1
|
Xu J, Ruan X. Schwann cell autotransplantation for the treatment of peripheral nerve injury. Life Sci 2024; 358:123129. [PMID: 39393574 DOI: 10.1016/j.lfs.2024.123129] [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: 03/08/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Peripheral nerve injury occurs in a relatively large proportion of trauma patients, in whom it generally results in severe functional impairment and permanent disability. At present, however, there are no effective treatments available. Studies have shown that Schwann cells play an indispensable role in removing myelin debris and guiding axonal regeneration, and transplantation using autologous Schwann cells has shown good efficacy for patients with peripheral nerve injury. In recent years, Schwann cell autologous transplantation therapy has become an area of intensive research and is anticipated to provide a new strategy for the clinical treatment of peripheral nerve injury. In this article, we review the rationale for selecting Schwann cell autotransplantation therapy and the latest progress in key aspects of cell transplantation and clinical efficacy, and also summarize the future directions of research on this therapy. All of the above provide a strong basis for the further improvement and clinical promotion of this therapy.
Collapse
Affiliation(s)
- Jialiang Xu
- China Medical University, Shenyang, Liaoning 110122, People's Republic of China.
| | - Xuelei Ruan
- Department of Neurobiology, China Medical University, Shenyang, Liaoning 110122, People's Republic of China.
| |
Collapse
|
2
|
He J, Shan S, Jiang T, Zhou S, Qin J, Li Q, Yu Z, Cao D, Fang B. Mechanical stretch preconditioned adipose-derived stem cells elicit polarization of anti-inflammatory M2-like macrophages and improve chronic wound healing. FASEB J 2024; 38:e23626. [PMID: 38739537 DOI: 10.1096/fj.202300586r] [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: 03/27/2023] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 05/16/2024]
Abstract
Transplantation of adipose-derived stem cells (ASCs) is a promising option in the field of chronic wounds treatment. However, the effectiveness of ASCs therapies has been hampered by highly inflammatory environment in chronic wound areas. These problems could be partially circumvented using efficient approaches that boost the survival and anti-inflammatory capacity of transplanted ASCs. Here, by application of mechanical stretch (MS), we show that ASCs exhibits increased survival and immunoregulatory properties in vitro. MS triggers the secretion of macrophage colony stimulating factor (M-CSF) from ASCs, a chemokine that is linked to anti-inflammatory M2-like macrophages polarization. When the MS-ASCs were transplanted to chronic wounds, the wound area yields significantly faster closure rate and lower inflammatory mediators, largely due to macrophages polarization driven by transplanted MS-ASCs. Thus, our work shows that mechanical stretch can be harnessed to enhance ASCs transplantation efficiency in chronic wounds treatment.
Collapse
Affiliation(s)
- Jiahao He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Taoran Jiang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sizheng Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Qin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheyuan Yu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dejun Cao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Fang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Kennedy O, Kitson A, Okpara C, Chow LW, Gonzalez-Fernandez T. Immunomodulatory Strategies for Cartilage Regeneration in Osteoarthritis. Tissue Eng Part A 2024; 30:259-271. [PMID: 38126327 DOI: 10.1089/ten.tea.2023.0255] [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: 12/23/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent musculoskeletal disorder and a leading cause of disability globally. Although many efforts have been made to treat this condition, current tissue engineering (TE) and regenerative medicine strategies fail to address the inflammatory tissue environment that leads to the rapid progression of the disease and prevents cartilage tissue formation. First, this review addresses in detail the current anti-inflammatory therapies for OA with a special emphasis on pharmacological approaches, gene therapy, and mesenchymal stromal cell (MSC) intra-articular administration, and discusses the reasons behind the limited clinical success of these approaches at enabling cartilage regeneration. Then, we analyze the state-of-the-art TE strategies and how they can be improved by incorporating immunomodulatory capabilities such as the optimization of biomaterial composition, porosity and geometry, and the loading of anti-inflammatory molecules within an engineered structure. Finally, the review discusses the future directions for the new generation of TE strategies for OA treatment, specifically focusing on the spatiotemporal modulation of anti-inflammatory agent presentation to allow for tailored patient-specific therapies. Impact statement Osteoarthritis (OA) is a prevalent and debilitating musculoskeletal disorder affecting millions worldwide. Despite significant advancements in regenerative medicine and tissue engineering (TE), mitigating inflammation while simultaneously promoting cartilage tissue regeneration in OA remains elusive. In this review article, we discuss current anti-inflammatory therapies and explore their potential synergy with cutting-edge cartilage TE strategies, with a special focus on novel spatiotemporal and patient-specific anti-inflammatory strategies.
Collapse
Affiliation(s)
- Orlaith Kennedy
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
| | - Andrew Kitson
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Chiebuka Okpara
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Lesley W Chow
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | | |
Collapse
|
4
|
Zhang Y, Yi D, Hong Q, Cao J, Geng X, Liu J, Xu C, Cao M, Chen C, Xu S, Zhang Z, Li M, Zhu Y, Peng N. Platelet-rich plasma-derived exosomes boost mesenchymal stem cells to promote peripheral nerve regeneration. J Control Release 2024; 367:265-282. [PMID: 38253204 DOI: 10.1016/j.jconrel.2024.01.043] [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: 09/15/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
Peripheral nerve injury (PNI) remains a severe clinical problem with debilitating consequences. Mesenchymal stem cell (MSC)-based therapy is promising, but the problems of poor engraftment and insufficient neurotrophic effects need to be overcome. Herein, we isolated platelet-rich plasma-derived exosomes (PRP-Exos), which contain abundant bioactive molecules, and investigated their potential to increase the regenerative capacity of MSCs. We observed that PRP-Exos significantly increased MSC proliferation, viability, and mobility, decreased MSC apoptosis under stress, maintained MSC stemness, and attenuated MSC senescence. In vivo, PRP-Exo-treated MSCs (pExo-MSCs) exhibited an increased retention rate and heightened therapeutic efficacy, as indicated by increased axonal regeneration, remyelination, and recovery of neurological function in a PNI model. In vitro, pExo-MSCs coculture promoted Schwann cell proliferation and dorsal root ganglion axon growth. Moreover, the increased neurotrophic behaviour of pExo-MSCs was mediated by trophic factors, particularly glia-derived neurotrophic factor (GDNF), and PRP-Exos activated the PI3K/Akt signalling pathway in MSCs, leading to the observed phenotypes. These findings demonstrate that PRP-Exos may be novel agents for increasing the ability of MSCs to promote neural repair and regeneration in patients with PNI.
Collapse
Affiliation(s)
- Yongyi Zhang
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China; State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; No.962 Hospital of the PLA Joint Logistic Support Force, Harbin 150080, China
| | - Dan Yi
- Medical School of Chinese PLA, Beijing 100853, China; Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Quan Hong
- State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jiangbei Cao
- Departments of Anaesthesiology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaodong Geng
- State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jinwei Liu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chuang Xu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Mengyu Cao
- Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Chen
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Shuaixuan Xu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhen Zhang
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Molin Li
- Medical School of Chinese PLA, Beijing 100853, China; Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yaqiong Zhu
- Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China.
| | - Nan Peng
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
5
|
Yun HH, Kim SG, Park SI, Jo W, Kang KK, Lee EJ, Kim DK, Jung HS, Son JY, Park JM, Park HS, Lee S, Shin HI, Hong IH, Jeong KS. Early Osteogenic-Induced Adipose-Derived Stem Cells and Canine Bone Regeneration Potential Analyzed Using Biodegradable Scaffolds. Bioengineering (Basel) 2023; 10:1311. [PMID: 38002434 PMCID: PMC10669612 DOI: 10.3390/bioengineering10111311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
The complex process of bone regeneration is influenced by factors such as inflammatory responses, tissue interactions, and progenitor cells. Currently, multiple traumas can interfere with fracture healing, causing the prolonging or failure of healing. In these cases, bone grafting is the most effective treatment. However, there are several drawbacks, such as morbidity at the donor site and availability of suitable materials. Advantages have been provided in this field by a variety of stem cell types. Adipose-derived stem cells (ASCs) show promise. In the radiological examination of this study, it was confirmed that the C/S group showed faster regeneration than the other groups, and Micro-CT also showed that the degree of bone formation in the defect area was highest in the C/S group. Compared to the control group, the change in cortical bone area in the defect area decreased in the sham group (0.874), while it slightly increased in the C/S group (1.027). An increase in relative vascularity indicates a decrease in overall bone density, but a weak depression filled with fibrous tissue was observed outside the compact bone. It was confirmed that newly formed cortical bone showed a slight difference in bone density compared to surrounding normal bone tissue due to increased distribution of cortical bone. In this study, we investigated the effect of bone regeneration by ADMSCs measured by radiation and pathological effects. These data can ultimately be applied to humans with important clinical applications in various bone diseases, regenerative, and early stages of formative differentiation.
Collapse
Affiliation(s)
- Hyun-Ho Yun
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea; (H.-H.Y.); (K.-K.K.); (E.-J.L.); (J.-Y.S.); (J.-M.P.)
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (S.-G.K.); (W.J.); (D.-K.K.); (H.-S.J.)
| | - Seong-Gon Kim
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (S.-G.K.); (W.J.); (D.-K.K.); (H.-S.J.)
| | - Se-Il Park
- Cardiovascular Product Evaluation Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Woori Jo
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (S.-G.K.); (W.J.); (D.-K.K.); (H.-S.J.)
| | - Kyung-Ku Kang
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea; (H.-H.Y.); (K.-K.K.); (E.-J.L.); (J.-Y.S.); (J.-M.P.)
