1
|
Francius G, Audoux K, Gaye I, Guedon E, Chebil L. Impact of Substrate Surface Chemistry on Wharton's Jelly Mesenchymal Stem Cell Morphological Characteristics, Adherence, and Detachment. ACS APPLIED BIO MATERIALS 2025; 8:4033-4045. [PMID: 40278837 DOI: 10.1021/acsabm.5c00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Using a multidisciplinary approach combining cell biology assays and physicochemical analyses, the effect of surface chemistry, usually involved in 2D and 3D cell cultures, on the adherence and detachment of Wharton's jelly mesenchymal stem cells (WJ-MSC) was clearly characterized. Values calculated from AFM measurements showed that the Young's modulus of the cells depends on the coating substrate: diethylaminoethyl-dextran (DEAE-D) and carboxypolystyrene (cPS). The adhesion of WJ-MSC was influenced by the composition, charge, and Young's modulus of the surface. High adhesion was observed on DEAE-D. Increasing the wall shear stress decreased the proportion of attached cells on DEAE-D down to ∼20%. For the cPS-coated glass substrate, the fraction of detached cells was higher than that for the DEAE-D surfaces, and at the end of the experiment (maximum wall shear stress of 1 Pa), almost complete detachment was achieved. The experimental strategy used in this work provides information in the field of surface chemistry and may help in the selection and design of efficient microcarriers for cell attachment and detachment during bioreactor cultivation.
Collapse
Affiliation(s)
| | - Kévin Audoux
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France
| | - Ibrahima Gaye
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France
| | | | - Latifa Chebil
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France
| |
Collapse
|
2
|
Jeong T, Lee MS, Jeon J, Park JH, Chung Y, Yang HS. Advanced stem cell therapy using both cell spheroids transplant and paracrine factor release hydrogel patches for myocardial infarction. Colloids Surf B Biointerfaces 2025; 253:114772. [PMID: 40378458 DOI: 10.1016/j.colsurfb.2025.114772] [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: 02/11/2025] [Revised: 04/25/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
Conventional micro-concave systems have been proposed as effective methods for facile cell spheroid formation, culture. However, these systems face challenges in terms of ease of cell transplantation and a low cell survival rate in ischemic disease. We present a novel open/close type hydrogel micro-concave patch (OC) designed for in situ 3D cell spheroid formation, culture, and a transplantable system utilizing a 3D printed mold. Open-type patches were fabricated with a rigid hydrogel, while closed-type patches were prepared with a combination of swellable soft hydrogel and rigid hydrogel. The open-type concave was intended for cell spheroid formation and subsequent transplantation into the ischemic region. Conversely, the close-type concave allowed released cytokines from cell spheroids, which were located inside the concave, to promote survival of transplanted cell spheroid. We hypothesized that transplant of open-type cell spheroids, combined with the release of paracrine factors from close-type cell spheroids, could enhance therapeutic outcomes in ischemic regions. The OC was prepared using different concentration ratios of swellable polyacrylamide (PAAM) hydrogel through 3D printed micropillar mold. Additionally, PAAM was characterized to enhance the compactness of close-type 3D cell spheroids. Transplantation of OC improved the therapeutic effect in a rat cardiac infarction model compared to open-type patches.
Collapse
Affiliation(s)
- Taekgwang Jeong
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Min Suk Lee
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jin Jeon
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jin Hee Park
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Youngdoo Chung
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Hee Seok Yang
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Department of Biomedical Science & Engineering, Dankook University, Cheonan 31116, Republic of Korea; Center for Bio-Medical Engineering Core-Facility, Dankook University, Cheonan 31116, Republic of Korea.
| |
Collapse
|
3
|
Morawska-Kozłowska M, Pitas M, Zhalniarovich Y. Mesenchymal Stem Cells in Veterinary Medicine-Still Untapped Potential. Animals (Basel) 2025; 15:1175. [PMID: 40282009 PMCID: PMC12024326 DOI: 10.3390/ani15081175] [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: 03/03/2025] [Revised: 04/05/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Mesenchymal stem cells (MSCs) hold significant therapeutic potential in veterinary medicine due to their regenerative and immunomodulatory properties. This review examines the clinical applications of MSCs across multiple animal species, including equine, canine, feline, and bovine medicine. MSC therapies have demonstrated promising outcomes in treating musculoskeletal disorders, osteoarthritis, inflammatory diseases, and tissue injuries, particularly in horses and dogs. In cats, MSCs show potential for managing chronic kidney disease, inflammatory bowel disease, and asthma, while in bovine medicine, they offer alternative treatment approaches for mastitis and orthopedic injuries. Despite these advancements, challenges such as treatment standardization, cell sourcing, and potential adverse effects, including tumorigenicity, remain under investigation. The emerging field of MSC-based veterinary medicine highlights its capacity to enhance healing, reduce inflammation, and improve clinical outcomes. However, further research is necessary to optimize treatment protocols and address safety concerns, ensuring the widespread adoption of MSC therapies in veterinary practice.
Collapse
Affiliation(s)
- Magdalena Morawska-Kozłowska
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Mateusz Pitas
- Veterinary Polyclinic, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Yauheni Zhalniarovich
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| |
Collapse
|
4
|
Deshpande P, Dornbrand-Lo M, Phondge V, Kelly P, Wong AK. Tissue engineering approaches for lymphedema: biomaterial innovations and clinical potential. Front Cell Dev Biol 2025; 13:1537050. [PMID: 40302939 PMCID: PMC12037638 DOI: 10.3389/fcell.2025.1537050] [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: 11/29/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
The lymphatic system plays a critical role in maintaining fluid balance and immune regulation. Lymphedema, and other lymphatic disorders, highlight the need for advanced therapeutic approaches, including tissue engineering. This review examines the latest developments in artificial lymphatic tissue engineering, focusing on scaffold materials, lymphangiogenic factors, and regenerative strategies to replicate the intricacy of lymphatic vessels and nodes. We conducted a thorough literature review of current practices and applications in lymphatic tissue engineering. Findings show that biomaterials such as hydrogels, decellularized matrices, and synthetic polymers provide effective scaffolds for lymphatic endothelial cell proliferation and lymphangiogenesis. Advances in growth factor delivery and stem-cell based therapies have further enhanced the viability of engineered lymphatic tissues. Despite promising progress, challenges in achieving functional replication of lymphatic structures and clinical translation of research remain. Ongoing research must address scaffold biocompatibility, optimized growth factor targeting, and scalable production to advance therapeutic options for lymphatic disorders. This review underscores the potential for transformative patient outcomes through innovative bioengineering solutions.
Collapse
Affiliation(s)
| | | | | | | | - Alex K. Wong
- Rutgers New Jersey Medical School, Division of Plastic and Reconstructive Surgery, Newark, NJ, United States
| |
Collapse
|
5
|
Shah J, Ong J, Lee R, Suh A, Waisberg E, Gibson CR, Berdahl J, Mader TH. Risk of Permanent Corneal Injury in Microgravity: Spaceflight-Associated Hazards, Challenges to Vision Restoration, and Role of Biotechnology in Long-Term Planetary Missions. Life (Basel) 2025; 15:602. [PMID: 40283157 PMCID: PMC12028470 DOI: 10.3390/life15040602] [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: 02/26/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Human space exploration presents an unparalleled opportunity to study life in extreme environments-but it also exposes astronauts to physiological stressors that jeopardize key systems like vision. Corneal health, essential for maintaining precise visual acuity, is threatened by microgravity-induced fluid shifts, cosmic radiation, and the confined nature of spacecraft living environments. These conditions elevate the risk of corneal abrasions, infections, and structural damage. In addition, Spaceflight-Associated Neuro-Ocular Syndrome (SANS)-while primarily affecting the posterior segment-has also been potentially linked to anterior segment alterations such as corneal edema and tear film instability. This review examines these ocular challenges and assesses current mitigation strategies. Traditional approaches, such as terrestrial eye banking and corneal transplantation, are impractical for spaceflight due to the limited viability of preserved tissues, surgical complexities, anesthetic risks, infection potential, and logistical constraints. The paper explores emerging technologies like 3D bioprinting and stem cell-based tissue engineering, which offer promising solutions by enabling the on-demand production of personalized corneal constructs. Complementary advancements, including adaptive protective eyewear, bioengineered tear substitutes, telemedicine, and AI-driven diagnostic tools, also show potential in autonomously managing ocular health during long-duration missions. By addressing the complex interplay of environmental stressors and biological vulnerabilities, these innovations not only safeguard astronaut vision and mission performance but also catalyze new pathways for regenerative medicine on Earth. The evolution of space-based ophthalmic care underscores the dual impact of space medicine investments across planetary exploration and terrestrial health systems.
Collapse
Affiliation(s)
- Jainam Shah
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI 48105, USA
| | - Ryung Lee
- Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Alex Suh
- Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ethan Waisberg
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, UK
| | | | - John Berdahl
- Vance Thompson Vision, Sioux Falls, SD 57105, USA
| | | |
Collapse
|
6
|
Wang Z, Zhong D, Yan T, Zheng Q, Zhou E, Ye Z, He X, Liu Y, Yan J, Yuan Y, Wang Y, Cai X. Stem Cells from Human Exfoliated Deciduous Teeth-Derived Exosomes for the Treatment of Acute Liver Injury and Liver Fibrosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:17948-17964. [PMID: 40087139 PMCID: PMC11955941 DOI: 10.1021/acsami.4c19748] [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] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Mesenchymal stem cells (MSCs) play a crucial role in regenerative medicine due to their regenerative potential. However, traditional MSC-based therapies are hindered by issues such as microvascular obstruction and low cell survival after transplantation. Exosomes derived from MSCs (MSC-Exo) provide a cell-free, nanoscale alternative, mitigating these risks and offering therapeutic potential for liver diseases. Nonetheless, the functional variability of MSCs from different sources complicates their clinical application. Stem cells derived from human exfoliated deciduous teeth (SHED) offer advantages such as ease of procurement and robust proliferative capacity, but their secretome, particularly SHED-Exo, remains underexplored in the context of liver disease therapy. This study analyzed MSC-Exo from various sources via small RNA sequencing to identify differences in microRNA profiles, aiding in the selection of optimal MSC sources for clinical use. SHED-Exo was subsequently tested in an acute liver injury model, showing notable regenerative effects, including enhanced hepatocyte proliferation, macrophage polarization, and reduced inflammation. Despite strong liver-targeting properties, the rapid hepatic clearance of SHED-Exo limits its effectiveness in chronic liver diseases. To address this challenge, a GelMA-based hydrogel was developed for in situ delivery, ensuring sustained release and enhanced antifibrotic efficacy, providing a promising strategy for chronic liver disease management.
Collapse
Affiliation(s)
- Ziyuan Wang
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Danyang Zhong
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Tingting Yan
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Qiang Zheng
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Enjie Zhou
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Zhichao Ye
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xiaoyan He
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yu Liu
- Department
of Cardiac Surgery, Zhejiang University
School of Medicine Sir Run Run Shaw Hospital, Hangzhou 310016, Zhejiang, China
| | - Jianing Yan
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yuyang Yuan
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yifan Wang
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- National
Engineering Research Center of Innovation and Application of Minimally
Invasive Instruments, Hangzhou 310016, China
- Zhejiang
Minimal Invasive Diagnosis and Treatment Technology Research Center
of Severe Hepatobiliary Disease, Hangzhou 310016, China
| | - Xiujun Cai
- Department
of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- National
Engineering Research Center of Innovation and Application of Minimally
Invasive Instruments, Hangzhou 310016, China
- Zhejiang
Minimal Invasive Diagnosis and Treatment Technology Research Center
of Severe Hepatobiliary Disease, Hangzhou 310016, China
| |
Collapse
|
7
|
Jin G, Porello EAL, Zhang J, Lim B. Heterogeneous Sox2 transcriptional dynamics mediate pluripotency maintenance in mESCs in response to LIF signaling perturbations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643751. [PMID: 40166162 PMCID: PMC11957043 DOI: 10.1101/2025.03.17.643751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The LIF signaling pathway and its regulation of internal factors like Sox2 is crucial for maintaining self-renewal and pluripotency in mESCs. However, the direct impact of LIF signaling on Sox2 transcriptional dynamics at the single-cell level remains elusive. Here, we employ PP7/PCP-mediated live imaging to analyze the transcriptional dynamics of Sox2 under perturbation of the LIF signaling pathway at single-cell resolution. Removal of the LIF ligand or addition of a JAK inhibitor heterogeneously affects the cell population, reducing the number of Sox2-active cells, rather than completely abolishing Sox2 expression. Moreover, Sox2-active cells under LIF perturbation exhibit significant reductions in mRNA production per cell. This reduction is characterized by decreased size and frequency of transcriptional bursting, resulting in shorter duration of Sox2 activity. Notably, cells with reduced or absent Sox2 expression demonstrate a significant loss in pluripotency, indicating that a reduction in Sox2 transcription (rather than a complete loss) is sufficient to trigger the transition from embryonic to an early differentiated state. In LIF-perturbed cells with Sox2 expression reduced to about 50% of non-perturbed levels, we observe a binary behavior, with cells either retaining or losing pluripotency-associated traits. Lastly, we find Sox2 expression is transcriptionally inherited across cell cycles, with Sox2-active mother cells more likely to reactivate Sox2 after mitosis compared to Sox2-inactive cells. This robust transcriptional memory is observed independent of LIF signaling perturbation. Our findings provide new insights into the transcriptional regulation of Sox2, advancing our understanding of the quantitative thresholds of gene expression required for pluripotency maintenance and highlighting the power of single-cell approaches to unravel dynamic regulatory mechanisms.
