removal. Cell therapy has played a crucial role in the development of regenerative medicine. Cell transplantation promotes regeneration of degenerative tissue. However, owing to the lack of vascular structure in IVD, sufficient nutrients cannot be provided for transplanted mesenchymal stem cells (MSCs). In addition, dead cells release harmful substances that aggravate IVDD. Extracellular vesicles (EVs) have been extensively studied as an emerging therapeutic approach. EVs generated by paracrine MSCs retain the potential of MSCs and serve as carriers to deliver their contents to target cells to regulate target cell activity. Owing to their double-layered membrane structure, EVs have a low immunogenicity and no immune rejection. Therefore, EVs are considered an emerging therapeutic modality in IVDD. However, they are limited by mass production and low loading rates. In this review, the structure of IVD and advantages of EVs are introduced, and the application of MSC-EVs in IVDD is discussed. The current limitations of EVs and future applications are described.
Collapse
Affiliation(s)
- Yuanliang Xia
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ruohan Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yulin Hou
- Department of Cardiology, Guangyuan Central Hospital, Guangyuan, China
| | - Hengyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuehong Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Changfeng Fu,
| |
Collapse
|
28
|
Selenium-Stimulated Exosomes Enhance Wound Healing by Modulating Inflammation and Angiogenesis. Int J Mol Sci 2022; 23:ijms231911543. [PMID: 36232844 PMCID: PMC9570007 DOI: 10.3390/ijms231911543] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/29/2022] Open
Abstract
Mesenchymal stem cell (MSC)-derived exosomes have emerged as an attractive cell-free tool in tissue engineering and regenerative medicine. The current study aimed to examine the anti-inflammatory, pro-angiogenic, and wound-repair effects of both exosomes and selenium-stimulated exosomes, and check whether the latter had superior wound healing capacity over others. The cellular and molecular network of exosomes, as a paracrine signal, was extensively studied by performing miRNA arrays to explore the key mediators of exosomes in wound healing. Selenium is known to play a critical role in enhancing the proliferation, multi-potency, and anti-inflammatory effects of MSCs. Selenium-stimulated exosomes showed significant effects in inhibiting inflammation and improving pro-angiogenesis in human umbilical vein endothelial cells. Cell growth and the migration of human dermal fibroblasts and wound regeneration were more enhanced in the selenium-stimulated exosome group than in the selenium and exosome groups, thereby further promoting the wound healing in vivo. Taken together, selenium was found to augment the therapeutic effects of adipose MSC-derived exosomes in tissue regeneration. We concluded that selenium may be considered a vital agent for wound healing in stem cell-based cell-free therapies.
Collapse
|
29
|
Luo Y, Li Z, Wang X, Wang J, Duan X, Li R, Peng Y, Ye Q, He Y. Characteristics of culture-condition stimulated exosomes or their loaded hydrogels in comparison with other extracellular vesicles or MSC lysates. Front Bioeng Biotechnol 2022; 10:1016833. [PMID: 36185445 PMCID: PMC9523448 DOI: 10.3389/fbioe.2022.1016833] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/05/2022] [Indexed: 12/12/2022] Open
Abstract
Recently, it has become popular to study the use of extracellular vesicles (EVs) secreted by stem cells to repair damaged tissues or lost cells. Various cell types and physiological fluids release EVs, and they play an important role in cell-to-cell communication. Moreover, EVs have been implicated in important processes, such as immune responses, homeostasis maintenance, coagulation, inflammation, cancer progression, angiogenesis, and antigen presentation. Thus, EVs participate in both physiological and pathological progression. The main classes of EVs include exosomes, microvesicles (MVs), and apoptotic bodies (ApoBDs). Exosomes, which carry a mass of signal molecules such as RNA, DNA, proteins, and lipids, are the most important of these EVs subsets. Currently, exosomes are generating substantial interest in the scientific community. Exosomes loaded hydrogels or under different cultural environments exhibit different properties and functions. Therefore, the exosomes obtained from different sources and conditions are worth reviewing. More importantly, no review article has compared the different EVs, such as exosomes, MVs, ApoBDs, and mesenchymal stem cell (MSC) lysates, which are special soluble substances. The differentiation between EVs and MSC lysates is a logical approach. Accordingly, this review provides an update on the latest progress in studying the roles of culture-condition stimulated exosomes or their loaded hydrogels and the differentiation between exosomes, MVs, ApoBDs, and MSC lysates. Published studies were retrieved from the PubMed® database for review.