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (S.-G.K.); (W.J.); (D.-K.K.); (H.-S.J.)
| | - Eun-Joo Lee
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea; (H.-H.Y.); (K.-K.K.); (E.-J.L.); (J.-Y.S.); (J.-M.P.)
| | - Dong-Kyu Kim
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (S.-G.K.); (W.J.); (D.-K.K.); (H.-S.J.)
| | - Hoe-Su Jung
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (S.-G.K.); (W.J.); (D.-K.K.); (H.-S.J.)
| | - Ji-Yoon Son
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea; (H.-H.Y.); (K.-K.K.); (E.-J.L.); (J.-Y.S.); (J.-M.P.)
| | - Jae-Min Park
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea; (H.-H.Y.); (K.-K.K.); (E.-J.L.); (J.-Y.S.); (J.-M.P.)
| | - Hyun-Sook Park
- Cell Engineering for Origin Research Center, Seoul 03150, Republic of Korea; (H.-S.P.); (S.L.)
| | - Sunray Lee
- Cell Engineering for Origin Research Center, Seoul 03150, Republic of Korea; (H.-S.P.); (S.L.)
| | - Hong-In Shin
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea;
| | - Il-Hwa Hong
- Department of Veterinary Pathology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Kyu-Shik Jeong
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea; (H.-H.Y.); (K.-K.K.); (E.-J.L.); (J.-Y.S.); (J.-M.P.)
- Institute for Next Generation Unified Technology, Hoseo University, Asan 31499, Republic of Korea
| |
Collapse
|
6
|
Li C, Hu Y. Extracellular Vesicles Derived from Mesenchymal Stem Cells as Cell-Free Therapy for Intrauterine Adhesion. Int J Stem Cells 2023; 16:260-268. [PMID: 37385632 PMCID: PMC10465336 DOI: 10.15283/ijsc21177] [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: 10/04/2021] [Accepted: 11/17/2021] [Indexed: 07/01/2023] Open
Abstract
Intrauterine adhesion (IUA) can occur after trauma to the basal layer of the endometrium, contributing to severe complications in females, such as infertility and amenorrhea. To date, the proposed therapeutic strategies are targeted to relieve IUA, such as hysteroscopic adhesiolysis, Foley catheter balloon, and hyaluronic acid injection have been applied in the clinic. However, these approaches showed limited effects in alleviating endometrial fibrosis and thin endometrium. Mesenchymal stem cells (MSCs) can offer the potential for endometrium regeneration owing to reduce inflammation and release growth factors. On this basis, MSCs have been proposed as promising methods to treat intrauterine adhesion. However, due to the drawbacks of cell therapy, the possible therapeutic use of extracellular vesicles released by stem cells is raising increasing interest. The paracrine effect, mediated by MSCs derived extracellular vehicles (MSC-EVs), has recently been suggested as a mechanism for their therapeutic properties. Here, we summarizes the main pathological mechanisms involved in intrauterine adhesion, the biogenesis and characteristics of extracellular vesicles, explaining how these vesicles could provide new opportunities for MSCs.
Collapse
Affiliation(s)
- Chao Li
- Department of Gynecology, Tianjin Medical University, Tianjin, China
- Department of Gynecology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Yuanjing Hu
- Department of Gynecology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| |
Collapse
|
7
|
Xu L, Mu J, Ma Z, Lin P, Xia F, Hu X, Wu J, Cao J, Liu S, Huang T, Ling D, Gao J, Li F. Nanozyme-Integrated Thermoresponsive In Situ Forming Hydrogel Enhances Mesenchymal Stem Cell Viability and Paracrine Effect for Efficient Spinal Cord Repair. ACS APPLIED MATERIALS & INTERFACES 2023; 15:37193-37204. [PMID: 37493513 DOI: 10.1021/acsami.3c06189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Mesenchymal stem cell (MSC)-based therapy has emerged as a promising strategy for the treatment of spinal cord injury (SCI). However, the hostile microenvironment of SCI, which can adversely affect the survival and paracrine effect of the implanted MSCs, severely limits the therapeutic efficacy of this approach. Here, we report on a ceria nanozyme-integrated thermoresponsive in situ forming hydrogel (CeNZ-gel) that can enable dual enhancement of MSC viability and paracrine effect, leading to highly efficient spinal cord repair. The sol-gel transition property of the CeNZ-gel at body temperature ensures uniform coverage of the hydrogel in injured spinal cord tissues. Our results demonstrate that the CeNZ-gel significantly increases the viability of transplanted MSCs in the microenvironment by attenuating oxidative stress and, more importantly, promotes the secretion of angiogenic factors from MSCs by inducing autophagy of MSCs. The synergy between the oxidative stress-relieving effect of CeNZs and the paracrine effect of MSCs accelerates angiogenesis, nerve repair, and motor function recovery after SCI, providing an efficient strategy for MSC-based SCI therapy.
Collapse
Affiliation(s)
- Lilan Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiafu Mu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhiyuan Ma
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Lin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fan Xia
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xi Hu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jiahe Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jian Cao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shanbiao Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianchen Huang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Daishun Ling
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo University, Zhejiang 315010, China
| | - Fangyuan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
8
|
Mori da Cunha MGMC, van der Veer BK, Giacomazzi G, Mackova K, Cattani L, Koh KP, Vande Velde G, Gijsbers R, Albersen M, Sampaolesi M, Deprest J. VEGF overexpressed mesoangioblasts enhance urethral and vaginal recovery following simulated vaginal birth in rats. Sci Rep 2023; 13:8622. [PMID: 37244975 DOI: 10.1038/s41598-023-35809-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Vaginal birth causes pelvic floor injury which may lead to urinary incontinence. Cell therapy has been proposed to assist in functional recovery. We aim to assess if intra-arterial injection of rat mesoangioblasts (MABs) and stable Vascular Endothelial Growth Factor (VEGF)-expressing MABs, improve recovery of urethral and vaginal function following simulated vaginal delivery (SVD). Female rats (n = 86) were assigned to either injection of saline (control), allogeneic-MABs (MABsallo), autologous-MABs (MABsauto) or allogeneic-MABs transduced to stably expressed VEGF (MABsallo-VEGF). One hour after SVD, 0.5 × 106 MABs or saline were injected into the aorta. Primary outcome was urethral (7d and 14d) and vaginal (14d) function; others were bioluminescent imaging for cell tracking (1, 3 and 7d), morphometry (7, 14 and 60d) and mRNAseq (3 and 7d). All MABs injected rats had external urethral sphincter and vaginal function recovery within 14d, as compared to only half of saline controls. Functional recovery was paralleled by improved muscle regeneration and microvascularization. Recovery rate was not different between MABsallo and MABsauto. MABsallo-VEGF accelerated functional recovery and increased GAP-43 expression at 7d. At 3d we detected major transcriptional changes in the urethra of both MABsallo and MABsallo-VEGF-injected animals, with upregulation of Rho/GTPase activity, epigenetic factors and dendrite development. MABSallo also upregulated transcripts that encode proteins involved in myogenesis and downregulated pro-inflammatory processes. MABsallo-VEGF also upregulated transcripts that encode proteins involved in neuron development and downregulated genes involved in hypoxia and oxidative stress. At 7d, urethras of MABsallo-VEGF-injected rats showed downregulation of oxidative and inflammatory response compared to MABSallo. Intra-arterial injection of MABsallo-VEGF enhances neuromuscular regeneration induced by untransduced MABs and accelerates the functional urethral and vaginal recovery after SVD.
Collapse
Affiliation(s)
- Marina G M C Mori da Cunha
- Group Biomedical Sciences, Centre for Surgical Technologies, KU Leuven, Leuven, Belgium.
- Group Biomedical Sciences, Woman and Child, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
- Department of Development and Regeneration, Experimental Gynecology Laboratory -Lok 05.30 ON3, Herestraat 49, Leuven, Belgium.
| | - Bernard K van der Veer
- Laboratory for Stem Cell and Developmental Epigenetics, Department of Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Giorgia Giacomazzi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology Unit, Department Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Katerina Mackova
- Group Biomedical Sciences, Centre for Surgical Technologies, KU Leuven, Leuven, Belgium
- Group Biomedical Sciences, Woman and Child, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Third Faculty of Medicine, Institute for the Care of the Mother and Child, Charles University, Prague, Czech Republic
| | - Laura Cattani
- Group Biomedical Sciences, Centre for Surgical Technologies, KU Leuven, Leuven, Belgium
- Group Biomedical Sciences, Woman and Child, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Kian Peng Koh
- Laboratory for Stem Cell and Developmental Epigenetics, Department of Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Department of Imaging and Pathology, Biomedical MRI/Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven, Belgium
| | - Rik Gijsbers
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Flanders, Belgium
- Leuven Viral Vector Core, KU Leuven, Leuven, Belgium
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology Unit, Department Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Jan Deprest
- Group Biomedical Sciences, Centre for Surgical Technologies, KU Leuven, Leuven, Belgium
- Group Biomedical Sciences, Woman and Child, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Pelvic Floor Unit, University Hospitals KU Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Zhu Y, Yang K, Cheng Y, Liu Y, Gu R, Liu X, Liu H, Zhang X, Liu Y. Apoptotic Vesicles Regulate Bone Metabolism via the miR1324/SNX14/SMAD1/5 Signaling Axis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205813. [PMID: 36670083 DOI: 10.1002/smll.202205813] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/17/2022] [Indexed: 06/17/2023]
Abstract
Mesenchymal stem cells (MSCs) are widely used in the treatment of diseases. After their in vivo application, MSCs undergo apoptosis and release apoptotic vesicles (apoVs). This study investigates the role of apoVs derived from human bone marrow mesenchymal stem cells (hBMMSCs) in bone metabolism and the molecular mechanism of the observed effects. The results show that apoVs can promote osteogenesis and inhibit osteoclast formation in vitro and in vivo. ApoVs may therefore attenuate the bone loss caused by primary and secondary osteoporosis and stimulate bone regeneration in areas of bone defect. The mechanisms responsible for apoV-induced bone regeneration include the release of miR1324, which inhibit expression of the target gene Sorting Nexin 14 (SNX14) and thus activate the SMAD1/5 pathway in target cells. Given that MSC-derived apoVs are easily obtained and stored, with low risks of immunological rejection and neoplastic transformation, The findings suggest a novel therapeutic strategy to treat bone loss, including via cell-free approaches to bone tissue engineering.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Kunkun Yang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Yawen Cheng
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Yaoshan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Ranli Gu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Xuenan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Hao Liu
- The Central Laboratory, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| |
Collapse
|
10
|
Jiang Z, Xu Y, Fu M, Zhu D, Li N, Yang G. Genetically modified cell spheroids for tissue engineering and regenerative medicine. J Control Release 2023; 354:588-605. [PMID: 36657601 DOI: 10.1016/j.jconrel.2023.01.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023]
Abstract
Cell spheroids offer cell-to-cell interactions and show advantages in survival rate and paracrine effect to solve clinical and biomedical inquiries ranging from tissue engineering and regenerative medicine to disease pathophysiology. Therefore, cell spheroids are ideal vehicles for gene delivery. Genetically modified spheroids can enhance specific gene expression to promote tissue regeneration. Gene deliveries to cell spheroids are via viral vectors or non-viral vectors. Some new technologies like CRISPR/Cas9 also have been used in genetically modified methods to deliver exogenous gene to the host chromosome. It has been shown that genetically modified cell spheroids had the potential to differentiate into bone, cartilage, vascular, nerve, cardiomyocytes, skin, and skeletal muscle as well as organs like the liver to replace the diseased organ in the animal and pre-clinical trials. This article reviews the recent articles about genetically modified spheroid cells and explains the fabrication, applications, development timeline, limitations, and future directions of genetically modified cell spheroid.