Collapse
|
8
|
Rajalekshmi R, Agrawal DK. Synergistic potential of stem cells and microfluidics in regenerative medicine. Mol Cell Biochem 2025; 480:1481-1493. [PMID: 39285093 PMCID: PMC11842489 DOI: 10.1007/s11010-024-05108-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/27/2024] [Indexed: 02/21/2025]
Abstract
Regenerative medicine has immense potential to revolutionize healthcare by using regenerative capabilities of stem cells. Microfluidics, a cutting-edge technology, offers precise control over cellular microenvironments. The integration of these two fields provides a deep understanding of stem cell behavior and enables the development of advanced therapeutic strategies. This critical review explores the use of microfluidic systems to culture and differentiate stem cells with precision. We examined the use of microfluidic platforms for controlled nutrient supply, mechanical stimuli, and real-time monitoring, providing an unprecedented level of detail in studying cellular responses. The convergence of stem cells and microfluidics holds immense promise for tissue repair, regeneration, and personalized medicine. It offers a unique opportunity to revolutionize the approach to regenerative medicine, facilitating the development of advanced therapeutic strategies and enhancing healthcare outcomes.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
9
|
Rahmatipour H, Shabestari SM, Benisi SZ, Samadikhah H. Pioneering pain management with botulinum toxin type A: From anti-inflammation to regenerative therapies. Heliyon 2025; 11:e42350. [PMID: 40028584 PMCID: PMC11870196 DOI: 10.1016/j.heliyon.2025.e42350] [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: 11/14/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
In the present paper, a comprehensive review was conducted to evaluate the performance of botulinum toxin type A (BTX-A) in managing various types of pain, including myofascial, muscular temporomandibular joint pain, orofacial pain, chronic migraines, and more. Firstly, the mechanism of action and anti-inflammatory effects of BTX-A was introduced. Following this, recent advancements in BTX-A applications were discussed, with an emphasis on emerging combination therapies, regenerative medicine, and personalized treatment strategies. Unlike previous reviews, this study explored a broader spectrum of pain conditions and highlighted BTX-A's versatility and potential as a long-term, minimally invasive pain management option. Additionally, the importance of tailoring BTX-A treatment was emphasized through the integration of biomarkers, genetic factors, and optimized dosing regimens to enhance efficacy and minimize side effects. Novel combinations with regenerative therapies, such as stem cells and tissue engineering, were identified as promising avenues for joint and nerve repair, providing both symptomatic relief and tissue regeneration. Furthermore, digital health tools and artificial intelligence were suggested as innovative approaches to monitor treatment responses and optimize dosing protocols in real-time, advancing personalized pain management. Overall, this review underscores BTX-A's potential in comprehensive and patient-centered pain management and offers recommendations to guide future studies in optimizing BTX-A therapy.
Collapse
Affiliation(s)
- Hamta Rahmatipour
- Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, P.O. Box 13185/768, Tehran, Iran
| | - Salar Mohammadi Shabestari
- Department of Polymer, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, P.O. Box 13185/768, Tehran, Iran
- Stem Cell and Cell Therapy Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hamidreza Samadikhah
- Department of Biology, Faculty of Basic Sciences, Central Tehran Branch, Islamic Azad University, P.O. Box 13145-784, Tehran, Iran
| |
Collapse
|
10
|
Artamonov MY, Pyatakovich FA, Minenko IA. Influence of Super-Low-Intensity Microwave Radiation on Mesenchymal Stem Cells. Int J Mol Sci 2025; 26:1705. [PMID: 40004170 PMCID: PMC11855362 DOI: 10.3390/ijms26041705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising tool for regenerative medicine due to their multipotency and immunomodulatory properties. According to recent research, exposing MSCs to super-low-intensity microwave radiation can have a significant impact on how they behave and operate. This review provides an overview of the most recent studies on the effects of microwave radiation on MSCs with power densities that are much below thermal values. Studies repeatedly show that non-thermal mechanisms affecting calcium signaling, membrane transport, mitochondrial activity, along ion channel activation may increase MSC proliferation, differentiation along mesodermal lineages, paracrine factor secretion, and immunomodulatory capabilities during brief, regulated microwave exposures. These bioeffects greatly enhance MSC regeneration capability in preclinical models of myocardial infarction, osteoarthritis, brain damage, and other diseases. Additional study to understand microwave treatment settings, biological processes, and safety assessments will aid in the translation of this unique, non-invasive strategy of activating MSCs with microwave radiation to improve cell engraftment, survival, and tissue healing results. Microwave-enhanced MSC treatment, if shown safe and successful, might have broad relevance as a novel cell-based approach for a variety of regenerative medicine applications.
Collapse
Affiliation(s)
| | - Felix A. Pyatakovich
- Department of Internal Medicine, Belgorod State University, Belgorod 308015, Russia;
| | - Inessa A. Minenko
- Department of Rehabilitation, Sechenov Medical University, Moscow 119991, Russia;
| |
Collapse
|
11
|
Hajinejad M, Far BF, Gorji A, Sahab-Negah S. The effects of self-assembling peptide on glial cell activation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1391-1402. [PMID: 39305327 DOI: 10.1007/s00210-024-03415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 08/26/2024] [Indexed: 02/14/2025]
Abstract
Glial cells play a critical role in the healthy and diseased phases of the central nervous system (CNS). CNS diseases involve a wide range of pathological conditions characterized by poor recovery of neuronal function. Glial cell-related target therapies are progressively gaining interest in inhibiting secondary injury-related death. Modulation of the extracellular matrix by artificial scaffolds plays a critical role in the behavior of glial cells after injury. Among numerous types of scaffolds, self-assembling peptides (SAPs) notably give attention to the design of a proper biophysical and biomechanical microenvironment for cellular homeostasis and tissue regeneration. Implementing SAPs in an injured brain can induce neural differentiation in transplanted stem cells, reducing inflammation and inhibiting glial scar formation. In this review, we investigate the recent findings to elucidate the pivotal role of SAPs in orchestrating the most pivotal secondary response following CNS injury. Notably, we explore their impact on the activation of glial cells and their modulatory effects on microglial and astrocytic polarization.
Collapse
Affiliation(s)
- Mehrdad Hajinejad
- Qaen Faculty of Medical Sciences, Birjand University of Medical Sciences, Birjand, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Farasati Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Epilepsy Research Center, Neurosurgery Department, Münster University, Münster, Germany
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran.
| |
Collapse
|
12
|
Heo S, Noh M, Kim Y, Park S. Stem Cell-Laden Engineered Patch: Advances and Applications in Tissue Regeneration. ACS APPLIED BIO MATERIALS 2025; 8:62-87. [PMID: 39701826 DOI: 10.1021/acsabm.4c01427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Stem cell-based therapies are emerging as significant approaches in tissue engineering and regenerative medicine, applicable to both fundamental scientific research and clinical practice. Despite remarkable results in clinical studies, challenges such as poor standardization of graft tissues, limited sources, and reduced functionality have hindered the effectiveness of these therapies. In this review, we summarize the engineering approaches involved in fabricating stem cell assisted patches and the substantial strategies for designing stem cell-laden engineered patches (SCP) to complement the existing stem cell-based therapies. We then outline the potential applications of SCP in advancing tissue regeneration and regenerative medicine. By combining living stem cells with engineered patches, SCP can enhance the functions of both components, particularly for tissue engineering applications. Finally, we addressed current challenges, such as ethical considerations, high costs, and regulatory hurdles and proposed future research directions to overcome these barriers.
Collapse
Affiliation(s)
- Seyeong Heo
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Minhyeok Noh
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Yeonseo Kim
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Sunho Park
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| |
Collapse
|
13
|
Ahmed S, Khan RA, Rashid TU. Cellulose nanocrystal based electrospun nanofiber for biomedical applications-A review. Carbohydr Polym 2025; 348:122838. [PMID: 39562112 DOI: 10.1016/j.carbpol.2024.122838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 11/21/2024]
Abstract
Electrospinning has become a revolutionized technique for nanofiber fabrication by offering versatile procedures to precisely regulate the nanofibers' properties suitable for a wide range of advanced applications. Nanofibers are utilized as carriers for delivering medications and other health supplements as well as their ability to discharge their contents can be easily programmed and tailored in a specific manner, while serving as tissue engineering scaffolds or medical devices. Cellulose nanocrystals (CNC) are one of the most significant natural biopolymers incorporated as reinforcing agents for nanostructured fibrous frameworks. The integration of electrospinning technology and CNC offers a viable method for manufacturing nanostructured porous substances with favorable functionality, a high ratio of surface area to volume, a tunable crystal structure along with non-toxicity and cytocompatibility, outstanding mechanical properties, flexibility, sustainability, and biodegradable properties. This article offers a thorough summary of the latest progress in the application of CNC based electrospun nanofibers in various biomedical fields such as drug delivery, tissue engineering, and wound healing. It covers the techniques and parameters used for their fabrication, the different types of raw materials employed, and their application criteria. The review concludes by discussing the prospects and challenges in this rapidly evolving research domains.
Collapse
Affiliation(s)
- Saifuddin Ahmed
- Department Applied Chemistry and Chemical Engineering, Faculty of Engineering and Technology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Ratul Ahmed Khan
- Department Applied Chemistry and Chemical Engineering, Faculty of Engineering and Technology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Taslim Ur Rashid
- Department Applied Chemistry and Chemical Engineering, Faculty of Engineering and Technology, University of Dhaka, Dhaka 1000, Bangladesh.
| |
Collapse
|
14
|
Yu C, Dong L, Lv Y, Shi X, Zhang R, Zhou W, Wu H, Li H, Li Y, Li Z, Luo D, Wei WB. Nanotherapy for Neural Retinal Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2409854. [PMID: 39807033 DOI: 10.1002/advs.202409854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Retinal diseases can severely impair vision and even lead to blindness, posing significant threats to both physical and mental health. Physical retinal regenerative therapies are poised to revolutionize the treatment of various disorders associated with blindness. However, these therapies must overcome the challenges posed by the protective inner and outer blood‒retinal barriers. Nanotechnology applications in ophthalmology have shown great potential in addressing the issue of drug delivery to the eye. Moreover, nanotechnology-based therapeutics can have profound clinical impacts on retinopathy, particularly retinal regeneration, thereby improving patient outcomes. Continuous advancements in nanotechnology are being applied to regenerate lost or damaged eye tissues and to treat vision loss and blindness caused by various retinal degenerative diseases. These approaches can be categorized into three main strategies: i) nanoparticles for delivering drugs, genes, and other essential substances; ii) nanoscaffolds for providing biocompatible support; and iii) nanocomposites for enhancing the functionality of primary or stem cells. The aim of this comprehensive review is to present the current understanding of nanotechnology-based therapeutics for retinal regeneration, with a focus on the perspective functions of nanomaterials.