Collapse
Affiliation(s)
- Yu Luo
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihua Li
- Department of Orthodontics, School and Hospital of Stomatology, Nanchang University, Nanchang, China
| | - Xinxin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Juan Wang
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xingxiang Duan
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruohan Li
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Youjian Peng
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingsong Ye
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Orthodontics, School and Hospital of Stomatology, Nanchang University, Nanchang, China
- *Correspondence: Qingsong Ye, ; Yan He,
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- *Correspondence: Qingsong Ye, ; Yan He,
| |
Collapse
|
30
|
Prospects and Challenges of Electrospun Cell and Drug Delivery Vehicles to Correct Urethral Stricture. Int J Mol Sci 2022; 23:ijms231810519. [PMID: 36142432 PMCID: PMC9502833 DOI: 10.3390/ijms231810519] [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: 07/30/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Current therapeutic modalities to treat urethral strictures are associated with several challenges and shortcomings. Therefore, significant strides have been made to develop strategies with minimal side effects and the highest therapeutic potential. In this framework, electrospun scaffolds incorporated with various cells or bioactive agents have provided promising vistas to repair urethral defects. Due to the biomimetic nature of these constructs, they can efficiently mimic the native cells’ niches and provide essential microenvironmental cues for the safe transplantation of multiple cell types. Furthermore, these scaffolds are versatile platforms for delivering various drug molecules, growth factors, and nucleic acids. This review discusses the recent progress, applications, and challenges of electrospun scaffolds to deliver cells or bioactive agents during the urethral defect repair process. First, the current status of electrospinning in urethral tissue engineering is presented. Then, the principles of electrospinning in drug and cell delivery applications are reviewed. Finally, the recent preclinical studies are summarized and the current challenges are discussed.
Collapse
|
31
|
Dai Z, Li Z, Zheng W, Yan Z, Zhang L, Yang J, Xiao J, Sun H, Li S, Huang W. Gallic Acid Ameliorates the Inflammatory State of Periodontal Ligament Stem Cells and Promotes Pro-Osteodifferentiation Capabilities of Inflammatory Stem Cell-Derived Exosomes. Life (Basel) 2022; 12:1392. [PMID: 36143428 PMCID: PMC9501550 DOI: 10.3390/life12091392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/22/2022] [Accepted: 09/02/2022] [Indexed: 11/20/2022] Open
Abstract
The slow proliferation rate and poor osteodifferentiation ability of inflammatory periodontal membrane stem cells extracted from periodontitis tissues (i-PDLSCs) account for poor efficiency in treating inflammatory bone loss. Exosomes reportedly have inducible and relatively stable components, allowing them to promote inflammatory bone repair, but obtaining i-PDLSCs exosomes with the ability to promote osteodifferentiation is challenging. In the present study, i-PDLSCs were extracted from periodontal membrane tissues of patients with severe periodontitis, and in vitro induction with gallic acid (GA) significantly promoted the proliferative activity of i-PDLSCs at a concentration of 10 mM, with TC0 of 11.057 mM and TC50 of 67.56 mM for i-PDLSCs. After mRNA sequencing, we found that GA could alleviate oxidative stress in i-PDLSCs and increase its mitochondrial membrane potential and glucose aerobic metabolism level, thus promoting the osteodifferentiation of i-PDLSCs. After exosomes of i-PDLSCs after GA induction (i-EXO-GA) were isolated by differential centrifugation, we found that 200 ug/mL of i-EXO-GA could remarkably promote the osteodifferentiation of i-PDLSCs. Overall, our results suggest that GA induction can enhance the proliferation and osteodifferentiation in primary cultures of i-PDLSCs in vitro, mediated by alleviating oxidative stress and glycometabolism levels in cells, which further influences the osteodifferentiation-promoting ability of i-EXO-GA. Overall, we provide a viable cell and exosome induction culture method for treating inflammatory alveolar defects associated with periodontitis.