Collapse
Affiliation(s)
- Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Yi Xu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Mengdie Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Na Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
11
|
Farooqui N, Mohan A, Isik B, Goksu BB, Thaler R, Zhu XY, Krier JD, Saadiq IM, Ferguson CM, Jordan KL, Tang H, Textor SC, Hickson LTJ, van Wijnen AJ, Eirin A, Lerman LO, Herrmann SM. Effect of Hypoxia Preconditioning on the Regenerative Capacity of Adipose Tissue Derived Mesenchymal Stem Cells in a Model of Renal Artery Stenosis. Stem Cells 2023; 41:50-63. [PMID: 36250949 PMCID: PMC9887092 DOI: 10.1093/stmcls/sxac073] [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: 04/18/2022] [Accepted: 08/26/2022] [Indexed: 02/02/2023]
Abstract
Atherosclerotic renal artery stenosis (ARAS) is associated with irreversible parenchymal renal disease and regenerative stem cell therapies may improve renal outcomes. Hypoxia preconditioning (HPC) may improve the regenerative functions of adipose tissue-derived mesenchymal stem cells (AMSC) by affecting DNA 5-hydroxymethylcytosine (5hmC) marks in angiogenic genes. Here, we investigated using a porcine ARAS model, whether growth of ARAS AMSCs in hypoxia (Hx) versus normoxia (Nx) would enhance renal tissue repair, and comprehensively analyze how HPC modifies DNA hydroxymethylation compared to untreated ARAS and healthy/normal pigs (n=5 each). ARAS pigs exhibited elevated serum cholesterol, serum creatinine and renal artery stenosis, with a concomitant decrease in renal blood flow (RBF) and increased blood pressure (BP) compared to healthy pigs. Renal artery injection of either autologous Nx or Hx AMSCs improved diastolic BP, reduced kidney tissue fibrosis, and inflammation (CD3+ T-cells) in ARAS pigs. In addition, renal medullary hypoxia significantly lowered with Nx but not Hx AMSC treatment. Mechanistically, levels of epigenetic 5hmC marks (which reflect gene activation) estimated using DNA immunoprecipitation technique were elevated in profibrotic and inflammatory genes in ARAS compared with normal AMSCs. HPC significantly reduced 5hmC levels in cholesterol biosynthesis and oxidative stress response pathways in ARAS AMSCs. Thus, autologous AMSCs improve key renovascular parameters and inflammation in ARAS pigs, with HPC mitigating pathological molecular effects on inflammatory and profibrotic genes which may play a role in augmenting regenerative capacity of AMSCs.
Collapse
Affiliation(s)
- Naba Farooqui
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Arjunmohan Mohan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Busra Isik
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Busra B Goksu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Xiang Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - James D Krier
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | | | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - La Tonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | | | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
12
|
Intra-Articular Mesenchymal Stem Cell Injection for Knee Osteoarthritis: Mechanisms and Clinical Evidence. Int J Mol Sci 2022; 24:ijms24010059. [PMID: 36613502 PMCID: PMC9819973 DOI: 10.3390/ijms24010059] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Knee osteoarthritis presents higher incidences than other joints, with increased prevalence during aging. It is a progressive process and may eventually lead to disability. Mesenchymal stem cells (MSCs) are expected to repair damaged issues due to trilineage potential, trophic effects, and immunomodulatory properties of MSCs. Intra-articular MSC injection was reported to treat knee osteoarthritis in many studies. This review focuses on several issues of intra-articular MSC injection for knee osteoarthritis, including doses of MSCs applied for injection and the possibility of cartilage regeneration following MSC injection. Intra-articular MSC injection induced hyaline-like cartilage regeneration, which could be seen by arthroscopy in several studies. Additionally, anatomical, biomechanical, and biochemical changes during aging and other causes participate in the development of knee osteoarthritis. Conversely, appropriate intervention based on these anatomical, biomechanical, biochemical, and functional properties and their interactions may postpone the progress of knee OA and facilitate cartilage repair induced by MSC injection. Hence, post-injection rehabilitation programs and related mechanisms are discussed.
Collapse
|
13
|
Augmenting Peripheral Nerve Regeneration with Adipose-Derived Stem Cells. Stem Cell Rev Rep 2022; 18:544-558. [PMID: 34417730 PMCID: PMC8858329 DOI: 10.1007/s12015-021-10236-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 02/03/2023]
Abstract
Peripheral nerve injuries (PNIs) are common and debilitating, cause significant health care costs for society, and rely predominately on autografts, which necessitate grafting a nerve section non-locally to repair the nerve injury. One possible approach to improving treatment is bolstering endogenous regenerative mechanisms or bioengineering new nervous tissue in the peripheral nervous system. In this review, we discuss critical-sized nerve gaps and nerve regeneration in rats, and summarize the roles of adipose-derived stem cells (ADSCs) in the treatment of PNIs. Several regenerative treatment modalities for PNI are described: ADSCs differentiating into Schwann cells (SCs), ADSCs secreting growth factors to promote peripheral nerve growth, ADSCs promoting myelination growth, and ADSCs treatments with scaffolds. ADSCs' roles in regenerative treatment and features are compared to mesenchymal stem cells, and the administration routes, cell dosages, and cell fates are discussed. ADSCs secrete neurotrophic factors and exosomes and can differentiate into Schwann cell-like cells (SCLCs) that share features with naturally occurring SCs, including the ability to promote nerve regeneration in the PNS. Future clinical applications are also discussed.
Collapse
|
14
|
Muscolino E, Di Stefano AB, Trapani M, Sabatino MA, Giacomazza D, Moschella F, Cordova A, Toia F, Dispenza C. Injectable xyloglucan hydrogels incorporating spheroids of adipose stem cells for bone and cartilage regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112545. [PMID: 34857257 DOI: 10.1016/j.msec.2021.112545] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/31/2021] [Accepted: 11/07/2021] [Indexed: 12/12/2022]
Abstract
Cartilage or bone regeneration approaches based on the direct injection of mesenchymal stem cells (MSCs) at the lesion site encounter several challenges, related to uncontrolled cell spreading and differentiation, reduced cell viability and poor engrafting. This work presents a simple and versatile strategy based on the synergic combination of in-situ forming hydrogels and spheroids of adipose stem cells (SASCs) with great potential for minimally invasive regenerative interventions aimed to threat bone and cartilage defects. Aqueous dispersions of partially degalactosylated xyloglucan (dXG) are mixed with SASCs derived from liposuction and either a chondroinductive or an osteoinductive medium. The dispersions rapidly set into hydrogels when temperature is brought to 37 °C. The physico-chemical and mechanical properties of the hydrogels are controlled by polymer concentration. The hydrogels, during 21 day incubation at 37 °C, undergo significant structural rearrangements that support cell proliferation and spreading. In formulations containing 1%w dXG cell viability increases up to 300% for SASCs-derived osteoblasts and up to 1000% for SASCs-derived chondrocytes if compared with control 2D cultures. The successful differentiation into the target cells is supported by the expression of lineage-specific genes. Cell-cell and cell-matrix interactions are also investigated. All formulations resulted injectable, and the incorporated cells are fully viable after injection.