Collapse
Affiliation(s)
- Chuyao Yu
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Key Laboratory of Intelligent Diagnosis, Treatment and Prevention of Blinding Eye Diseases, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Li Dong
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Key Laboratory of Intelligent Diagnosis, Treatment and Prevention of Blinding Eye Diseases, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yujia Lv
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Xuhan Shi
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Key Laboratory of Intelligent Diagnosis, Treatment and Prevention of Blinding Eye Diseases, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Ruiheng Zhang
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Key Laboratory of Intelligent Diagnosis, Treatment and Prevention of Blinding Eye Diseases, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Wenda Zhou
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Key Laboratory of Intelligent Diagnosis, Treatment and Prevention of Blinding Eye Diseases, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Haotian Wu
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Key Laboratory of Intelligent Diagnosis, Treatment and Prevention of Blinding Eye Diseases, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Heyan Li
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Key Laboratory of Intelligent Diagnosis, Treatment and Prevention of Blinding Eye Diseases, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yitong Li
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Key Laboratory of Intelligent Diagnosis, Treatment and Prevention of Blinding Eye Diseases, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Dan Luo
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Wen-Bin Wei
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Key Laboratory of Intelligent Diagnosis, Treatment and Prevention of Blinding Eye Diseases, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| |
Collapse
|
15
|
Han JJ, Li J, Huang DH. Mesenchymal Stem Cell-Derived Extracellular Vesicles Carrying Circ-Tulp4 Attenuate Diabetes Mellitus with Nonalcoholic Fatty Liver Disease by Inhibiting Cell Pyroptosis through the HNRNPC/ABHD6 Axis. Tissue Eng Regen Med 2025; 22:23-41. [PMID: 39546192 PMCID: PMC11711725 DOI: 10.1007/s13770-024-00675-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/15/2024] [Accepted: 09/22/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Diabetes mellitus with nonalcoholic fatty liver disease (DM-NAFLD) represents a complex metabolic syndrome with significant clinical challenges. This study explores the therapeutic potential and underlying mechanisms of umbilical cord-derived mesenchymal stem cells (UCMSCs)-derived extracellular vesicles (EVs) in DM-NAFLD. METHODS UCMSCs-EVs were isolated and characterized. DM-NAFLD mouse model was developed through high-energy diet and streptozotocin injection. Additionally, primary mouse hepatocytes were exposed to high glucose to simulate cellular conditions. Hepatic tissue damage, body weight changes, lipid levels, glucose and insulin homeostasis, and hepatic lipid accumulation were evaluated. The interaction between UCMSCs-EVs and hepatocytes was assessed, focusing on the localization and function of circ-Tulp4. The study also investigated the expression of circularRNA TUB-like protein 4 (circ-Tulp4), heterogeneous nuclear ribonucleoprotein C (HNRNPC), abhydrolase domain containing 6 (ABHD6), cleaved Caspase-1, NLR family pyrin domain containing 3 (NLRP3) and cleaved N-terminal gasdermin D (GSDMD-N). The binding of circ-Tulp4 to lysine demethylase 6B (KDM6B) and the subsequent epigenetic regulation of ABHD6 by H3K27me3 were analyzed. RESULTS Circ-Tulp4 was reduced, while HNRNPC and ABHD6 were elevated in DM-NAFLD models. UCMSCs-EVs attenuated hepatic steatosis and inhibited the NLRP3/cleaved Caspase-1/GSDMD-N pathway. EVs delivered circ-Tulp4 into hepatocytes, thereby restoring circ-Tulp4 expression. Elevated circ-Tulp4 enhanced the recruitment of H3K27me3 to the HNRNPC promoter through interaction with KDM6B, thus suppressing HNRNPC and ABHD6. Overexpression of HNRNPC or ABHD6 counteracted the protective effects of UCMSCs-EVs, exacerbating pyroptosis and hepatic steatosis in DM-NAFLD. CONCLUSION UCMSCs-EVs deliver circ-Tulp4 into hepatocytes, where circ-Tulp4 inhibits the HNRNPC/ABHD6 axis, thereby reducing pyroptosis and alleviating DM-NAFLD. These findings provide a novel therapeutic avenue for targeting DM-NAFLD through modulation of cell pyroptosis.
Collapse
Affiliation(s)
- Jing-Jing Han
- Department of Endocrinology, The Second Affiliated Hospital of Shandong First Medical University, Taishan Street Road No.366, Tai'an, 271000, Shandong, China
| | - Jing Li
- Department of Pediatric, The Secondary TCM Hospital of Tai'an City, Lingshan Street No.265, Tai'an, 271000, Shandong, China
| | - Dong-Hui Huang
- Department of Endocrinology, The Second Affiliated Hospital of Shandong First Medical University, Taishan Street Road No.366, Tai'an, 271000, Shandong, China.
| |
Collapse
|
16
|
Zou X, Brigstock D. Extracellular Vesicles from Mesenchymal Stem Cells: Potential as Therapeutics in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Biomedicines 2024; 12:2848. [PMID: 39767754 PMCID: PMC11673942 DOI: 10.3390/biomedicines12122848] [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: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/03/2025] Open
Abstract
Background/Objectives: Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by the accumulation of triglycerides within hepatocytes, which can progress to more severe conditions, such as metabolic dysfunction-associated steatohepatitis (MASH), which may include progressive fibrosis, leading to cirrhosis, cancer, and death. This goal of this review is to highlight recent research showing the potential of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) in reducing the key pathogenic pathways of MASLD or MASH. Methods: Relevant published studies were identified using PubMed with one or more of the following search terms: MASLD, MASH, NAFLD, NASH, exosome, extracellular vesicle (EV), therapy, and/or mesenchymal stem cells (MSC). The primary literature were subsequently downloaded and summarized. Results: Using in vitro or in vivo models, MSC-EVs have been found to counteract oxidative stress, a significant contributor to liver injury in MASH, and to suppress disease progression, including steatosis, inflammation, and, in a few instances, fibrosis. Some of these outcomes have been attributed to specific EV cargo components including microRNAs and proteins. Thus, MSC-EVs enriched with these types of molecules may have improved the therapeutic efficacy for MASLD/MASH and represent a novel approach to potentially halt or reverse the disease process. Conclusions: MSC-EVs are attractive therapeutic agents for treating MASLD/MASH. Further studies are necessary to validate the clinical applicability and efficacy of MSC-EVs in human MASH patients, focusing on optimizing delivery strategies and identifying the pathogenic pathways that are targeted by specific EV components.
Collapse
Affiliation(s)
- Xue Zou
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - David Brigstock
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43212, USA
| |
Collapse
|
17
|
Demmer W, Schinacher J, Wiggenhauser PS, Giunta RE. Use of Acellular Matrices as Scaffolds in Cartilage Regeneration: A Systematic Review. Adv Wound Care (New Rochelle) 2024; 13:625-638. [PMID: 38775424 DOI: 10.1089/wound.2024.0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024] Open
Abstract
Significance: Cartilage regeneration remains a significant challenge in the field of regenerative medicine. Acellular matrix (AM)-based cartilage tissue regeneration offers an innovative approach to repairing cartilage defects by providing a scaffold for new tissue growth. Its significance lies in its potential to restore joint function, mitigate pain, and improve the quality of life for patients suffering from cartilage-related injuries and conditions. Recent Advances: Recent advances in AM-based cartilage regeneration have focused on enhancing scaffold properties for improved cell adhesion, proliferation, and differentiation. Moreover, several scaffold techniques such as combining acellular dermal matrix (ADM) and acellular cartilage matrix (ACM) with cartilage tissue, as well as biphasic scaffolding, enjoy rising research activity. Incorporating bioactive factors and advanced manufacturing techniques holds promise for producing more biomimetic scaffolds, advancing efficient cartilage repair and regeneration. Critical Issues: Obstacles in AM-based cartilage regeneration include achieving proper integration with the surrounding tissue and ensuring long-term durability of the regenerated cartilage. Furthermore, issues such as high costs and limited availability of suitable cells for scaffold seeding must be considered. The heterogeneity and limited regenerative capabilities of cartilage need to be addressed for successful clinical translation. Future Directions: Research should focus on exploring advanced biomaterials and developing new techniques, regarding easily reproducible scaffolds, ideally constructed from clinically validated and readily available commercial products. Findings underline the potential of AM-based approaches, especially the rising exploration of tissue-derived ADM and ACM. In future, the primary objective should not only be the regeneration of small cartilage defects but rather focus on fully regenerating a joint or larger cartilage defect.
Collapse
|
18
|
Hussen BM, Taheri M, Yashooa RK, Abdullah GH, Abdullah SR, Kheder RK, Mustafa SA. Revolutionizing medicine: recent developments and future prospects in stem-cell therapy. Int J Surg 2024; 110:8002-8024. [PMID: 39497543 PMCID: PMC11634165 DOI: 10.1097/js9.0000000000002109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/27/2024] [Indexed: 12/13/2024]
Abstract
Stem-cell therapy is a revolutionary frontier in modern medicine, offering enormous capacity to transform the treatment landscape of numerous debilitating illnesses and injuries. This review examines the revolutionary frontier of treatments utilizing stem cells, highlighting the distinctive abilities of stem cells to undergo regeneration and specialized cell differentiation into a wide variety of phenotypes. This paper aims to guide researchers, physicians, and stakeholders through the intricate terrain of stem-cell therapy, examining the processes, applications, and challenges inherent in utilizing stem cells across diverse medical disciplines. The historical journey from foundational contributions in the late 19th and early 20th centuries to recent breakthroughs, including ESC isolation and iPSC discovery, has set the stage for monumental leaps in medical science. Stem cells' regenerative potential spans embryonic, adult, induced pluripotent, and perinatal stages, offering unprecedented therapeutic opportunities in cancer, neurodegenerative disorders, cardiovascular ailments, spinal cord injuries, diabetes, and tissue damage. However, difficulties, such as immunological rejection, tumorigenesis, and precise manipulation of stem-cell behavior, necessitate comprehensive exploration and innovative solutions. This manuscript summarizes recent biotechnological advancements, critical trial evaluations, and emerging technologies, providing a nuanced understanding of the triumphs, difficulties, and future trajectories in stem cell-based regenerative medicine. Future directions, including precision medicine integration, immune modulation strategies, advancements in gene-editing technologies, and bioengineering synergy, offer a roadmap in stem cell treatment. The focus on stem-cell therapy's potential highlights its significant influence on contemporary medicine and points to a future in which individualized regenerative therapies will alleviate various medical disorders.
Collapse
Affiliation(s)
- Bashdar M. Hussen
- Department of Biomedical Sciences, Cihan University-Erbil
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Raya Kh. Yashooa
- General Directorate of Scientific Research Center, Salahaddin University-Erbil
| | | | - Snur R. Abdullah
- Department of Medical Laboratory Science, College of Health sciences, Lebanese French University, Erbil, Kurdistan Region, Erbil, Iraq
| | - Ramiar Kamal Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania, Sulaymaniyah, Iraq
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Suhad A. Mustafa
- General Directorate of Scientific Research Center, Salahaddin University-Erbil
| |
Collapse
|
19
|
Arjmand B, Alavi-Moghadam S, Khorsand G, Sarvari M, Arjmand R, Rezaei-Tavirani M, Rajaeinejad M, Mosaed R. Cell-Based Vaccines: Frontiers in Medical Technology for Cancer Treatment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:480-499. [DOI: 10.1007/s40883-024-00338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 01/13/2024] [Accepted: 02/17/2024] [Indexed: 01/03/2025]
|
20
|
Vaishya R, Gupta BM, Kappi MM, Mamdapur GMN, Ali KS, Vaish A. Scientometric analysis of global research on delayed and nonunion of fractures (2004-2023): Insights from the Web of Science core collections. Injury 2024; 55:111882. [PMID: 39332225 DOI: 10.1016/j.injury.2024.111882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND AND AIMS Nonunion of fractures continue to be the most challenging complication. The increased interest in this area has been anecdotally recognized through published research. The study aimed to conduct a comprehensive bibliometric analysis of global research on nonunion and delayed union of fractures to identify key trends, hotspots, and potential areas for future research development. METHODS Using a pre-defined search strategy, articles related to "Nonunion and Delayed Union of Fractures" that were published from January 1st, 2004 to December 31st, 2023, were retrieved from the Web of Science Core Collection for scientometric analysis. The bibliometrics method was used to analyze the publication dates, countries, institutions, journals, authors, highly cited literature and research hotspots. RESULTS A total of 1997 publications were identified, which registered an average of 21.42 citations per paper (CPP). The 30.39 % and 3.55 % of the 1997 global publications have received external funding support and were highly-cited papers, respectively. The USA, China and UK, were the most productive countries, while Canada, Scotland, and Belgium registered the highest citation impact. University Leeds, Shanghai Jiao Tong University and Kobe University were the most productive organizations, while Vanderbilt University, University of Edinburgh and University Leeds registered the highest citation impact. Giannoudis PV, Egol KA, and Konda SR were the most productive authors, while Bhandari M, Mehta S, and Schemitsch EH registered the highest citation impact. Injury, Journal of Orthopaedic Trauma and Archives of Orthopaedic and Trauma Surgery contributed the most papers, while Journal of Bone and Joint Surgery-American, Injury and Clinical Orthopaedics and Related Research registered the highest citation impact per paper. CONCLUSION The present bibliometric analysis shows the characteristics and trends of non-union fracture research and illuminates the current research situation and developmental trends.