Collapse
Affiliation(s)
- Zhenning Dai
- Department of Stomatology, Guangdong Key Laboratory of Traditional Chinese Medicine Research and Development, The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangdong Second Traditional Chinese Medicine Hospital, Guangzhou 510095, China
| | - Ziyue Li
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
- Department of Anatomy, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weihan Zheng
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
- Department of Anatomy, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zi Yan
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
- Department of Anatomy, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lijun Zhang
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jiaxin Yang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jing Xiao
- Division of Spine Surgery, Section II, Department of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
| | - Hanxiao Sun
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Shiyu Li
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
- Department of Anatomy, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenhua Huang
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
- Department of Anatomy, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
32
|
Krishnan A, Muthusamy S, Fernandez FB, Kasoju N. Mesenchymal Stem Cell-Derived Extracellular Vesicles in the Management of COVID19-Associated Lung Injury: A Review on Publications, Clinical Trials and Patent Landscape. Tissue Eng Regen Med 2022; 19:659-673. [PMID: 35384633 PMCID: PMC8985390 DOI: 10.1007/s13770-022-00441-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023] Open
Abstract
The unprecedented COVID-19 pandemic situation forced the scientific community to explore all the possibilities from various fields, and so far we have seen a lot of surprises, eureka moments and disappointments. One of the approaches from the cellular therapists was exploiting the immunomodulatory and regenerative potential of mesenchymal stromal cells (MSCs), more so of MSC-derived extracellular vesicles (EVs)-particularly exosomes, in order to alleviate the cytokine storm and regenerate the damaged lung tissues. Unlike MSCs, the EVs are easier to store, deliver, and are previously shown to be as effective as MSCs, yet less immunogenic. These features attracted the attention of many and thus led to a tremendous increase in publications, clinical trials and patent applications. This review presents the current landscape of the field and highlights some interesting findings on MSC-derived EVs in the context of COVID-19, including in silico, in vitro, in vivo and case reports. The data strongly suggests the potential of MSC-derived EVs as a therapeutic regime for the management of acute lung injury and associated complications in COVID-19 and beyond.
Collapse
Affiliation(s)
- Anand Krishnan
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India
| | - Senthilkumar Muthusamy
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India
| | - Francis B Fernandez
- Department of Biomaterial Science and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India
| | - Naresh Kasoju
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India.
| |
Collapse
|
33
|
Tesovnik T, Jenko Bizjan B, Šket R, Debeljak M, Battelino T, Kovač J. Technological Approaches in the Analysis of Extracellular Vesicle Nucleotide Sequences. Front Bioeng Biotechnol 2021; 9:787551. [PMID: 35004647 PMCID: PMC8733665 DOI: 10.3389/fbioe.2021.787551] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Together with metabolites, proteins, and lipid components, the EV cargo consists of DNA and RNA nucleotide sequence species, which are part of the intracellular communication network regulating specific cellular processes and provoking distinct target cell responses. The extracellular vesicle (EV) nucleotide sequence cargo molecules are often investigated in association with a particular pathology and may provide an insight into the physiological and pathological processes in hard-to-access organs and tissues. The diversity and biological function of EV nucleotide sequences are distinct regarding EV subgroups and differ in tissue- and cell-released EVs. EV DNA is present mainly in apoptotic bodies, while there are different species of EV RNAs in all subgroups of EVs. A limited sample volume of unique human liquid biopsy provides a small amount of EVs with limited isolated DNA and RNA, which can be a challenging factor for EV nucleotide sequence analysis, while the additional difficulty is technical variability of molecular nucleotide detection. Every EV study is challenged with its first step of the EV isolation procedure, which determines the EV's purity, yield, and diameter range and