Collapse
Affiliation(s)
- Emanuela Muscolino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy
| | - Anna Barbara Di Stefano
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Marco Trapani
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Maria Antonietta Sabatino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy
| | - Daniela Giacomazza
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146, Palermo, Italy
| | - Francesco Moschella
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy; Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Adriana Cordova
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy; Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Francesca Toia
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Clelia Dispenza
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy; Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146, Palermo, Italy.
| |
Collapse
|
15
|
Lu Y, Yu CH, Yang G, Sun N, Jiang F, Zhou M, Wu X, Luo J, Huang C, Zhang W, Jiang X. A rapidly magnetically assembled stem cell microtissue with "hamburger" architecture and enhanced vascularization capacity. Bioact Mater 2021; 6:3756-3765. [PMID: 33898876 PMCID: PMC8044908 DOI: 10.1016/j.bioactmat.2021.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 12/19/2022] Open
Abstract
With the development of magnetic manipulation technology based on magnetic nanoparticles (MNPs), scaffold-free microtissues can be constructed utilizing the magnetic attraction of MNP-labeled cells. The rapid in vitro construction and in vivo vascularization of microtissues with complex hierarchical architectures are of great importance to the viability and function of stem cell microtissues. Endothelial cells are indispensable for the formation of blood vessels and can be used in the prevascularization of engineered tissue constructs. Herein, safe and rapid magnetic labeling of cells was achieved by incubation with MNPs for 1 h, and ultrathick scaffold-free microtissues with different sophisticated architectures were rapidly assembled, layer by layer, in 5 min intervals. The in vivo transplantation results showed that in a stem cell microtissue with trisection architecture, the two separated human umbilical vein endothelial cell (HUVEC) layers would spontaneously extend to the stem cell layers and connect with each other to form a spatial network of functional blood vessels, which anastomosed with the host vasculature. The "hamburger" architecture of stem cell microtissues with separated HUVEC layers could promote vascularization and stem cell survival. This study will contribute to the construction and application of structural and functional tissues or organs in the future.
Collapse
Affiliation(s)
- Yuezhi Lu
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chun-Hua Yu
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Guangzheng Yang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ningjia Sun
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Fei Jiang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Mingliang Zhou
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiaolin Wu
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jiaxin Luo
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Cui Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedical Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, China
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
16
|
Zhang Y, Gao S, Liang K, Wu Z, Yan X, Liu W, Li J, Wu B, Du Y. Exendin-4 gene modification and microscaffold encapsulation promote self-persistence and antidiabetic activity of MSCs. SCIENCE ADVANCES 2021; 7:eabi4379. [PMID: 34215590 PMCID: PMC11060038 DOI: 10.1126/sciadv.abi4379] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/20/2021] [Indexed: 06/13/2023]
Abstract
Mesenchymal stem cell (MSC)-based therapy to combat diabetic-associated metabolic disorders is hindered by impoverished cell survival and limited therapeutic effects under high glucose stress. Here, we genetically engineered MSCs with Exendin-4 (MSC-Ex-4), a glucagon-like peptide-1 (GLP-1) analog, and demonstrated their boosted cellular functions and antidiabetic efficacy in the type 2 diabetes mellitus (T2DM) mouse model. Mechanistically, MSC-Ex-4 achieved self-augmentation and improved survival under high glucose stress via autocrine activation of the GLP-1R-mediated AMPK signaling pathway. Meanwhile, MSC-Ex-4-secreted Exendin-4 suppressed senescence and apoptosis of pancreatic β cells through endocrine effects, while MSC-Ex-4-secreted bioactive factors (e.g., IGFBP2 and APOM) paracrinely augmented insulin sensitivity and decreased lipid accumulation in hepatocytes through PI3K-Akt activation. Furthermore, we encapsulated MSC-Ex-4 in 3D gelatin microscaffolds for single-dose administration to extend the therapeutic effect for 3 months. Together, our findings provide mechanistic insights into Exendin-4-mediated MSCs self-persistence and antidiabetic activity that offer more effective MSC-based therapy for T2DM.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shuang Gao
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhaozhao Wu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaojun Yan
- Beijing CytoNiche Biotechnology Co. Ltd., Beijing 100195, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd., Beijing 100195, China
| | - Junyang Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bingjie Wu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
17
|
Lian M, Sun B, Han Y, Yu B, Xin W, Xu R, Ni B, Jiang W, Hao Y, Zhang X, Shen Y, Qiao Z, Dai K. A low-temperature-printed hierarchical porous sponge-like scaffold that promotes cell-material interaction and modulates paracrine activity of MSCs for vascularized bone regeneration. Biomaterials 2021; 274:120841. [PMID: 33984633 DOI: 10.1016/j.biomaterials.2021.120841] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/31/2021] [Accepted: 04/16/2021] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) secrete paracrine trophic factors that are beneficial for tissue regeneration. In this study, a sponge-like scaffold with hierarchical and interconnected pores was developed using low-temperature deposition modeling (LDM) printing. Its effects on the cellular behavior, especially on the paracrine secretion patterns of MSCs, were comprehensively investigated. We found that compared with the scaffolds printed via the fused deposition modeling (FDM) technique, the LDM-printed sponges enhanced the adhesion, retention, survival, and ingrowth of MSCs and promoted cell-material interactions. Moreover, the paracrine functions of the cultured MSCs on the LDM-printed sponges were improved, with significant secretion of upregulated immunomodulatory, angiogenic, and osteogenic factors. MSCs on the LDM-printed sponges exert beneficial paracrine effects on multiple regenerative processes, including macrophage polarization, tube formation, and osteogenesis, verifying the enhanced immunomodulatory, angiogenic, and osteogenic potential. Further protein function assays indicated that focal adhesion kinase (FAK), downstream AKT, and yes-associated-protein (YAP) signaling might participate in the required mechanotransductive pathways, through which the hierarchical porous structures stimulated the paracrine effects of MSCs. In a rat distal femoral defect model, the MSC-laden LDM-printed sponges significantly promoted vascularized bone regeneration. The results of the present study demonstrate that the hierarchical porous biomimetic sponges prepared via LDM printing have potential applications in tissue engineering based on their cell-material interaction promotion and MSC paracrine function modulation effects. Furthermore, our findings suggest that the optimization of biomaterial properties to direct the paracrine signaling of MSCs would enhance tissue regeneration.
Collapse
Affiliation(s)
- Meifei Lian
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Binbin Sun
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yu Han
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bin Yu
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Weiwei Xin
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China
| | - Ruida Xu
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China
| | - Bing Ni
- School of Life Science, East China Normal University, Shanghai, 200241, China
| | - Wenbo Jiang
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yongqiang Hao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiuyin Zhang
- Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi Shen
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China.
| | - Zhiguang Qiao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China.
| | - Kerong Dai
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
18
|
Murata Y, Jo JI, Tabata Y. Visualization of Apoptosis in Three-Dimensional Cell Aggregates Based on Molecular Beacon Imaging. Tissue Eng Part C Methods 2021; 27:264-275. [PMID: 33619986 DOI: 10.1089/ten.tec.2020.0338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The objective of this study is to visualize cell apoptosis in three-dimensional (3D) cell aggregates based on molecular beacons (MB). Two types of MB for messenger RNA were used: caspase-3 MB as a target for apoptosis and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) MB as a control of stable fluorescence in cells. To enhance the MB internalization into cells, caspase-3 and GAPDH MB were incorporated in cationized gelatin nanospheres (cGNS), respectively (cGNScasp3 MB and cGNSGAP MB). In addition, cGNS co-incorporating caspase-3 and GAPDH MB (cGNSdual MB) were prepared to perform the dual-color imaging for the same cell aggregate. The cGNSMB were incubated with mouse mesenchymal stem cells to label with MB in the two-dimensional culture. The cell apoptosis mediated by the addition of antibody for a death receptor Fas was ratiometrically detected by the cGNSdual MB to the same extent as single MB. The cell aggregates were prepared from MB-labeled cells, and the MB fluorescence was detected from almost all the cells even in the 3D aggregates to show the homogenous distribution. In addition to the Fas-mediated apoptosis, the aggregates were treated with camptothecin of a low-molecular weight apoptosis inducer. The fluorescence of caspase-3 MB was mainly distributed at the surface surrounding site of Fas-mediated apoptotic aggregates rather than the center site, while that of GAPDH MB was detected even in the interior site. On the other hand, in the camptothecin-induced apoptotic aggregates, both caspae-3 and GAPDH MB fluorescence were detected from the interior site of aggregates as well as the surrounding site. It is likely that the MB fluorescence reflected the localization of apoptotic position caused by the different molecular sizes of apoptosis inducer and the consequent penetration into the aggregates. It is concluded that the cGMSMB are a promising system to visualize cell apoptosis in 3D cell aggregates without the destruction of aggregates.
Collapse
Affiliation(s)
- Yuki Murata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Jun-Ichiro Jo
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
19
|
Toia F, Di Stefano AB, Muscolino E, Sabatino MA, Giacomazza D, Moschella F, Cordova A, Dispenza C. In-situ gelling xyloglucan formulations as 3D artificial niche for adipose stem cell spheroids. Int J Biol Macromol 2020; 165:2886-2899. [PMID: 33470202 DOI: 10.1016/j.ijbiomac.2020.10.158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022]
Abstract
Three-dimensional spheroidal cell aggregates of adipose stem cells (SASCs) are a distinct upstream population of stem cells present in adipose tissue, with enhanced regeneration properties in vivo. The preservation of the 3D structure of the cells, from extraction to administration, can be a promising strategy to ensure optimal conditions for cell viability and maintenance of stemness potential. With this aim, an artificial niche was created by incorporating the spheroids into an injectable, in-situ gelling solution of partially degalactosylated xyloglucan (dXG) and an ad hoc formulated culture medium for the preservation of stem cell spheroid features. The evolution of the mechanical properties and the morphological structure of this artificial niche was investigated by small amplitude rheological analysis and scanning electron microscopy, respectively. Comparatively, systems produced with the same polymer and the typical culture medium (DMEM) used for adipose stem cell (ASC) growth in adherent cell culture conditions were also characterised. Cell viability of both SASCs and ASCs incorporated inside the hydrogel or seeded on top of the hydrogel were investigated as well as the preservation of SASC stemness conditions when embedded in the hydrogel.