Collapse
Affiliation(s)
- Raju Vaishya
- Department of Orthopaedics, Indraprastha Apollo Hospitals, New Delhi 110076, India.
| | | | - Mallikarjun M Kappi
- Library and Information Centre, Government First Grade College, Hosapete 583201, Karnataka, India
| | - Ghouse Modin N Mamdapur
- Yenepoya (Deemed to be University), Department of Library and Information Science, Deralakatte Mangalore 575018, Karnataka, India.
| | - K S Ali
- Yenepoya (Deemed to be University), Department of Library and Information Science, Deralakatte Mangalore 575018, Karnataka, India.
| | - Abhishek Vaish
- Department of Orthopaedics, Indraprastha Apollo Hospitals, New Delhi 110076, India
| |
Collapse
|
21
|
Su R, Ai Y, Wang J, Wu L, Sun H, Ding M, Xie R, Liang Q. Engineered Microfibers for Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:5823-5840. [PMID: 39145987 DOI: 10.1021/acsabm.4c00615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Hydrogel microfibers are hydrogel materials engineered into fiber structures. Techniques such as wet spinning, microfluidic spinning, and 3D bioprinting are often used to prepare microfibers due to their ability to precisely control the size, morphology, and structure of the microfibers. Microfibers with different structural morphologies have different functions; they provide a flow-through culture environment for cells to improve viability, and can also be used to induce the differentiation of cells such as skeletal muscle and cardiac muscle cells to eventually form functional organs in vitro through special morphologies. This Review introduces recent advances in microfluidics, 3D bioprinting, and wet spinning in the preparation of microfibers, focusing on the materials and fabrication methods. The applications of microfibers in tissue engineering are highlighted by summarizing their contributions in engineering biomimetic blood vessels, vascularized tissues, bone, heart, pancreas, kidney, liver, and fat. Furthermore, applications of engineered fibers in tissue repair and drug screening are also discussed.
Collapse
Affiliation(s)
- Riguga Su
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Jingyu Wang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Lei Wu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Hua Sun
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Mingyu Ding
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Ruoxiao Xie
- Department of Materials, Design and Manufacturing Engineering, School of Engineering, University of Liverpool, Liverpool L69 3BX, U.K
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
22
|
Meretsky CR, Polychronis A, Liovas D, Schiuma AT. Advances in Tissue Engineering and Its Future in Regenerative Medicine Compared to Traditional Reconstructive Techniques: A Comparative Analysis. Cureus 2024; 16:e68872. [PMID: 39376883 PMCID: PMC11457798 DOI: 10.7759/cureus.68872] [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] [Accepted: 09/06/2024] [Indexed: 10/09/2024] Open
Abstract
Tissue engineering represents a revolutionary approach in regenerative medicine, offering promising alternatives to traditional reconstructive techniques. This systematic review explores recent advances in tissue engineering, comparing their efficacy, postoperative outcomes, and patient satisfaction to conventional methods. A comprehensive literature search was conducted across PubMed, Cochrane Library, and Google Scholar, covering studies published from 2000 to 2024. Fourteen studies were selected for final analysis based on inclusion criteria focusing on outcomes such as scar quality, postoperative pain, and patient satisfaction. The review demonstrated that tissue engineering techniques consistently provided superior cosmetic outcomes with minimal scarring compared to traditional methods. Patients undergoing tissue-engineered procedures experienced mild-to-moderate postoperative pain with rapid resolution, whereas traditional techniques resulted in moderate to severe pain requiring extended management. Furthermore, patients treated with tissue engineering reported high satisfaction rates due to improved cosmetic and functional outcomes. Despite challenges such as ensuring adequate vascularization, controlling scaffold degradation, and overcoming regulatory and cost barriers, ongoing research and development are essential to fully realize the potential of these innovative therapies. Tissue engineering offers significant advantages over traditional reconstructive techniques and has the potential to profoundly improve patient care in regenerative medicine.
Collapse
Affiliation(s)
| | - Andreas Polychronis
- General Surgery, St. George's University School of Medicine, Great River, USA
| | - Dimitria Liovas
- Medicine, St. George's University School of Medicine, Great River, USA
| | | |
Collapse
|
23
|
Le QV, Shim G. Biorobotic Drug Delivery for Biomedical Applications. Molecules 2024; 29:3663. [PMID: 39125066 PMCID: PMC11314275 DOI: 10.3390/molecules29153663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 08/12/2024] Open
Abstract
Despite extensive efforts, current drug-delivery systems face biological barriers and difficulties in bench-to-clinical use. Biomedical robotic systems have emerged as a new strategy for drug delivery because of their innovative diminutive engines. These motors enable the biorobots to move independently rather than relying on body fluids. The main components of biorobots are engines controlled by external stimuli, chemical reactions, and biological responses. Many biorobot designs are inspired by blood cells or microorganisms that possess innate swimming abilities and can incorporate living materials into their structures. This review explores the mechanisms of biorobot locomotion, achievements in robotic drug delivery, obstacles, and the perspectives of translational research.
Collapse
Affiliation(s)
- Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam;
| | - Gayong Shim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
- Integrative Institute of Basic Sciences, Soongsil University, Seoul 06978, Republic of Korea
| |
Collapse
|
24
|
Tamimi A, Javid M, Sedighi-Pirsaraei N, Mirdamadi A. Exosome prospects in the diagnosis and treatment of non-alcoholic fatty liver disease. Front Med (Lausanne) 2024; 11:1420281. [PMID: 39144666 PMCID: PMC11322140 DOI: 10.3389/fmed.2024.1420281] [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: 04/19/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
The growing prevalence of NAFLD and its global health burden have provoked considerable research on possible diagnostic and therapeutic options for NAFLD. Although various pathophysiological mechanisms and genetic factors have been identified to be associated with NAFLD, its treatment remains challenging. In recent years, exosomes have attracted widespread attention for their role in metabolic dysfunctions and their efficacy as pathological biomarkers. Exosomes have also shown tremendous potential in treating a variety of disorders. With increasing evidence supporting the significant role of exosomes in NAFLD pathogenesis, their theragnostic potential has become a point of interest in NAFLD. Expectedly, exosome-based treatment strategies have shown promise in the prevention and amelioration of NAFLD in preclinical studies. However, there are still serious challenges in preparing, standardizing, and applying exosome-based therapies as a routine clinical option that should be overcome. Due to the great potential of this novel theragnostic agent in NAFLD, further investigations on their safety, clinical efficacy, and application standardization are highly recommended.
Collapse
|
25
|
Lele M, Kapur S, Hargett S, Sureshbabu NM, Gaharwar AK. Global trends in clinical trials involving engineered biomaterials. SCIENCE ADVANCES 2024; 10:eabq0997. [PMID: 39018412 PMCID: PMC466960 DOI: 10.1126/sciadv.abq0997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/11/2024] [Indexed: 07/19/2024]
Abstract
Engineered biomaterials are materials specifically designed to interact with biological systems for biomedical applications. This paper offers the comprehensive analysis of global clinical trial trends involving such materials. We surveyed 834 studies in the ClinicalTrials.gov database and explored biomaterial types, their initiation points, and durations in clinical trials. Predominantly, synthetic and natural polymers, particularly silicone and collagen, are used. Trials, focusing on ophthalmology, dentistry, and vascular medicine, are primarily conducted in the United States, Canada, and Italy. These trials encompass a broad demographic, and trials enrolled fewer than 100 participants. The study duration varied ranging from 0.5 to 4.5 years. These biomaterials are mainly bioresorbable or bioinert, with the integration of cells in biomaterials remaining an underexplored area. Our findings shed light on current practices and future potentials of engineered biomaterials in clinical research, offering insights for advancing this dynamic field globally.
Collapse
Affiliation(s)
- Mahim Lele
- Bridgeland High School, 10707 Mason Rd., Cypress, TX 77433, USA
| | - Shaunak Kapur
- Seven Lakes High School, 9251 S Fry Rd., Katy, TX 77494, USA
| | - Sarah Hargett
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Nivedhitha Malli Sureshbabu
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA
- Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX 77843, USA
- Department of Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
26
|
Goushki MA, Kharat Z, Kehtari M, Sohi AN, Ahvaz HH, Rad I, HosseinZadeh S, Kouhkan F, Kabiri M. Applications of extraembryonic tissue-derived cells in vascular tissue regeneration. Stem Cell Res Ther 2024; 15:205. [PMID: 38982541 PMCID: PMC11234723 DOI: 10.1186/s13287-024-03784-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024] Open
Abstract
Vascular tissue engineering is a promising approach for regenerating damaged blood vessels and developing new therapeutic approaches for heart disease treatment. To date, different sources of cells have been recognized that offer assistance within the recovery of heart supply routes and veins with distinctive capacities and are compelling for heart regeneration. However, some challenges still remain that need to be overcome to establish the full potential application of these cells. In this paper, we review the different cell sources used for vascular tissue engineering, focusing on extraembryonic tissue-derived cells (ESCs), and elucidate their roles in cardiovascular disease. In addition, we highlight the intricate interplay between mechanical and biochemical factors in regulating mesenchymal stem cell (MSC) differentiation, offering insights into optimizing their application in vascular tissues.
Collapse
Affiliation(s)
- Mehdi Amiri Goushki
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, 14395-1561, Iran
| | - Zahra Kharat
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, 14395-1561, Iran
| | - Mousa Kehtari
- School of Biology, College of Sciences, University of Tehran, Tehran, 1417614411, Iran
| | - Alireza Naderi Sohi
- National Institute of Genetic Engineering and Biotechnology, Tehran, 1497716316, Iran
| | | | - Iman Rad
- Stem Cell Technology Research Center, Tehran, 15856-36473, Iran
| | - Simzar HosseinZadeh
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Kouhkan
- Stem Cell Technology Research Center, Tehran, 15856-36473, Iran
| | - Mahboubeh Kabiri
- Department of Biotechnology, College of Science, University of Tehran, Tehran, 14155-6455, Iran.
| |
Collapse
|
27
|
Komatsu D, Cabrera ARE, Quevedo BV, Asami J, Cristina Motta A, de Moraes SC, Duarte MAT, Hausen MDA, Aparecida de Rezende Duek E. Meniscal repair with additive manufacture of bioresorbable polymer: From physicochemical characterization to implantation of 3D printed poly (L-co-D, L lactide-co-trimethylene carbonate) with autologous stem cells in rabbits. J Biomater Appl 2024; 39:66-79. [PMID: 38646887 DOI: 10.1177/08853282241248517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Three-dimensional (3D) structures are actually the state-of-the-art technique to create porous scaffolds for tissue engineering. Since regeneration in cartilage tissue is limited due to intrinsic cellular properties this study aims to develop and characterize three-dimensional porous scaffolds of poly (L-co-D, L lactide-co-trimethylene carbonate), PLDLA-TMC, obtained by 3D fiber deposition technique. The PLDLA-TMC terpolymer scaffolds (70:30), were obtained and characterized by scanning electron microscopy, gel permeation chromatography, differential scanning calorimetry, thermal gravimetric analysis, compression mechanical testing and study on in vitro degradation, which showed its amorphous characteristics, cylindrical geometry, and interconnected pores. The in vitro degradation study showed significant loss of mechanical properties compatible with a decrease in molar mass, accompanied by changes in morphology. The histocompatibility association of mesenchymal stem cells from rabbit's bone marrow, and PLDLA-TMC scaffolds, were evaluated in the meniscus regeneration, proving the potential of cell culture at in vivo tissue regeneration. Nine New Zealand rabbits underwent total medial meniscectomy, yielding three treatments: implantation of the seeded PLDLA-TMC scaffold, implantation of the unseeded PLDLA-TMC and negative control (defect without any implant). After 24 weeks, the results revealed the presence of fibrocartilage in the animals treated with polymer. However, the regeneration obtained with the seeded PLDLA-TMC scaffolds with mesenchymal stem cells had become intimal to mature fibrocartilaginous tissue of normal meniscus both macroscopically and histologically. This study demonstrated the effectiveness of the PLDLA-TMC scaffold in meniscus regeneration and the potential of mesenchymal stem cells in tissue engineering, without the use of growth factors. It is concluded that bioresorbable polymers represent a promising alternative for tissue regeneration.