has an impact on the EV's downstream analysis with a significant impact on the final result. The gold standard EV isolation procedure with ultracentrifugation provides a low output and not highly pure isolated EVs, while modern techniques increase EV's yield and purity. Different EV DNA and RNA detection techniques include the PCR procedure for nucleotide sequence replication of the molecules of interest, which can undergo a small-input EV DNA or RNA material. The nucleotide sequence detection approaches with their advantages and disadvantages should be considered to appropriately address the study problem and to extract specific EV nucleotide sequence information with the detection using qPCR or next-generation sequencing. Advanced next-generation sequencing techniques allow the detection of total EV genomic or transcriptomic data even at the single-molecule resolution and thus, offering a sensitive and accurate EV DNA or RNA biomarker detection. Additionally, with the processes where the EV genomic or transcriptomic data profiles are compared to identify characteristic EV differences in specific conditions, novel biomarkers could be discovered. Therefore, a suitable differential expression analysis is crucial to define the EV DNA or RNA differences between conditions under investigation. Further bioinformatics analysis can predict molecular cell targets and identify targeted and affected cellular pathways. The prediction target tools with functional studies are essential to help specify the role of the investigated EV-targeted nucleotide sequences in health and disease and support further development of EV-related therapeutics. This review will discuss the biological diversity of human liquid biopsy-obtained EV nucleotide sequences DNA and RNA species reported as potential biomarkers in health and disease and methodological principles of their detection, from human liquid biopsy EV isolation, EV nucleotide sequence extraction, techniques for their detection, and their cell target prediction.
Collapse
Affiliation(s)
- Tine Tesovnik
- Institute for Special Laboratory Diagnostics, University Medical Centre Ljubljana, University Children’s Hospital, Ljubljana, Slovenia
| | - Barbara Jenko Bizjan
- Institute for Special Laboratory Diagnostics, University Medical Centre Ljubljana, University Children’s Hospital, Ljubljana, Slovenia
| | - Robert Šket
- Institute for Special Laboratory Diagnostics, University Medical Centre Ljubljana, University Children’s Hospital, Ljubljana, Slovenia
| | - Maruša Debeljak
- Institute for Special Laboratory Diagnostics, University Medical Centre Ljubljana, University Children’s Hospital, Ljubljana, Slovenia
| | - Tadej Battelino
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, University Children’s Hospital, Ljubljana, Slovenia
- Faculty of Medicine, Chair of Paediatrics, University of Ljubljana, Ljubljana, Slovenia
| | - Jernej Kovač
- Institute for Special Laboratory Diagnostics, University Medical Centre Ljubljana, University Children’s Hospital, Ljubljana, Slovenia
| |
Collapse
|
34
|
Li T, Tan X, Li S, Al-Nusaif M, Le W. Role of Glia-Derived Extracellular Vesicles in Neurodegenerative Diseases. Front Aging Neurosci 2021; 13:765395. [PMID: 34744700 PMCID: PMC8563578 DOI: 10.3389/fnagi.2021.765395] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/28/2021] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs), as nano-sized vesicles secreted by almost all cells, have been recognized as the essential transmitter for cell-to-cell communication and participating in multiple biological processes. Neurodegenerative diseases (ND), such as Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis, share common mechanisms of the aggregation and propagation of distinct pathologic proteins among cells in the nervous systems and neuroinflammatory reactions mediated by glia during the pathogenic process. This feature indicates the vital role of crosstalk between neurons and glia in the pathogenesis of ND. In recent years, glia-derived EVs have been investigated as potential mediators of signals between neurons and glia, which provides a new direction and strategy for understanding ND. By a comprehensive summary, it can be concluded that glia-derived EVs have both a beneficial and/or a detrimental effect in the process of ND. Therefore, this review article conveys the role of glia-derived EVs in the pathogenesis of ND and raises current limitations of their potential application in the diagnosis and treatment of ND.
Collapse
Affiliation(s)
- Tianbai Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiang Tan
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Murad Al-Nusaif
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.,Institute of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|