Collapse
Affiliation(s)
- F Toia
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy; BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - A B Di Stefano
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - E Muscolino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy
| | - M A Sabatino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy
| | - D Giacomazza
- Istituto di BioFisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - F Moschella
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - A Cordova
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy; BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - C Dispenza
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy; Istituto di BioFisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy.
| |
Collapse
|
20
|
Laurent A, Hirt-Burri N, Scaletta C, Michetti M, de Buys Roessingh AS, Raffoul W, Applegate LA. Holistic Approach of Swiss Fetal Progenitor Cell Banking: Optimizing Safe and Sustainable Substrates for Regenerative Medicine and Biotechnology. Front Bioeng Biotechnol 2020; 8:557758. [PMID: 33195124 PMCID: PMC7644790 DOI: 10.3389/fbioe.2020.557758] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
Safety, quality, and regulatory-driven iterative optimization of therapeutic cell source selection has constituted the core developmental bedrock for primary fetal progenitor cell (FPC) therapy in Switzerland throughout three decades. Customized Fetal Transplantation Programs were pragmatically devised as straightforward workflows for tissue procurement, traceability maximization, safety, consistency, and robustness of cultured progeny cellular materials. Whole-cell bioprocessing standardization has provided plethoric insights into the adequate conjugation of modern biotechnological advances with current restraining legislative, ethical, and regulatory frameworks. Pioneer translational advances in cutaneous and musculoskeletal regenerative medicine continuously demonstrate the therapeutic potential of FPCs. Extensive technical and clinical hindsight was gathered by managing pediatric burns and geriatric ulcers in Switzerland. Concomitant industrial transposition of dermal FPC banking, following good manufacturing practices, demonstrated the extensive potential of their therapeutic value. Furthermore, in extenso, exponential revalorization of Swiss FPC technology may be achieved via the renewal of integrative model frameworks. Consideration of both longitudinal and transversal aspects of simultaneous fetal tissue differential processing allows for a better understanding of the quasi-infinite expansion potential within multi-tiered primary FPC banking. Multiple fetal tissues (e.g., skin, cartilage, tendon, muscle, bone, lung) may be simultaneously harvested and processed for adherent cell cultures, establishing a unique model for sustainable therapeutic cellular material supply chains. Here, we integrated fundamental, preclinical, clinical, and industrial developments embodying the scientific advances supported by Swiss FPC banking and we focused on advances made to date for FPCs that may be derived from a single organ donation. A renewed model of single organ donation bioprocessing is proposed, achieving sustained standards and potential production of billions of affordable and efficient therapeutic doses. Thereby, the aim is to validate the core therapeutic value proposition, to increase awareness and use of standardized protocols for translational regenerative medicine, potentially impacting millions of patients suffering from cutaneous and musculoskeletal diseases. Alternative applications of FPC banking include biopharmaceutical therapeutic product manufacturing, thereby indirectly and synergistically enhancing the power of modern therapeutic armamentariums. It is hypothesized that a single qualifying fetal organ donation is sufficient to sustain decades of scientific, medical, and industrial developments, as technological optimization and standardization enable high efficiency.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
- Tec-Pharma SA, Bercher, Switzerland
- LAM Biotechnologies SA, Épalinges, Switzerland
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Anthony S. de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Wassim Raffoul
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
- Oxford Suzhou Center for Advanced Research, Science and Technology Co., Ltd., Oxford University, Suzhou, China
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Yang Z, Zheng C, Zhang F, Lin B, Cao M, Tian X, Zhang J, Zhang X, Shen J. Magnetic resonance imaging of enhanced nerve repair with mesenchymal stem cells combined with microenvironment immunomodulation in neurotmesis. Muscle Nerve 2020; 61:815-825. [PMID: 32170960 DOI: 10.1002/mus.26862] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 02/04/2020] [Accepted: 03/11/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The immuno-microenvironment of injured nerves adversely affects mesenchymal stem cell (MSC) therapy for neurotmesis. Magnetic resonance imaging (MRI) can be used noninvasively to monitor nerve degeneration and regeneration. The aim of this study was to investigate nerve repair after MSC transplantation combined with microenvironment immunomodulation in neurotmesis by using multiparametric MRI. METHODS Rats with sciatic nerve transection and surgical coaptation were treated with MSCs combined with immunomodulation or MSCs alone. Serial multiparametric MRI examinations were performed over an 8-week period after surgery. RESULTS Nerves treated with MSCs combined with immunomodulation showed better functional recovery, rapid recovery of nerve T2, fractional anisotropy and radial diffusivity values, and more rapid restoration of the fiber tracks than nerves treated with MSCs alone. DISCUSSION Transplantation of MSCs in combination with immunomodulation can exert a synergistic repair effect on neurotmesis, which can be monitored by multiparametric MRI.
Collapse
Affiliation(s)
- Zehong Yang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chushan Zheng
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Binglin Lin
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Minghui Cao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xuwei Tian
- Department of Radiology, The First People's Hospital of Kashgar, Kashgar, China
| | - Jingzhong Zhang
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Xiao Zhang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
22
|
Ding S, Xu Y, Yan X, Lin Y, Tan Q. Effect of Collagen Scaffold With Bcl-2-Modified Adipose-Derived Stem Cells on Diabetic Mice Wound Healing. INT J LOW EXTR WOUND 2019; 19:139-147. [PMID: 31680592 DOI: 10.1177/1534734619880055] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
This study aimed at evaluating the effects of collagen scaffold with Bcl-2-modified adipose-derived stem cells (ADSCs) on wound repair in streptozotocin-induced diabetic mice. A round full thickness skin defect with a diameter of 7 mm was made in the mice model. The experimental mice were divided into 4 groups (n = 12 each): group A (control group), group B (scaffold group), group C (ADSCs-scaffold group), and group D (Bcl-2-ADSCs-scaffold group). On days 3, 7, 10, and 14 after surgery, characteristics of wound healing was observed, and wound tissues were sampled for histology characteristics via hematoxylin-eosin staining and immunohistochemical staining. Compared with other groups, the wound healing rate was significantly higher in group D a week after operation (P < .05). On the seventh day postoperation, group D exhibited higher blood vessel in the wounds granulation tissue than other groups according to results of hematoxylin-eosin staining and immunohistochemistry. In conclusion, these findings demonstrated that collagen scaffold with Bcl-2 modified ADSCs may effectively improve the wound healing process in diabetic mice.
Collapse
Affiliation(s)
- Sheng Ding
- Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China.,Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Ye Xu
- Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Xin Yan
- Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Yue Lin
- Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Qian Tan
- Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| |
Collapse
|
23
|
Ma DJ, Lim MS, Park UC, Park JB, Ji SY, Yu HG. Magnetic Iron Oxide Nanoparticle Labeling of Photoreceptor Precursors for Magnetic Resonance Imaging. Tissue Eng Part C Methods 2019; 25:532-542. [PMID: 31418341 DOI: 10.1089/ten.tec.2019.0136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IMPACT STATEMENT This study describes the methods and results of superparamagnetic iron oxide nanoparticle (SPION) labeling and magnetic resonance imaging (MRI) tracking of human embryonic stem cell-derived photoreceptor precursors transplanted into the subretinal space of Royal College of Surgeons rats. SPION labeling and MRI tracking provide information about the biodistribution of transplanted photoreceptor precursors, which is necessary for improving the functional benefits of cell therapy for degenerative retinal diseases.
Collapse
Affiliation(s)
- Dae Joong Ma
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul, Republic of Korea.,Retinal Degeneration Research Lab, Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea
| | - Mi-Sun Lim
- R&D Center, Jeil Pharmaceutical Co., Ltd., Yongin-si, Republic of Korea.,Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Un Chul Park
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul, Republic of Korea.,Retinal Degeneration Research Lab, Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea
| | - Jung-Bum Park
- Retinal Degeneration Research Lab, Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea
| | - So Yeon Ji
- R&D Center, Jeil Pharmaceutical Co., Ltd., Yongin-si, Republic of Korea.,Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyeong Gon Yu
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul, Republic of Korea.,Retinal Degeneration Research Lab, Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea.,Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
24
|
Rbia N, Bulstra LF, Thaler R, Hovius SER, van Wijnen AJ, Shin AY. In Vivo Survival of Mesenchymal Stromal Cell-Enhanced Decellularized Nerve Grafts for Segmental Peripheral Nerve Reconstruction. J Hand Surg Am 2019; 44:514.e1-514.e11. [PMID: 30301645 DOI: 10.1016/j.jhsa.2018.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/09/2018] [Accepted: 07/18/2018] [Indexed: 02/02/2023]
Abstract
PURPOSE Adipose-derived mesenchymal stromal cells (MSCs) have emerged as promising tools for peripheral nerve reconstruction. There is a paucity of information regarding the ultimate survivorship of implanted MSCs or whether these cells remain where they are placed. The aim of the present study was to track the in vivo distribution and survival of MSCs seeded on a decellularized nerve allograft reconstruction of a peripheral nerve defect using luciferase-based bioluminescence imaging (BLI). METHODS To determine the in vivo survivability of MSCs, autologous Lewis rat MSCs were stably labeled with luciferase by lentiviral particles. Labeled cells were dynamically seeded onto a Sprague Dawley decellularized rat nerve allograft and used to bridge a 10-mm sciatic nerve defect. The MSC survival was determined by performing in vivo BLI to detect living cells. Twelve animals were examined at 24 hours after implantation, 3, 7, 9, 11, and 14 days, and at daily intervals thereafter if signals were still present. RESULTS Labeled MSCs could be detected for up to 29 days. Gradually diminishing BLI signals were observed within the first week following implantation. Implanted MSCs were not detected anywhere other than the site of surgery. CONCLUSIONS The MSCs seeded on decellularized nerve allografts can survive in vivo but have finite survival after implantation. There was no evidence of migration of MSCs to surrounding tissues. CLINICAL RELEVANCE The findings support a therapeutic approach that combines MSCs with a biological scaffold for peripheral nerve surgery. It provides understanding of the viability and distribution of implanted MSCs, which is a prerequisite before clinical translation can be considered.