Collapse
Affiliation(s)
- Daniel Komatsu
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
| | | | - Bruna Vanessa Quevedo
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
- Post-Graduation Program in Materials Sciences (PPGCM), Federal University of São Carlos (UFSCar), Sorocaba, Brazil
| | - Jessica Asami
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
- Post-Graduation Program of School of Mechanical Engineering (FEM), University of Campinas (UNICAMP), Campinas, Brazil
| | - Adriana Cristina Motta
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
| | | | | | - Moema de Alencar Hausen
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
- Post-Graduation Program of Biomaterials and Regenerative Medicine (PPGBMR), Surgery Department, PUC-SP, Sorocaba, Brazil
| | - Eliana Aparecida de Rezende Duek
- Laboratory of Biomaterials, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, Brazil
- Post-Graduation Program of School of Mechanical Engineering (FEM), University of Campinas (UNICAMP), Campinas, Brazil
- Post-Graduation Program in Materials Sciences (PPGCM), Federal University of São Carlos (UFSCar), Sorocaba, Brazil
- Post-Graduation Program of Biomaterials and Regenerative Medicine (PPGBMR), Surgery Department, PUC-SP, Sorocaba, Brazil
| |
Collapse
|
28
|
Zhang W, Hou Y, Yin S, Miao Q, Lee K, Zhou X, Wang Y. Advanced gene nanocarriers/scaffolds in nonviral-mediated delivery system for tissue regeneration and repair. J Nanobiotechnology 2024; 22:376. [PMID: 38926780 PMCID: PMC11200991 DOI: 10.1186/s12951-024-02580-8] [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: 03/09/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Tissue regeneration technology has been rapidly developed and widely applied in tissue engineering and repair. Compared with traditional approaches like surgical treatment, the rising gene therapy is able to have a durable effect on tissue regeneration, such as impaired bone regeneration, articular cartilage repair and cancer-resected tissue repair. Gene therapy can also facilitate the production of in situ therapeutic factors, thus minimizing the diffusion or loss of gene complexes and enabling spatiotemporally controlled release of gene products for tissue regeneration. Among different gene delivery vectors and supportive gene-activated matrices, advanced gene/drug nanocarriers attract exceptional attraction due to their tunable physiochemical properties, as well as excellent adaptive performance in gene therapy for tissue regeneration, such as bone, cartilage, blood vessel, nerve and cancer-resected tissue repair. This paper reviews the recent advances on nonviral-mediated gene delivery systems with an emphasis on the important role of advanced nanocarriers in gene therapy and tissue regeneration.
Collapse
Affiliation(s)
- Wanheng Zhang
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Shanghai, 200444, China
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yan Hou
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Shanghai, 200444, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, 200444, China
| | - Shiyi Yin
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi Miao
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kyubae Lee
- Department of Biomedical Materials, Konyang University, Daejeon, 35365, Republic of Korea
| | - Xiaojian Zhou
- Department of Pediatrics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Yongtao Wang
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Shanghai, 200444, China.
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
29
|
Park S, Sharma H, Safdar M, Lee J, Kim W, Park S, Jeong HE, Kim J. Micro/nanoengineered agricultural by-products for biomedical and environmental applications. ENVIRONMENTAL RESEARCH 2024; 250:118490. [PMID: 38365052 DOI: 10.1016/j.envres.2024.118490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Agriculturally derived by-products generated during the growth cycles of living organisms as secondary products have attracted increasing interest due to their wide range of biomedical and environmental applications. These by-products are considered promising candidates because of their unique characteristics including chemical stability, profound biocompatibility and offering a green approach by producing the least impact on the environment. Recently, micro/nanoengineering based techniques play a significant role in upgrading their utility, by controlling their structural integrity and promoting their functions at a micro and nano scale. Specifically, they can be used for biomedical applications such as tissue regeneration, drug delivery, disease diagnosis, as well as environmental applications such as filtration, bioenergy production, and the detection of environmental pollutants. This review highlights the diverse role of micro/nano-engineering techniques when applied on agricultural by-products with intriguing properties and upscaling their wide range of applications across the biomedical and environmental fields. Finally, we outline the future prospects and remarkable potential that these agricultural by-products hold in establishing a new era in the realms of biomedical science and environmental research.
Collapse
Affiliation(s)
- Sunho Park
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang, 50463, Republic of Korea
| | - Harshita Sharma
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Mahpara Safdar
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jeongryun Lee
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Woochan Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sangbae Park
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Biosystems Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hoon Eui Jeong
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Jangho Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
30
|
Moyo MTG, Adali T, Tulay P. Exploring gellan gum-based hydrogels for regenerating human embryonic stem cells in age-related macular degeneration therapy: A literature review. Regen Ther 2024; 26:235-250. [PMID: 38966602 PMCID: PMC11222715 DOI: 10.1016/j.reth.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/15/2024] [Accepted: 05/26/2024] [Indexed: 07/06/2024] Open
Abstract
Age-related macular degeneration (AMD) is a progressive ocular disease marked by the deterioration of retinal photoreceptor cells, leading to central vision decline, predominantly affecting the elderly population worldwide. Current treatment modalities, such as anti-VEGF agents, laser therapy, and photodynamic therapy, aim to manage the condition, with emerging strategies like stem cell replacement therapy showing promise. However, challenges like immune rejection and cell survival hinder the efficacy of stem cell interventions. Regenerative medicine faces obstacles in maximizing stem cell potential due to limitations in mimicking the dynamic cues of the extracellular matrix (ECM) crucial for guiding stem cell behaviour. Innovative biomaterials like gellan gum hydrogels offer tailored microenvironments conducive to enhancing stem cell culture efficacy and tissue regeneration. Gellan gum-based hydrogels, renowned for biocompatibility and customizable mechanical properties, provide crucial support for cell viability, differentiation, and controlled release of therapeutic factors, making them an ideal platform for culturing human embryonic stem cells (hESCs). These hydrogels mimic native tissue mechanics, promoting optimal hESC differentiation while minimizing immune responses and facilitating localized delivery. This review explores the potential of Gellan Gum-Based Hydrogels in regenerative AMD therapy, emphasizing their role in enhancing hESC regeneration and addressing current status, treatment limitations, and future directions.
Collapse
Affiliation(s)
- Mthabisi Talent George Moyo
- Near East University, Faculty of Engineering, Department of Biomedical Engineering, P.O. Box: 99138, Nicosia, Cyprus, Mersin 10, Turkey
- Girne American University, Faculty of Medicine, Department of Medical Biochemistry, PO Box 99428, Karmi Campus, Karaoglanoglu, Kyrenia, Cyprus, Mersin 10, Turkey
- Girne American University, Research and Application Center of Biomedical Sciences, PO Box 99428, Karmi Campus, Karaoglanoglu, Kyrenia, North Cyprus, Mersin 10, Turkey
| | - Terin Adali
- Girne American University, Faculty of Medicine, Department of Medical Biochemistry, PO Box 99428, Karmi Campus, Karaoglanoglu, Kyrenia, Cyprus, Mersin 10, Turkey
- Girne American University, Research and Application Center of Biomedical Sciences, PO Box 99428, Karmi Campus, Karaoglanoglu, Kyrenia, North Cyprus, Mersin 10, Turkey
| | - Pinar Tulay
- Near East University, Faculty of Medicine, Department of Medical Genetics, Nicosia, Cyprus, Mersin 10, Turkey
- Near East University, DESAM Research Institute, Nicosia, Cyprus, Mersin 10, Turkey
| |
Collapse
|
31
|
Safiaghdam H, Baniameri S, Aminianfar H, Mohajeri SF, Dehghan MM, Tayebi L, Nokhbatolfoghahaei H, Khojasteh A. Evaluating osteogenic potential of a 3D-printed bioceramic-based scaffold for critical-sized defect treatment: an in vivo and in vitro investigation. In Vitro Cell Dev Biol Anim 2024; 60:657-666. [PMID: 38743380 DOI: 10.1007/s11626-024-00912-4] [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: 12/30/2023] [Accepted: 04/08/2024] [Indexed: 05/16/2024]
Abstract
The integration of precision medicine principles into bone tissue engineering has ignited a wave of research focused on customizing intricate scaffolds through advanced 3D printing techniques. Bioceramics, known for their exceptional biocompatibility and osteoconductivity, have emerged as a promising material in this field. This article aims to evaluate the regenerative capabilities of a composite scaffold composed of 3D-printed gelatin combined with hydroxyapatite/tricalcium phosphate bioceramics (G/HA/TCP), incorporating human dental pulp-derived stem cells (hDPSCs). Using 3D powder printing, we created cross-shaped biphasic calcium phosphate scaffolds with a gelatin layer. The bone-regenerating potential of these scaffolds, along with hDPSCs, was assessed through in vitro analyses and in vivo studies with 60 rats and critical-sized calvarial defects. The assessment included analyzing cellular proliferation, differentiation, and alkaline phosphatase activity (ALP), and concluded with a detailed histological evaluation of bone regeneration. Our study revealed a highly favorable scenario, displaying not only desirable cellular attachment and proliferation on the scaffolds but also a notable enhancement in the ALP activity of hDPSCs, underscoring their pivotal role in bone regeneration. However, the histological examination of calvarial defects at the 12-wk mark yielded a rather modest level of bone regeneration across all experimental groups. The test and cell group exhibited significant bone formation compared to all other groups except the control and cell group. This underscores the complexity of the regenerative process and paves the way for further in-depth investigations aimed at improving the potential of the composite scaffolds.
Collapse
Affiliation(s)
- Hannaneh Safiaghdam
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Baniameri
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Aminianfar
- Department of Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Saeed Farzad Mohajeri
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Lobat Tayebi
- School of Materials Science and Engineering, Helmerich Advanced Technology Research Center, Oklahoma State University, Tulsa, OK, 74106, USA
- Department of Developmental Sciences, Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arash Khojasteh
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Quek J, Vizetto-Duarte C, Teoh SH, Choo Y. Towards Stem Cell Therapy for Critical-Sized Segmental Bone Defects: Current Trends and Challenges on the Path to Clinical Translation. J Funct Biomater 2024; 15:145. [PMID: 38921519 PMCID: PMC11205181 DOI: 10.3390/jfb15060145] [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: 04/24/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
The management and reconstruction of critical-sized segmental bone defects remain a major clinical challenge for orthopaedic clinicians and surgeons. In particular, regenerative medicine approaches that involve incorporating stem cells within tissue engineering scaffolds have great promise for fracture management. This narrative review focuses on the primary components of bone tissue engineering-stem cells, scaffolds, the microenvironment, and vascularisation-addressing current advances and translational and regulatory challenges in the current landscape of stem cell therapy for critical-sized bone defects. To comprehensively explore this research area and offer insights for future treatment options in orthopaedic surgery, we have examined the latest developments and advancements in bone tissue engineering, focusing on those of clinical relevance in recent years. Finally, we present a forward-looking perspective on using stem cells in bone tissue engineering for critical-sized segmental bone defects.