Collapse
Affiliation(s)
- Nadia Rbia
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Liselotte F Bulstra
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | - Steven E R Hovius
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
25
|
Lopa S, Colombini A, Moretti M, de Girolamo L. Injective mesenchymal stem cell-based treatments for knee osteoarthritis: from mechanisms of action to current clinical evidences. Knee Surg Sports Traumatol Arthrosc 2019; 27:2003-2020. [PMID: 30159741 PMCID: PMC6541568 DOI: 10.1007/s00167-018-5118-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/14/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE Osteoarthritis (OA) represents a relevant social and economic burden worldwide. "Mesenchymal stem cells" or, as recently proposed, "medicinal signaling cells" (MSCs) have been recently introduced as injective treatments for OA with the aim of restoring joint homeostasis. The aim of this review is to provide the reader with the tools necessary to interpret the currently available clinical data, focusing on the MSC mechanisms of action which might help to clarify what we should expect from this treatment. METHODS Clinical studies reporting MSC injections for the treatment of knee OA, either freshly isolated or culture-expanded cells, have been included and commented in relation to the supposed therapeutic effect that MSCs might exert giving their supposed mode of actions. RESULTS The majority of the studies reports significant improvements in terms of pain and knee function compared to baseline values, up to 24 months of follow-up. Although these data support the expected therapeutic effect of this therapy giving the features of these cells, only 14% of the studies present a control group and more than one-third of them report the results on less than ten patients. CONCLUSIONS Despite the constant presence of positive and satisfactory results in the studies analyzed, the complexity of MSC metabolism and related therapeutic effects as well as the weakness of most of the studies do not allow withdrawing definitive conclusions about the superiority of one tissue source over another, as well as about the best cell dose and the long-term durability of the effects of these procedures. Given the high potential value of these therapies in the treatment of OA, further studies accurately designed, carefully defining the type of patients to be included and pursuing minimal standard requirements in terms of follow-up, number of patients, and types of measurements should be conducted to finally assess the efficacy of MSC-based injective treatments.
Collapse
Affiliation(s)
- Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy
| | - Alessandra Colombini
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900 Lugano, Switzerland
- Swiss Institute for Regenerative Medicine, Lugano, Switzerland
| | - Laura de Girolamo
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy
| |
Collapse
|
26
|
Kiaie N, Aghdam RM, Tafti SHA, Gorabi AM. Stem Cell-Mediated Angiogenesis in Tissue Engineering Constructs. Curr Stem Cell Res Ther 2018; 14:249-258. [PMID: 30394215 DOI: 10.2174/1574888x13666181105145144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/09/2018] [Accepted: 10/31/2018] [Indexed: 11/22/2022]
Abstract
Angiogenesis has always been a concern in the field of tissue engineering. Poor vascularization of engineered constructs is a problem for the clinical success of these structures. Among the various methods employed to induce angiogenesis, stem cells provide a promising tool for the future. The present review aims to present the application of stem cells in the induction of angiogenesis. Additionally, it summarizes recent advancements in stem cell-mediated angiogenesis of different tissue engineering constructs.
Collapse
Affiliation(s)
- Nasim Kiaie
- School of Metallurgy & Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran.,Department of Tissue Engineering, Amirkabir University of Technology, Tehran 15875, Iran
| | - Rouhollah M Aghdam
- School of Metallurgy & Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Seyed H Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Armita M Gorabi
- Department of Basic and Clinical Research, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Murata Y, Jo JI, Tabata Y. Preparation of cationized gelatin nanospheres incorporating molecular beacon to visualize cell apoptosis. Sci Rep 2018; 8:14839. [PMID: 30287861 PMCID: PMC6172245 DOI: 10.1038/s41598-018-33231-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 09/17/2018] [Indexed: 12/16/2022] Open
Abstract
The objective of this study is to prepare cationized gelatin nanospheres (cGNS) incorporating a molecular beacon (MB), and visualize cellular apoptosis. Two types of MB to detect the messenger RNA (mRNA) of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (GAP MB), and caspase-3 (casp3 MB) were incorporated in cGNS, respectively. MB incorporated in cGNS showed the DNA sequence specificity in hybridization. The cGNS incorporation enabled MB to enhance the stability against nuclease to a significantly great extent compared with free MB. The cGNS incorporating GAP MB were internalized into the KUM6 of a mouse bone marrow-derived stem cell by an endocytotic pathway. The cGNS were not distributed at the lysosomes. After the incubation with cGNS, the cell apoptosis was induced at different concentrations of camptothecin. No change in the intracellular fluorescence was observed for cGNSGAPMB. On the other hand, for the cGNScasp3MB, the fluorescent intensity significantly enhanced by the apoptosis induction of cells. It is concluded that cGNS incorporating MB is a promising system for the visualization of cellular apoptosis.
Collapse
Affiliation(s)
- Yuki Murata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Jun-Ichiro Jo
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
28
|
Farzamfar S, Salehi M, Ehterami A, Naseri-Nosar M, Vaez A, Zarnani AH, Sahrapeyma H, Shokri MR, Aleahmad M. Promotion of excisional wound repair by a menstrual blood-derived stem cell-seeded decellularized human amniotic membrane. Biomed Eng Lett 2018; 8:393-398. [PMID: 30603224 DOI: 10.1007/s13534-018-0084-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/19/2018] [Accepted: 09/04/2018] [Indexed: 02/06/2023] Open
Abstract
This is the first study demonstrating the efficacy of menstrual blood-derived stem cell (MenSC) transplantation via decellularized human amniotic membrane (DAM), for the promotion of skin excisional wound repair. The DAM was seeded with MenSCs at the density of 3 × 104 cells/cm2 and implanted onto a rat's 1.50 × 1.50 cm2 full-thickness excisional wound defect. The results of wound closure and histopathological examinations demonstrated that the MenSC-seeded DAM could significantly improve the wound healing compared with DAM-treatment. All in all, our data indicated that the MenSCs can be a potential source for cell-based therapies to regenerate skin injuries.
Collapse
Affiliation(s)
- Saeed Farzamfar
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Salehi
- 2Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,3Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Arian Ehterami
- 2Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mahdi Naseri-Nosar
- 3Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ahmad Vaez
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hassan Zarnani
- 4Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,5Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Hamed Sahrapeyma
- 6Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Shokri
- 7Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Aleahmad
- 4Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Ye X, Cheng S, Dong Y, Ren J, Su L, Liu J, Zhou J, Liu Q, Zhu N. Exendin-4 promotes proliferation of adipose-derived stem cells through PI3K/Akt-Wnt signaling pathways. Neurosci Lett 2018; 685:196-202. [PMID: 29920298 DOI: 10.1016/j.neulet.2018.06.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 06/15/2018] [Accepted: 06/15/2018] [Indexed: 01/27/2023]
Abstract
Adipose-derived stem cell (ADSC) transplantation has emerged as a potential tool for the treatment of cardiovascular disease and skin wounds. However, with a limited renewal capacity and the need for mass cells during the engraftment, strategies are needed to enhance ADSC proliferative capacity. In this study, we explored the effects of Exendin-4, a glucagon-like peptide-1 analog, on the growth of ADSCs, focusing in particular on phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt) and Wnt signaling pathways. Firstly, ADSCs were isolated and cultured in vitro. Then, flow cytometry demonstrated that ADSCs were positive for CD44, CD90 and CD29 but negative for CD31, CD34, and CD45. Exendin-4 (0-200 nM) treatment increased ADSC proliferation. In order to examine specific signaling pathways, a western blotting assay was performed. Our results demonstrate that after treated with 50 nM Exendin-4 for 48 h, the phosphorylation of PI3K, Akt, and GSK3β were increased and phosphorylation of β-catenin was decreased. From these results, we concluded that PI3K-Akt and Wnt-β-catenin signaling pathways mediate Exendin-4 induced ADSC proliferation, the function of which might contribute to the regulation of ADSC proliferation. Our findings provided new insights into the function of the mechanisms underlying Exendin-4 of ADSCs.
Collapse
Affiliation(s)
- Xiaolu Ye
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Shimeng Cheng
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Yabing Dong
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Jie Ren
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Lina Su
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Jianlan Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Jing Zhou
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Qingmei Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Ningwen Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China.
| |
Collapse
|
30
|
Zhang F, Duan X, Lu L, Zhang X, Chen M, Mao J, Cao M, Shen J. In Vivo Long-Term Tracking of Neural Stem Cells Transplanted into an Acute Ischemic Stroke model with Reporter Gene-Based Bimodal MR and Optical Imaging. Cell Transplant 2017; 26:1648-1662. [PMID: 29251112 PMCID: PMC5753979 DOI: 10.1177/0963689717722560] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 06/01/2017] [Accepted: 06/07/2017] [Indexed: 12/18/2022] Open
Abstract
Transplantation of neural stem cells (NSCs) is emerging as a new therapeutic approach for stroke. Real-time imaging of transplanted NSCs is essential for successful cell delivery, safety monitoring, tracking cell fate and function, and understanding the interactions of transplanted cells with the host environment. Magnetic resonance imaging (MRI) of magnetic nanoparticle-labeled cells has been the most widely used means to track stem cells in vivo. Nevertheless, it does not allow for the reliable discrimination between live and dead cells. Reporter gene-based MRI was considered as an alternative strategy to overcome this shortcoming. In this work, a class of lentiviral vector-encoding ferritin heavy chain (FTH) and enhanced green fluorescent protein (EGFP) was constructed to deliver reporter genes into NSCs. After these transgenic NSCs were transplanted into the contralateral hemisphere of rats with acute ischemic stroke, MRI and fluorescence imaging (FLI) were performed in vivo for tracking the fate of transplanted cells over a long period of 6 wk. The results demonstrated that the FTH and EGFP can be effectively and safely delivered to NSCs via the designed lentiviral vector. The distribution and migration of grafted stem cells could be monitored by bimodal MRI and FLI. FTH can be used as a robust MRI reporter for reliable reporting of the short-term viability of cell grafts, whereas its capacity for tracking the long-term viability of stem cells remains dependent on several confounding factors such as cell death and the concomitant reactive inflammation.