Collapse
Affiliation(s)
- Jolene Quek
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| | - Catarina Vizetto-Duarte
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| | - Swee Hin Teoh
- Centre for Advanced Medical Engineering, College of Materials Science and Engineering, Hunan University, Changsha 410012, China
| | - Yen Choo
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| |
Collapse
|
33
|
Zhang X, Wan J, Huang T, Tang P, Yang L, Bu X, Zhang W, Zhong L. Rapid and accurate identification of stem cell differentiation stages via SERS and convolutional neural networks. BIOMEDICAL OPTICS EXPRESS 2024; 15:2753-2766. [PMID: 38855654 PMCID: PMC11161375 DOI: 10.1364/boe.519093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 06/11/2024]
Abstract
Monitoring the transition of cell states during induced pluripotent stem cell (iPSC) differentiation is crucial for clinical medicine and basic research. However, both identification category and prediction accuracy need further improvement. Here, we propose a method combining surface-enhanced Raman spectroscopy (SERS) with convolutional neural networks (CNN) to precisely identify and distinguish cell states during stem cell differentiation. First, mitochondria-targeted probes were synthesized by combining AuNRs and mitochondrial localization signal (MLS) peptides to obtain effective and stable SERS spectra signals at various stages of cell differentiation. Then, the SERS spectra served as input datasets, and their distinctive features were learned and distinguished by CNN. As a result, rapid and accurate identification of six different cell states, including the embryoid body (EB) stage, was successfully achieved throughout the stem cell differentiation process with an impressive prediction accuracy of 98.5%. Furthermore, the impact of different spectral feature peaks on the identification results was investigated, which provides a valuable reference for selecting appropriate spectral bands to identify cell states. This is also beneficial for shortening the spectral acquisition region to enhance spectral acquisition speed. These results suggest the potential for SERS-CNN models in quality monitoring of stem cells, advancing the practical applications of stem cells.
Collapse
Affiliation(s)
- Xiao Zhang
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| | - Jianhui Wan
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| | - Tao Huang
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| | - Ping Tang
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Liwei Yang
- Guangdong Provincial Key Laboratory of Nanophotonic Functional Materials and Devices, South China Normal University, Guangzhou 510006, China
| | - Xiaoya Bu
- Guangdong Provincial Key Laboratory of Nanophotonic Functional Materials and Devices, South China Normal University, Guangzhou 510006, China
| | - Weina Zhang
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| | - Liyun Zhong
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
34
|
Rajput SN, Naeem BK, Ali A, Salim A, Khan I. Expansion of human umbilical cord derived mesenchymal stem cells in regenerative medicine. World J Stem Cells 2024; 16:410-433. [PMID: 38690517 PMCID: PMC11056638 DOI: 10.4252/wjsc.v16.i4.410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Stem cells are undifferentiated cells that possess the potential for self-renewal with the capacity to differentiate into multiple lineages. In humans, their limited numbers pose a challenge in fulfilling the necessary demands for the regeneration and repair of damaged tissues or organs. Studies suggested that mesenchymal stem cells (MSCs), necessary for repair and regeneration via transplantation, require doses ranging from 10 to 400 million cells. Furthermore, the limited expansion of MSCs restricts their therapeutic application. AIM To optimize a novel protocol to achieve qualitative and quantitative expansion of MSCs to reach the targeted number of cells for cellular transplantation and minimize the limitations in stem cell therapy protocols. METHODS Human umbilical cord (hUC) tissue derived MSCs were obtained and re-cultured. These cultured cells were subjected to the following evaluation procedures: Immunophenotyping, immunocytochemical staining, trilineage differentiation, population doubling time and number, gene expression markers for proliferation, cell cycle progression, senescence-associated β-galactosidase assay, human telomerase reverse transcriptase (hTERT) expression, mycoplasma, cytomegalovirus and endotoxin detection. RESULTS Analysis of pluripotent gene markers Oct4, Sox2, and Nanog in recultured hUC-MSC revealed no significant differences. The immunophenotypic markers CD90, CD73, CD105, CD44, vimentin, CD29, Stro-1, and Lin28 were positively expressed by these recultured expanded MSCs, and were found negative for CD34, CD11b, CD19, CD45, and HLA-DR. The recultured hUC-MSC population continued to expand through passage 15. Proliferative gene expression of Pax6, BMP2, and TGFb1 showed no significant variation between recultured hUC-MSC groups. Nevertheless, a significant increase (P < 0.001) in the mitotic phase of the cell cycle was observed in recultured hUC-MSCs. Cellular senescence markers (hTERT expression and β-galactosidase activity) did not show any negative effect on recultured hUC-MSCs. Additionally, quality control assessments consistently confirmed the absence of mycoplasma, cytomegalovirus, and endotoxin contamination. CONCLUSION This study proposes the development of a novel protocol for efficiently expanding stem cell population. This would address the growing demand for larger stem cell doses needed for cellular transplantation and will significantly improve the feasibility of stem cell based therapies.
Collapse
Affiliation(s)
- Shafiqa Naeem Rajput
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Bushra Kiran Naeem
- Surgical Unit 4, Dr. Ruth KM Pfau Civil Hospital, Karachi 74400, Pakistan
| | - Anwar Ali
- Department of Physiology, University of Karachi, Karachi 75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
- Center for Regenerative Medicine and Stem Cells Research, and Department of Ophthalmology and Visual Sciences, The Aga Khan University, Karachi 74800, Sindh, Pakistan.
| |
Collapse
|
35
|
Elshazly N, Nasr FE, Hamdy A, Saied S, Elshazly M. Advances in clinical applications of bioceramics in the new regenerative medicine era. World J Clin Cases 2024; 12:1863-1869. [PMID: 38660540 PMCID: PMC11036528 DOI: 10.12998/wjcc.v12.i11.1863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/31/2024] [Accepted: 03/20/2024] [Indexed: 04/11/2024] Open
Abstract
In this editorial, we comment on the hard and soft tissue applications of different ceramic-based scaffolds prepared by different mechanisms such as 3D printing, sol-gel, and electrospinning. The new concept of regenerative medicine relies on biomaterials that can trigger in situ tissue regeneration and stem cell recruitment at the defect site. A large percentage of these biomaterials is ceramic-based as they provide the essential requirements of biomaterial principles such as tailored multisize porosity, antibacterial properties, and angiogenic properties. All these previously mentioned properties put bioceramics on top of the hierarchy of biomaterials utilized to stimulate tissue regeneration in soft and hard tissue wounds. Multiple clinical applications registered the use of these materials in triggering soft tissue regeneration in healthy and diabetic patients such as bioactive glass nanofibers. The results were promising and opened new frontiers for utilizing these materials on a larger scale. The same results were mentioned when using different forms and formulas of bioceramics in hard defect regeneration. Some bioceramics were used in combination with other polymers and biological scaffolds to improve their regenerative and mechanical properties. All this progress will enable a larger scale of patients to receive such services with ease and decrease the financial burden on the government.
Collapse
Affiliation(s)
- Noha Elshazly
- Tissue Engineering Laboratory, Faculty of Dentistry, Alexandria University, Alexandria 21526, Egypt
| | - Fayza Eid Nasr
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21526, Egypt
| | - Ayat Hamdy
- Tissue Engineering Laboratory, Faculty of Dentistry, Alexandria University, Alexandria 21526, Egypt
- Public Dental Clinic, Central Administration of Dentistry, Ministry of Health and Population, Alexandria 21554, Egypt
| | - Safa Saied
- Tissue Engineering Laboratory, Faculty of Dentistry, Alexandria University, Alexandria 21526, Egypt
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21526, Egypt
| | - Mohamed Elshazly
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria 21526, Egypt
| |
Collapse
|
36
|
Al Mamun A, Shao C, Geng P, Wang S, Xiao J. The Mechanism of Pyroptosis and Its Application Prospect in Diabetic Wound Healing. J Inflamm Res 2024; 17:1481-1501. [PMID: 38463193 PMCID: PMC10924950 DOI: 10.2147/jir.s448693] [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: 11/17/2023] [Accepted: 02/13/2024] [Indexed: 03/12/2024] Open
Abstract
Pyroptosis defines a form of pro-inflammatory-dependent programmed cell death triggered by gasdermin proteins, which creates cytoplasmic pores and promotes the activation and accumulation of immune cells by releasing several pro-inflammatory mediators and immunogenic substances upon cell rupture. Pyroptosis comprises canonical (mediated by Caspase-1) and non-canonical (mediated by Caspase-4/5/11) molecular signaling pathways. Numerous studies have explored the contributory roles of inflammasome and pyroptosis in the progression of multiple pathological conditions such as tumors, nerve injury, inflammatory diseases and metabolic disorders. Accumulating evidence indicates that the activation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome results in the activation of pyroptosis and inflammation. Current evidence suggests that pyroptosis-dependent cell death plays a progressive role in the development of diabetic complications including diabetic wound healing (DWH) and diabetic foot ulcers (DFUs). This review presents a brief overview of the molecular mechanisms underlying pyroptosis and addresses the current research on pyroptosis-dependent signaling pathways in the context of DWH. In this review, we also present some prospective therapeutic compounds/agents that can target pyroptotic signaling pathways, which may serve as new strategies for the effective treatment and management of diabetic wounds.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui City, Zhejiang, 323000, People's Republic of China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Chuxiao Shao
- Central Laboratory of the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui City, Zhejiang, 323000, People's Republic of China
| | - Peiwu Geng
- Central Laboratory of the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui City, Zhejiang, 323000, People's Republic of China
| | - Shuanghu Wang
- Central Laboratory of the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui City, Zhejiang, 323000, People's Republic of China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
- Department of Wound Healing, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| |
Collapse
|
37
|
Bellon A. Comparing stem cells, transdifferentiation and brain organoids as tools for psychiatric research. Transl Psychiatry 2024; 14:127. [PMID: 38418498 PMCID: PMC10901833 DOI: 10.1038/s41398-024-02780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 03/01/2024] Open
Abstract
The inaccessibility of neurons coming directly from patients has hindered our understanding of mental illnesses at the cellular level. To overcome this obstacle, six different cellular approaches that carry the genetic vulnerability to psychiatric disorders are currently available: Olfactory Neuroepithelial Cells, Mesenchymal Stem Cells, Pluripotent Monocytes, Induced Pluripotent Stem Cells, Induced Neuronal cells and more recently Brain Organoids. Here we contrast advantages and disadvantages of each of these six cell-based methodologies. Neuronal-like cells derived from pluripotent monocytes are presented in more detail as this technique was recently used in psychiatry for the first time. Among the parameters used for comparison are; accessibility, need for reprograming, time to deliver differentiated cells, differentiation efficiency, reproducibility of results and cost. We provide a timeline on the discovery of these cell-based methodologies, but, our main goal is to assist researchers selecting which cellular approach is best suited for any given project. This manuscript also aims to help readers better interpret results from the published literature. With this goal in mind, we end our work with a discussion about the differences and similarities between cell-based techniques and postmortem research, the only currently available tools that allow the study of mental illness in neurons or neuronal-like cells coming directly from patients.
Collapse
Affiliation(s)
- Alfredo Bellon
- Penn State Hershey Medical Center, Department of Psychiatry and Behavioral Health, Hershey, PA, USA.
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, USA.
| |
Collapse
|
38
|
Aoki K, Ideta H, Komatsu Y, Tanaka A, Kito M, Okamoto M, Takahashi J, Suzuki S, Saito N. Bone-Regeneration Therapy Using Biodegradable Scaffolds: Calcium Phosphate Bioceramics and Biodegradable Polymers. Bioengineering (Basel) 2024; 11:180. [PMID: 38391666 PMCID: PMC10886059 DOI: 10.3390/bioengineering11020180] [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: 01/08/2024] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Calcium phosphate-based synthetic bone is broadly used for the clinical treatment of bone defects caused by trauma and bone tumors. Synthetic bone is easy to use; however, its effects depend on the size and location of the bone defect. Many alternative treatment options are available, such as joint arthroplasty, autologous bone grafting, and allogeneic bone grafting. Although various biodegradable polymers are also being developed as synthetic bone material in scaffolds for regenerative medicine, the clinical application of commercial synthetic bone products with comparable performance to that of calcium phosphate bioceramics have yet to be realized. This review discusses the status quo of bone-regeneration therapy using artificial bone composed of calcium phosphate bioceramics such as β-tricalcium phosphate (βTCP), carbonate apatite, and hydroxyapatite (HA), in addition to the recent use of calcium phosphate bioceramics, biodegradable polymers, and their composites. New research has introduced potential materials such as octacalcium phosphate (OCP), biologically derived polymers, and synthetic biodegradable polymers. The performance of artificial bone is intricately related to conditions such as the intrinsic material, degradability, composite materials, manufacturing method, structure, and signaling molecules such as growth factors and cells. The development of new scaffold materials may offer more efficient bone regeneration.