Collapse
Affiliation(s)
- Fang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaohui Duan
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Liejing Lu
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Meiwei Chen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jiaji Mao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Minghui Cao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Duan X, Lu L, Wang Y, Zhang F, Mao J, Cao M, Lin B, Zhang X, Shuai X, Shen J. The long-term fate of mesenchymal stem cells labeled with magnetic resonance imaging-visible polymersomes in cerebral ischemia. Int J Nanomedicine 2017; 12:6705-6719. [PMID: 28932115 PMCID: PMC5598550 DOI: 10.2147/ijn.s146742] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Understanding the long-term fate and potential mechanisms of mesenchymal stem cells (MSCs) after transplantation is essential for improving functional benefits of stem cell-based stroke treatment. Magnetic resonance imaging (MRI) is considered an attractive and clinically translatable tool for longitudinal tracking of stem cells, but certain controversies have arisen in this regard. In this study, we used SPION-loaded cationic polymersomes to label green fluorescent protein (GFP)-expressing MSCs to determine whether MRI can accurately reflect survival, long-term fate, and potential mechanisms of MSCs in ischemic stroke therapy. Our results showed that MSCs could improve the functional outcome and reduce the infarct volume of stroke in the brain. In vivo MRI can verify the biodistribution and migration of grafted cells when pre-labeled with SPION-loaded polymersome. The dynamic change of low signal volume on MRI can reflect the tendency of cell survival and apoptosis, but may overestimate long-term survival owing to the presence of iron-laden macrophages around cell graft. Only a small fraction of grafted cells survived up to 8 weeks after transplantation. A minority of these surviving cells were differentiated into astrocytes, but not into neurons. MSCs might exert their therapeutic effect via secreting paracrine factors rather than directing cell replacement through differentiation into neuronal and/or glial phenotypes.
Collapse
Affiliation(s)
- Xiaohui Duan
- Department of Radiology, Sun Yat-Sen Memorial Hospital
| | - Liejing Lu
- Department of Radiology, Sun Yat-Sen Memorial Hospital
| | - Yong Wang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering
| | - Fang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital
| | - Jiaji Mao
- Department of Radiology, Sun Yat-Sen Memorial Hospital
| | - Minghui Cao
- Department of Radiology, Sun Yat-Sen Memorial Hospital
| | - Bingling Lin
- Department of Radiology, Sun Yat-Sen Memorial Hospital
| | - Xiang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering
- BME Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital
| |
Collapse
|
32
|
Cortese FAB, Aguiar S, Santostasi G. Induced Cell Turnover: A Novel Therapeutic Modality for In Situ Tissue Regeneration. Hum Gene Ther 2017; 28:703-716. [PMID: 28557533 DOI: 10.1089/hum.2016.167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Induced cell turnover (ICT) is a theoretical intervention in which the targeted ablation of damaged, diseased, and/or nonfunctional cells is coupled with replacement by partially differentiated induced pluripotent stem cells in a gradual and multiphasic manner. Tissue-specific ablation can be achieved using pro-apoptotic small molecule cocktails, peptide mimetics, and/or tissue-tropic adeno-associated virus-delivered suicide genes driven by cell type-specific promoters. Replenishment with new cells can be mediated by systemic administration of cells engineered for homing, robustness, and even enhanced function and disease resistance. Otherwise, the controlled release of cells can be achieved using implanted biodegradable scaffolds, hydrogels, and polymer matrixes. In theory, ICT would enable in situ tissue regeneration without the need for surgical transplantation of organs produced ex vivo, and addresses non-transplantable tissues (such as the vasculature, lymph nodes, and the nervous system). This article outlines several complimentary strategies for overcoming barriers to ICT in an effort to stimulate further research at this promising interface of cell therapy, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Francesco Albert Bosco Cortese
- 1 Biogerontology Research Foundation, Oxford, United Kingdom .,2 Department of Biomedical and Molecular Sciences, Queen's University School of Medicine, Queen's University, Kingston, Canada
| | - Sebastian Aguiar
- 3 Neurobiology Department, Swammerdam Institute of Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Giovanni Santostasi
- 4 Department of Neurology, Feinberg School of Medicine, Northwestern University , Chicago, Illinois
| |
Collapse
|
33
|
Daldrup-Link HE, Chan C, Lenkov O, Taghavigarmestani S, Nazekati T, Nejadnik H, Chapelin F, Khurana A, Tong X, Yang F, Pisani L, Longaker M, Gambhir SS. Detection of Stem Cell Transplant Rejection with Ferumoxytol MR Imaging: Correlation of MR Imaging Findings with Those at Intravital Microscopy. Radiology 2017; 284:495-507. [PMID: 28128708 DOI: 10.1148/radiol.2017161139] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Purpose To determine whether endogenous labeling of macrophages with clinically applicable nanoparticles enables noninvasive detection of innate immune responses to stem cell transplants with magnetic resonance (MR) imaging. Materials and Methods Work with human stem cells was approved by the institutional review board and the stem cell research oversight committee, and animal experiments were approved by the administrative panel on laboratory animal care. Nine immunocompetent Sprague-Dawley rats received intravenous injection of ferumoxytol, and 18 Jax C57BL/6-Tg (Csf1r-EGFP-NGFR/FKBP1A/TNFRSF6) 2Bck/J mice received rhodamine-conjugated ferumoxytol. Then, 48 hours later, immune-matched or mismatched stem cells were implanted into osteochondral defects of the knee joints of experimental rats and calvarial defects of Jax mice. All animals underwent serial MR imaging and intravital microscopy (IVM) up to 4 weeks after surgery. Macrophages of Jax C57BL/6-Tg (Csf1r-EGFP-NGFR/FKBP1A/TNFRSF6) 2Bck/J mice express enhanced green fluorescent protein (GFP), which enables in vivo correlation of ferumoxytol enhancement at MR imaging with macrophage quantities at IVM. All quantitative data were compared between experimental groups by using a mixed linear model and t tests. Results Immune-mismatched stem cell implants demonstrated stronger ferumoxytol enhancement than did matched stem cell implants. At 4 weeks, T2 values of mismatched implants were significantly lower than those of matched implants in osteochondral defects of female rats (mean, 10.72 msec for human stem cells and 11.55 msec for male rat stem cells vs 15.45 msec for sex-matched rat stem cells; P = .02 and P = .04, respectively) and calvarial defects of recipient mice (mean, 21.7 msec vs 27.1 msec, respectively; P = .0444). This corresponded to increased recruitment of enhanced GFP- and rhodamine-ferumoxytol-positive macrophages into stem cell transplants, as visualized with IVM and histopathologic examination. Conclusion Endogenous labeling of macrophages with ferumoxytol enables noninvasive detection of innate immune responses to stem cell transplants with MR imaging. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Heike E Daldrup-Link
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Carmel Chan
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Olga Lenkov
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Seyedmeghdad Taghavigarmestani
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Toktam Nazekati
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Hossein Nejadnik
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Fanny Chapelin
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Aman Khurana
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Xinming Tong
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Fan Yang
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Laura Pisani
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Michael Longaker
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Sanjiv Sam Gambhir
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| |
Collapse
|
34
|
Bellio MA, Rodrigues CO, Landin AM, Hatzistergos KE, Kuznetsov J, Florea V, Valasaki K, Khan A, Hare JM, Schulman IH. Physiological and hypoxic oxygen concentration differentially regulates human c-Kit+ cardiac stem cell proliferation and migration. Am J Physiol Heart Circ Physiol 2016; 311:H1509-H1519. [PMID: 27694215 PMCID: PMC5206337 DOI: 10.1152/ajpheart.00449.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/24/2016] [Indexed: 02/07/2023]
Abstract
Cardiac stem cells (CSCs) are being evaluated for their efficacy in the treatment of heart failure. However, numerous factors impair the exogenously delivered cells' regenerative capabilities. Hypoxia is one stress that contributes to inadequate tissue repair. Here, we tested the hypothesis that hypoxia impairs cell proliferation, survival, and migration of human CSCs relative to physiological and room air oxygen concentrations. Human endomyocardial biopsy-derived CSCs were isolated, selected for c-Kit expression, and expanded in vitro at room air (21% O2). To assess the effect on proliferation, survival, and migration, CSCs were transferred to physiological (5%) or hypoxic (0.5%) O2 concentrations. Physiological O2 levels increased proliferation (P < 0.05) but did not affect survival of CSCs. Although similar growth rates were observed in room air and hypoxia, a significant reduction of β-galactosidase activity (-4,203 fluorescent units, P < 0.05), p16 protein expression (0.58-fold, P < 0.001), and mitochondrial content (0.18-fold, P < 0.001) in hypoxia suggests that transition from high (21%) to low (0.5%) O2 reduces senescence and promotes quiescence. Furthermore, physiological O2 levels increased migration (P < 0.05) compared with room air and hypoxia, and treatment with mesenchymal stem cell-conditioned media rescued CSC migration under hypoxia to levels comparable to physiological O2 migration (2-fold, P < 0.05 relative to CSC media control). Our finding that physiological O2 concentration is optimal for in vitro parameters of CSC biology suggests that standard room air may diminish cell regenerative potential. This study provides novel insights into the modulatory effects of O2 concentration on CSC biology and has important implications for refining stem cell therapies.