Collapse
Affiliation(s)
- Kaoru Aoki
- Physical Therapy Division, School of Health Sciences, Shinshu University, Matsumoto 390-8621, Japan
| | - Hirokazu Ideta
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Yukiko Komatsu
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Atsushi Tanaka
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Munehisa Kito
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Masanori Okamoto
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Jun Takahashi
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Shuichiro Suzuki
- Department of Orthopaedic Surgery, Matsumoto Medical Center, Matsumoto 390-8621, Japan
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto 390-8621, Japan
| |
Collapse
|
39
|
Yadav P, Singh SK, Rajput S, Allawadhi P, Khurana A, Weiskirchen R, Navik U. Therapeutic potential of stem cells in regeneration of liver in chronic liver diseases: Current perspectives and future challenges. Pharmacol Ther 2024; 253:108563. [PMID: 38013053 DOI: 10.1016/j.pharmthera.2023.108563] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/04/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
The deposition of extracellular matrix and hyperplasia of connective tissue characterizes chronic liver disease called hepatic fibrosis. Progression of hepatic fibrosis may lead to hepatocellular carcinoma. At this stage, only liver transplantation is a viable option. However, the number of possible liver donors is less than the number of patients needing transplantation. Consequently, alternative cell therapies based on non-stem cells (e.g., fibroblasts, chondrocytes, keratinocytes, and hepatocytes) therapy may be able to postpone hepatic disease, but they are often ineffective. Thus, novel stem cell-based therapeutics might be potentially important cutting-edge approaches for treating liver diseases and reducing patient' suffering. Several signaling pathways provide targets for stem cell interventions. These include pathways such as TGF-β, STAT3/BCL-2, NADPH oxidase, Raf/MEK/ERK, Notch, and Wnt/β-catenin. Moreover, mesenchymal stem cells (MSCs) stimulate interleukin (IL)-10, which inhibits T-cells and converts M1 macrophages into M2 macrophages, producing an anti-inflammatory environment. Furthermore, it inhibits the action of CD4+ and CD8+ T cells and reduces the activity of TNF-α and interferon cytokines by enhancing IL-4 synthesis. Consequently, the immunomodulatory and anti-inflammatory capabilities of MSCs make them an attractive therapeutic approach. Importantly, MSCs can inhibit the activation of hepatic stellate cells, causing their apoptosis and subsequent promotion of hepatocyte proliferation, thereby replacing dead hepatocytes and reducing liver fibrosis. This review discusses the multidimensional therapeutic role of stem cells as cell-based therapeutics in liver fibrosis.
Collapse
Affiliation(s)
- Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sumeet Kumar Singh
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sonu Rajput
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Prince Allawadhi
- Department of Pharmacy, Vaish Institute of Pharmaceutical Education and Research (VIPER), Pandit Bhagwat Dayal Sharma University of Health Sciences (Pt. B. D. S. UHS), Rohtak, Haryana 124001, India
| | - Amit Khurana
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| |
Collapse
|
40
|
Ye P, Gu R, Zhu H, Chen J, Han F, Nie X. SOX family transcription factors as therapeutic targets in wound healing: A comprehensive review. Int J Biol Macromol 2023; 253:127243. [PMID: 37806414 DOI: 10.1016/j.ijbiomac.2023.127243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
The SOX family plays a vital role in determining the fate of cells and has garnered attention in the fields of cancer research and regenerative medicine. It also shows promise in the study of wound healing, as it actively participates in the healing processes of various tissues such as skin, fractures, tendons, and the cornea. However, our understanding of the mechanisms behind the SOX family's involvement in wound healing is limited compared to its role in cancer. Gaining insight into its role, distribution, interaction with other factors, and modifications in traumatized tissues could provide valuable new knowledge about wound healing. Based on current research, SOX2, SOX7, and SOX9 are the most promising members of the SOX family for future interventions in wound healing. SOX2 and SOX9 promote the renewal of cells, while SOX7 enhances the microvascular environment. The SOX family holds significant potential for advancing wound healing research. This article provides a comprehensive review of the latest research advancements and therapeutic tools related to the SOX family in wound healing, as well as the potential benefits and challenges of targeting the SOX family for wound treatment.
Collapse
Affiliation(s)
- Penghui Ye
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Rifang Gu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; School Medical Office, Zunyi Medical University, Zunyi 563006, China
| | - Huan Zhu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Jitao Chen
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Felicity Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xuqiang Nie
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
41
|
Ranjbar N, Bakhshandeh B, Pennisi CP. Electroconductive Nanofibrous Scaffolds Enable Neuronal Differentiation in Response to Electrical Stimulation without Exogenous Inducing Factors. Bioengineering (Basel) 2023; 10:1438. [PMID: 38136029 PMCID: PMC10740536 DOI: 10.3390/bioengineering10121438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Among the various biochemical and biophysical inducers for neural regeneration, electrical stimulation (ES) has recently attracted considerable attention as an efficient means to induce neuronal differentiation in tissue engineering approaches. The aim of this in vitro study was to develop a nanofibrous scaffold that enables ES-mediated neuronal differentiation in the absence of exogenous soluble inducers. A nanofibrous scaffold composed of polycaprolactone (PCL), poly-L-lactic acid (PLLA), and single-walled nanotubes (SWNTs) was fabricated via electrospinning and its physicochemical properties were investigated. The cytocompatibility of the electrospun composite with the PC12 cell line and bone marrow-derived mesenchymal stem cells (BMSCs) was investigated. The results showed that the PCL/PLLA/SWNT nanofibrous scaffold did not exhibit cytotoxicity and supported cell attachment, spreading, and proliferation. ES was applied to cells cultured on the nanofibrous scaffolds at different intensities and the expression of the three neural markers (Nestin, Microtubule-associated protein 2, and β tubulin-3) was evaluated using RT-qPCR analysis. The results showed that the highest expression of neural markers could be achieved at an electric field intensity of 200 mV/cm, suggesting that the scaffold in combination with ES can be an efficient tool to accelerate neural differentiation in the absence of exogenous soluble inducers. This has important implications for the regeneration of nerve injuries and may provide insights for further investigations of the mechanisms underlying ES-mediated neuronal commitment.
Collapse
Affiliation(s)
- Nika Ranjbar
- Department of Biotechnology, College of Science, University of Tehran, Tehran 14155-6455, Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran 14155-6455, Iran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, DK-9260 Gistrup, Denmark
| |
Collapse
|
42
|
Hong J, Ahn H, Moon SY, Kang HJ, Yi KW. Effect of collagen endometrial patch loaded with adipose-derived mesenchymal stem cells on endometrial regeneration in rats with a thin endometrium. Front Endocrinol (Lausanne) 2023; 14:1287789. [PMID: 38089603 PMCID: PMC10714005 DOI: 10.3389/fendo.2023.1287789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 10/31/2023] [Indexed: 12/18/2023] Open
Abstract
Background This study aimed to investigate the effects of a collagen endometrial patch (EM patch) loaded with adipose-derived mesenchymal stem cells (ADSCs) on endometrial regeneration in a rat model with thin endometrium. Materials and methods Thin endometrium was induced in female rats and divided into treatment groups as outlined: control, group 1(G1), local injection of ADSCs into the uterus, group 2 (G2), an EM patch without ADSCs, group 3 (G3), and an EM patch loaded with ADSCs, group 4 (G4). The rats were euthanized at either two weeks or four weeks after modeling and treatment followed by histological and biochemical analyses to examine the regenerative effects on the injured endometrium. Results Transplantation of the ADSC-loaded EM patch significantly promoted endometrial proliferation and increased the luminal epithelial area. Two weeks after treatment, the mean number of von Villebrand factor (vWF)+ or cluster of differentiation (CD) 31+-stained blood vessels was significantly higher in G4 than in G1 and G2. The mRNA and protein expression levels of TGF-β and FGF2 were significantly upregulated in G4 compared to those in the control. G4 exhibited significantly increased LIF mRNA levels and immunoreactivity compared with the other groups at both two weeks and four weeks after treatment. Cell tracking after ADSCs treatment revealed the presence of a substantial number of ADSCs grafted in the uterine tissues of G4, whereas a low number of ADSCs that were focally clustered were present in G2. Conclusion Transplantation of EM patches loaded with ADSCs resulted in the histological and biochemical restoration of an injured endometrium. The strategic integration of EM patches and ADSCs holds significant promise as an innovative therapeutic approach for effectively treating impaired endometrial conditions.
Collapse
Affiliation(s)
- Juyeon Hong
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyojin Ahn
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Soo Young Moon
- Department of Biomedical Laboratory Science, Honam University, Gwangju, Republic of Korea
| | - Hyo Jin Kang
- Department of Biomedical Laboratory Science, Honam University, Gwangju, Republic of Korea
| | - Kyong Wook Yi
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
43
|
Kwon JW, Savitri C, An B, Yang SW, Park K. Mesenchymal stem cell-derived secretomes-enriched alginate/ extracellular matrix hydrogel patch accelerates skin wound healing. Biomater Res 2023; 27:107. [PMID: 37904231 PMCID: PMC10617187 DOI: 10.1186/s40824-023-00446-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/15/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND The secretomes of mesenchymal stem cells (MSCs) have great therapeutic potential and thereby their efficient delivery into the target site is of particular interest. Here, we propose a new strategy of hMSCs-derived secretomes delivery for advanced wound healing upon harnessing the working principle of extracellular matrix (ECM)-growth factors interaction in vivo. METHODS We prepared an alginate hydrogel based wound patch, where it contains both human MSC-derived secretomes and ECM. The ECM was obtained from the decellularization of in vitro cultured human lung fibroblasts. The alginate solution was blended with ECM suspension, crosslinked, air-dried, then rehydrated with the secretomes contained in the concentrated conditioned media (CCM) as a highly saturated form of conditioned media (CM). We tested four different groups, with or without the ECM to investigate not only the role of ECM but the therapeutic effect of secretomes. RESULTS The secretomes reserved many, diverse bioactive factors, such as VEGF, HGF, IGFBPs, IL-6, and IL-8. Alginate/ECM/CCM (AEC) patch could hold significantly larger amount of secretomes and release them longer than the other groups. Our AEC patch was the most effective in stimulating not only cell migration and proliferation but the collagen synthesis of dermal fibroblasts in vitro. Moreover, the AEC patch-treated full-thickness skin wounds disclosed significantly better wound healing indications: cell recruitment, neovascularization, epidermis thickness, keratinocyte migration, and mature collagen deposition, as assessed via histology (H&E, Herovici staining) and immunofluorescence, respectively. In particular, our AEC patch enabled a phenotype shift of myofibroblast into fibroblast over time and led to mature blood vessel formation at 14 day. CONCLUSIONS We believe that ECM certainly contributed to generate a secretomes-enriched milieu via ECM-secretomes interactions and thereby such secretomes could be delivered more efficiently, exerting significant therapeutic impact either individually or collectively during wound healing process.
Collapse
Affiliation(s)
- Jae Won Kwon
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST school, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Cininta Savitri
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Byoungha An
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST school, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Seung Won Yang
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST school, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
- Division of Bio-Medical Science and Technology, KIST school, University of Science and Technology (UST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
44
|
Fenberg R, vonWindheim N, Malara M, Ahmed M, Cowen E, Melaragno L, Vankoevering K. Tissue Engineering: Current Technology for Facial Reconstruction. Facial Plast Surg 2023; 39:489-495. [PMID: 37290454 DOI: 10.1055/s-0043-1769808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
Facial reconstruction is a complex surgical process that requires intricate three-dimensional (3D) concepts for optimal functional and aesthetic outcomes. Conventional reconstruction of structural facial anomalies, such as those including cartilage or bony defects, typically rely on hand-carving autologous constructs harvested from a separate donor site, and shaping that cartilage or bone into a new structural framework. Tissue engineering has emerged in recent decades as a potential approach to mitigate the need for donor site morbidity while improving precision in the design of reconstructive construct. Computer-aided design and computer-aided manufacturing have allowed for a digital 3D workflow to digitally execute the planned reconstruction in virtual space. 3D printing and other manufacturing techniques can then be utilized to create custom-fabricated scaffolds and guides to improve the reconstructive efficiency. Tissue engineering can be paired with custom 3D-manufactured scaffolds to theoretically create an ideal framework for structural reconstruction. In the past decade, there have been several compelling preclinical studies demonstrating the capacity to induce chondrogenesis or osteogenesis in a custom scaffold. However, to date, these preclinical data have not yet translated into significant clinical experience. This translation has been hindered by a lack of consensus on the ideal materials and cellular progenitors to be utilized in these constructs and a lack of regulatory guidance and control to enable clinical application. In this review, we highlight the current state of tissue engineering in facial reconstruction and exciting potential for future applications as the field continues to advance.
Collapse
Affiliation(s)
- Rachel Fenberg
- School of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Natalia vonWindheim
- Center for Design and Manufacturing Excellence, The Ohio State University College of Engineering, Columbus, Ohio
| | - Megan Malara
- Center for Design and Manufacturing Excellence, The Ohio State University College of Engineering, Columbus, Ohio
| | - Maariyah Ahmed
- Center for Design and Manufacturing Excellence, The Ohio State University College of Engineering, Columbus, Ohio
| | - Erin Cowen
- Center for Design and Manufacturing Excellence, The Ohio State University College of Engineering, Columbus, Ohio
| | - Luigi Melaragno
- Center for Design and Manufacturing Excellence, The Ohio State University College of Engineering, Columbus, Ohio
| | - Kyle Vankoevering
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
45
|
Stougiannou TM, Christodoulou KC, Georgakarakos E, Mikroulis D, Karangelis D. Promising Novel Therapies in the Treatment of Aortic and Visceral Aneurysms. J Clin Med 2023; 12:5878. [PMID: 37762818 PMCID: PMC10531975 DOI: 10.3390/jcm12185878] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Aortic and visceral aneurysms affect large arterial vessels, including the thoracic and abdominal aorta, as well as visceral arterial branches, such as the splenic, hepatic, and mesenteric arteries, respectively. Although these clinical entities have not been equally researched, it seems that they might share certain common pathophysiological changes and molecular mechanisms. The yet limited published data, with regard to newly designed, novel therapies, could serve as a nidus for the evaluation and potential implementation of such treatments in large artery aneurysms. In both animal models and clinical trials, various novel treatments have been employed in an attempt to not only reduce the complications of the already implemented modalities, through manufacturing of more durable materials, but also to regenerate or replace affected tissues themselves. Cellular populations like stem and differentiated vascular cell types, large diameter tissue-engineered vascular grafts (TEVGs), and various molecules and biological factors that might target aspects of the pathophysiological process, including cell-adhesion stabilizers, metalloproteinase inhibitors, and miRNAs, could potentially contribute significantly to the treatment of these types of aneurysms. In this narrative review, we sought to collect and present relevant evidence in the literature, in an effort to unveil promising biological therapies, possibly applicable to the treatment of aortic aneurysms, both thoracic and abdominal, as well as visceral aneurysms.
Collapse
Affiliation(s)
- Theodora M. Stougiannou
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (E.G.); (D.M.); (D.K.)
| | | | | | | | | |
Collapse
|
46
|
Giannotti L, Di Chiara Stanca B, Spedicato F, Nitti P, Damiano F, Demitri C, Calabriso N, Carluccio MA, Palermo A, Siculella L, Stanca E. Progress in Regenerative Medicine: Exploring Autologous Platelet Concentrates and Their Clinical Applications. Genes (Basel) 2023; 14:1669. [PMID: 37761809 PMCID: PMC10530962 DOI: 10.3390/genes14091669] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
The goal of regenerative medicine is to achieve tissue regeneration. In the past, commonly used techniques included autologous or allogeneic transplantation and stem cell therapy, which have limitations, such as a lack of donor sites in the case of autologous transplantation and the invasiveness of stem cell harvesting. In recent years, research has, therefore, focused on new and less invasive strategies to achieve tissue regeneration. A step forward in this direction has been made with the development of autologous platelet concentrates (APCs), which are derived from the patient's own blood. They can be classified into three generations: platelet-rich plasma (PRP), platelet-rich fibrin (PRF), and concentrated growth factors (CGFs). These APCs have different structural characteristics, depending on the distinctive preparation method, and contain platelets, leukocytes, and multiple growth factors, including those most involved in regenerative processes. The purpose of this review is to clarify the most used techniques in the field of regenerative medicine in recent years, comparing the different types of APCs and analyzing the preparation protocols, the composition of the growth factors, the level of characterization achieved, and their clinical applications to date.
Collapse
Affiliation(s)
- Laura Giannotti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Benedetta Di Chiara Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Francesco Spedicato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Paola Nitti
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy; (P.N.); (C.D.)
| | - Fabrizio Damiano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Christian Demitri
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy; (P.N.); (C.D.)
| | - Nadia Calabriso
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100 Lecce, Italy; (N.C.); (M.A.C.)
| | - Maria Annunziata Carluccio
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100 Lecce, Italy; (N.C.); (M.A.C.)
| | - Andrea Palermo
- Implant Dentistry College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Luisa Siculella
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| | - Eleonora Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.G.); (B.D.C.S.); (F.S.); (F.D.); (E.S.)
| |
Collapse
|
47
|
Panagiotou N, McGuinness D, Jaminon AMG, Mees B, Selman C, Schurgers L, Shiels PG. Microvesicle-Mediated Tissue Regeneration Mitigates the Effects of Cellular Ageing. Cells 2023; 12:1707. [PMID: 37443741 PMCID: PMC10340655 DOI: 10.3390/cells12131707] [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: 06/01/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Extracellular vesicles (EVs), comprising microvesicles (MVs) and exosomes (Exos), are membranous vesicles secreted by cells which mediate the repair of cellular and tissue damage via paracrine mechanisms. The action of EVs under normative and morbid conditions in the context of ageing remains largely unexplored. We demonstrate that MVs, but not Exos, from Pathfinder cells (PCs), a putative stem cell regulatory cell type, enhance the repair of human dermal fibroblast (HDF) and mesenchymal stem cell (MSC) co-cultures, following both mechanical and genotoxic stress. Critically, this effect was found to be both cellular age and stress specific. Notably, MV treatment was unable to repair mechanical injury in older co-cultures but remained therapeutic following genotoxic stress. These observations were further confirmed in human dermal fibroblast (HDF) and vascular smooth muscle cell (VSMC) co-cultures of increasing cellular age. In a model of comorbidity comprising co-cultures of HDFs and highly senescent abdominal aortic aneurysm (AAA) VSMCs, MV administration appeared to be senotherapeutic, following both mechanical and genotoxic stress. Our data provide insights into EVs and the specific roles they play during tissue repair and ageing. These data will potentiate the development of novel cell-free therapeutic interventions capable of attenuating age-associated morbidities and avoiding undesired effects.
Collapse
Affiliation(s)
- Nikolaos Panagiotou
- Davidson Building, School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, UK; (N.P.)
| | - Dagmara McGuinness
- School of Infection & Immunity, University of Glasgow, Glasgow G12 8QQ, UK; (D.M.)
| | - Armand M. G. Jaminon
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University,
Maastricht, 6229 ER Maastricht, NetherlandsThe Netherlands
| | - Barend Mees
- Department of Vascular Surgery, Maastricht University Medical Centre (MUMC),
Maastricht, The Netherlands;
| | - Colin Selman
- Graham Kerr Building, College of Medical, Veterinary & Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK;
| | - Leon Schurgers
- School of Infection & Immunity, University of Glasgow, Glasgow G12 8QQ, UK; (D.M.)
- Graham Kerr Building, College of Medical, Veterinary & Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK;
| | - Paul G. Shiels
- Davidson Building, School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, UK; (N.P.)
| |
Collapse
|
48
|
Shi G, Zhou Y, Liu W, Chen C, Wei Y, Yan X, Wu L, Wang W, Sun L, Zhang T. Bone-derived MSCs encapsulated in alginate hydrogel prevent collagen-induced arthritis in mice through the activation of adenosine A 2A/2B receptors in tolerogenic dendritic cells. Acta Pharm Sin B 2023; 13:2778-2794. [PMID: 37425054 PMCID: PMC10326293 DOI: 10.1016/j.apsb.2023.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 07/11/2023] Open
Abstract
Tolerogenic dendritic cells (tolDCs) facilitate the suppression of autoimmune responses by differentiating regulatory T cells (Treg). The dysfunction of immunotolerance results in the development of autoimmune diseases, such as rheumatoid arthritis (RA). As multipotent progenitor cells, mesenchymal stem cells (MSCs), can regulate dendritic cells (DCs) to restore their immunosuppressive function and prevent disease development. However, the underlying mechanisms of MSCs in regulating DCs still need to be better defined. Simultaneously, the delivery system for MSCs also influences their function. Herein, MSCs are encapsulated in alginate hydrogel to improve cell survival and retention in situ, maximizing efficacy in vivo. The three-dimensional co-culture of encapsulated MSCs with DCs demonstrates that MSCs can inhibit the maturation of DCs and the secretion of pro-inflammatory cytokines. In the collagen-induced arthritis (CIA) mice model, alginate hydrogel encapsulated MSCs induce a significantly higher expression of CD39+CD73+ on MSCs. These enzymes hydrolyze ATP to adenosine and activate A2A/2B receptors on immature DCs, further promoting the phenotypic transformation of DCs to tolDCs and regulating naïve T cells to Tregs. Therefore, encapsulated MSCs obviously alleviate the inflammatory response and prevent CIA progression. This finding clarifies the mechanism of MSCs-DCs crosstalk in eliciting the immunosuppression effect and provides insights into hydrogel-promoted stem cell therapy for autoimmune diseases.
Collapse
Affiliation(s)
- Gaona Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yu Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Wenshuai Liu
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Chengjuan Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yazi Wei
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing 100124, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Lan Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
49
|
Matos BMD, Stimamiglio MA, Correa A, Robert AW. Human pluripotent stem cell-derived extracellular vesicles: From now to the future. World J Stem Cells 2023; 15:453-465. [PMID: 37342215 PMCID: PMC10277970 DOI: 10.4252/wjsc.v15.i5.453] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/14/2023] [Accepted: 04/13/2023] [Indexed: 05/26/2023] Open
Abstract
Extracellular vesicles (EVs) are nanometric particles that enclose cell-derived bioactive molecules in a lipid bilayer and serve as intercellular communication tools. Accordingly, in various biological contexts, EVs are reported to engage in immune modulation, senescence, and cell proliferation and differentiation. Therefore, EVs could be key elements for potential off-the-shelf cell-free therapy. Little has been studied regarding EVs derived from human pluripotent stem cells (hPSC-EVs), even though hPSCs offer good opportunities for induction of tissue regeneration and unlimited proliferative ability. In this review article, we provide an overview of studies using hPSC-EVs, focusing on identifying the conditions in which the cells are cultivated for the isolation of EVs, how they are characterized, and applications already demonstrated. The topics reported in this article highlight the incipient status of the studies in the field and the significance of hPSC-EVs’ prospective applications as PSC-derived cell-free therapy products.
Collapse
Affiliation(s)
- Bruno Moises de Matos
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute, Curitiba 81350010, Paraná, Brazil
| | | | - Alejandro Correa
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute, Curitiba 81350010, Paraná, Brazil
| | - Anny Waloski Robert
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute, Curitiba 81350010, Paraná, Brazil
| |
Collapse
|
50
|
Park D, Lee SJ, Choi DK, Park JW. Therapeutic Agent-Loaded Fibrous Scaffolds for Biomedical Applications. Pharmaceutics 2023; 15:pharmaceutics15051522. [PMID: 37242764 DOI: 10.3390/pharmaceutics15051522] [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: 04/05/2023] [Revised: 04/28/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Tissue engineering is a sophisticated field that involves the integration of various disciplines, such as clinical medicine, material science, and life science, to repair or regenerate damaged tissues and organs. To achieve the successful regeneration of damaged or diseased tissues, it is necessary to fabricate biomimetic scaffolds that provide structural support to the surrounding cells and tissues. Fibrous scaffolds loaded with therapeutic agents have shown considerable potential in tissue engineering. In this comprehensive review, we examine various methods for fabricating bioactive molecule-loaded fibrous scaffolds, including preparation methods for fibrous scaffolds and drug-loading techniques. Additionally, we delved into the recent biomedical applications of these scaffolds, such as tissue regeneration, inhibition of tumor recurrence, and immunomodulation. The aim of this review is to discuss the latest research trends in fibrous scaffold manufacturing methods, materials, drug-loading methods with parameter information, and therapeutic applications with the goal of contributing to the development of new technologies or improvements to existing ones.
Collapse
Affiliation(s)
- Dongsik Park
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Su Jin Lee
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Dong Kyu Choi
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Jee-Woong Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| |
Collapse
|