Collapse
Affiliation(s)
- Michael A Bellio
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Claudia O Rodrigues
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Ana Marie Landin
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | | | - Jeffim Kuznetsov
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Victoria Florea
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Krystalenia Valasaki
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Ivonne Hernandez Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; and
- Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
35
|
Saad A, Zhu XY, Herrmann S, Hickson L, Tang H, Dietz AB, van Wijnen AJ, Lerman L, Textor S. Adipose-derived mesenchymal stem cells from patients with atherosclerotic renovascular disease have increased DNA damage and reduced angiogenesis that can be modified by hypoxia. Stem Cell Res Ther 2016; 7:128. [PMID: 27612459 PMCID: PMC5016873 DOI: 10.1186/s13287-016-0389-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/27/2016] [Accepted: 08/23/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Adipose-derived MSC (AMSCs) possess angiogenic and immunomodulatory properties that may modulate kidney regeneration. Whether these properties are retained in older patients with atherosclerotic vascular disease is poorly understood. Hypoxic conditions are known to modify properties and growth characteristics of AMSCs. We tested the hypothesis that AMSCs from older patients with atherosclerotic renovascular disease (RVD) differ from normal kidney donors, and whether hypoxia changes their functional and molecular properties to promote angiogenesis. METHODS AMSCs from 11 patients with RVD (mean age =74.5 years) and 10 healthy kidney donors (mean age = 51.2 years) were cultured under normoxia (20 % O2) and hypoxia (1 % O2) for 3-4 days until they reached 80 % confluency. We analyzed expression of genes and microRNAs using RNA sequencing and real-time quantitative rt-PCR. Protein expression of selected angiogenic factors (VEGF, IGF, HGF and EGF) were quantified in conditioned media using ELISAs. Apoptosis was tested using Annexin IV staining. RESULTS Normoxic AMSC from RVD patients grew normally, but exhibited increased DNA damage and reduced migration. VEGF protein secretion was significantly lower in the RVD AMSCs (0.08 vs 2.4 ng/mL/ cell, p <0.05) while HGF was higher. Both trends were reversed during growth under hypoxic conditions. Hypoxia upregulated pro-angiogenic mRNAs expression in AMSCs (VEGF, FGF, STC and ANGPTL4), and downregulated expression of many miRNAs (e.g., miR-15a, miR-16, miR-93, miR-424, 126, 132, 221) except miR-210. CONCLUSIONS Thus, although AMSC from patients with RVD had increased DNA damage and reduced migration, hypoxia stimulated pro-angiogenic responses via increased expression of angiogenic genes, VEGF secretion and induction of the hypoxia-inducible miR-210, while downregulating angiogenesis-related miRNAs.
Collapse
Affiliation(s)
- Ahmed Saad
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester Minnesota, 200 First Street SW, Rochester, MN USA
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester Minnesota, 200 First Street SW, Rochester, MN USA
| | - Sandra Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester Minnesota, 200 First Street SW, Rochester, MN USA
| | - LaTonya Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester Minnesota, 200 First Street SW, Rochester, MN USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester Minnesota, 200 First Street SW, Rochester, MN USA
| | - Allan B. Dietz
- Division of Transfusion Medicine, Mayo Clinic, Rochester Minnesota, 200 First Street SW, Rochester, MN USA
| | - Andre J. van Wijnen
- Department of Orthopedic Surgery, Biochemistry and Molecular Biology, Mayo Clinic, Rochester Minnesota, 200 First Street SW, Rochester, MN USA
| | - Lilach Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester Minnesota, 200 First Street SW, Rochester, MN USA
| | - Stephen Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester Minnesota, 200 First Street SW, Rochester, MN USA
| |
Collapse
|
36
|
Nollet E, Hoymans VY, Van Craenenbroeck AH, Vrints CJ, Van Craenenbroeck EM. Improving stem cell therapy in cardiovascular diseases: the potential role of microRNA. Am J Physiol Heart Circ Physiol 2016; 311:H207-18. [PMID: 27208159 DOI: 10.1152/ajpheart.00239.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/11/2016] [Indexed: 11/22/2022]
Abstract
The initial promising prospect of autologous bone marrow-derived stem cell therapy in the setting of cardiovascular diseases has been overshadowed by functional shortcomings of the stem cell product. As powerful epigenetic regulators of (stem) cell function, microRNAs are valuable targets for novel therapeutic strategies. Indeed, modulation of specific miRNA expression could contribute to improved therapeutic efficacy of stem cell therapy. First, this review elaborates on the functional relevance of miRNA dysregulation in bone marrow-derived progenitor cells in different cardiovascular diseases. Next, we provide a comprehensive overview of the current evidence on the effect of specific miRNA modulation in several types of progenitor cells on cardiac and/or vascular regeneration. By elaborating on the cardioprotective regulation of progenitor cells on cardiac miRNAs, more insight in the underlying mechanisms of stem cell therapy is provided. Finally, some considerations are made regarding the potential of circulating miRNAs as regulators of the miRNA signature of progenitor cells in cardiovascular diseases.
Collapse
Affiliation(s)
- Evelien Nollet
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium
| | - Vicky Y Hoymans
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium
| | - Amaryllis H Van Craenenbroeck
- Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium; Department of Nephrology, Antwerp University Hospital, Antwerp, Belgium; Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium; and
| | - Christiaan J Vrints
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium; Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Emeline M Van Craenenbroeck
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium; Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
37
|
Han LH, Conrad B, Chung MT, Deveza L, Jiang X, Wang A, Butte MJ, Longaker MT, Wan D, Yang F. Winner of the Young Investigator Award of the Society for Biomaterials at the 10th World Biomaterials Congress, May 17-22, 2016, Montreal QC, Canada: Microribbon-based hydrogels accelerate stem cell-based bone regeneration in a mouse critical-size cranial defect model. J Biomed Mater Res A 2016; 104:1321-31. [PMID: 26991141 DOI: 10.1002/jbm.a.35715] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 02/23/2016] [Accepted: 03/09/2016] [Indexed: 12/31/2022]
Abstract
Stem cell-based therapies hold great promise for enhancing tissue regeneration. However, the majority of cells die shortly after transplantation, which greatly diminishes the efficacy of stem cell-based therapies. Poor cell engraftment and survival remain a major bottleneck to fully exploiting the power of stem cells for regenerative medicine. Biomaterials such as hydrogels can serve as artificial matrices to protect cells during delivery and guide desirable cell fates. However, conventional hydrogels often lack macroporosity, which restricts cell proliferation and delays matrix deposition. Here we report the use of injectable, macroporous microribbon (μRB) hydrogels as stem cell carriers for bone repair, which supports direct cell encapsulation into a macroporous scaffold with rapid spreading. When transplanted in a critical-sized, mouse cranial defect model, μRB-based hydrogels significantly enhanced the survival of transplanted adipose-derived stromal cells (ADSCs) (81%) and enabled up to three-fold cell proliferation after 7 days. In contrast, conventional hydrogels only led to 27% cell survival, which continued to decrease over time. MicroCT imaging showed μRBs enhanced and accelerated mineralized bone repair compared to hydrogels (61% vs. 34% by week 6), and stem cells were required for bone repair to occur. These results suggest that paracrine signaling of transplanted stem cells are responsible for the observed bone repair, and enhancing cell survival and proliferation using μRBs further promoted the paracrine-signaling effects of ADSCs for stimulating endogenous bone repair. We envision μRB-based scaffolds can be broadly useful as a novel scaffold for enhancing stem cell survival and regeneration of other tissue types. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1321-1331, 2016.
Collapse
Affiliation(s)
- Li-Hsin Han
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| | - Bogdan Conrad
- Program of Stem Cell Biology and Regenerative Medicine, Stanford University, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| | - Michael T Chung
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Dr, Hagey Building Room GK106, Stanford, California 94305
| | - Lorenzo Deveza
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| | - Xinyi Jiang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| | - Andrew Wang
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Dr, Grant Building Room H307A, Stanford, California 94305
| | - Manish J Butte
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Dr, Grant Building Room H307A, Stanford, California 94305
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 257 Campus Dr, Hagey Building Room GK106, Stanford, California 94305
| | - Derrick Wan
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Dr, Hagey Building Room GK106, Stanford, California 94305
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305.,Department of Bioengineering, Stanford University, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| |
Collapse
|
38
|
Tollemar V, Collier ZJ, Mohammed MK, Lee MJ, Ameer GA, Reid RR. Stem cells, growth factors and scaffolds in craniofacial regenerative medicine. Genes Dis 2016; 3:56-71. [PMID: 27239485 PMCID: PMC4880030 DOI: 10.1016/j.gendis.2015.09.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/22/2015] [Indexed: 02/08/2023] Open
Abstract
Current reconstructive approaches to large craniofacial skeletal defects are often complicated and challenging. Critical-sized defects are unable to heal via natural regenerative processes and require surgical intervention, traditionally involving autologous bone (mainly in the form of nonvascularized grafts) or alloplasts. Autologous bone grafts remain the gold standard of care in spite of the associated risk of donor site morbidity. Tissue engineering approaches represent a promising alternative that would serve to facilitate bone regeneration even in large craniofacial skeletal defects. This strategy has been tested in a myriad of iterations by utilizing a variety of osteoconductive scaffold materials, osteoblastic stem cells, as well as osteoinductive growth factors and small molecules. One of the major challenges facing tissue engineers is creating a scaffold fulfilling the properties necessary for controlled bone regeneration. These properties include osteoconduction, osetoinduction, biocompatibility, biodegradability, vascularization, and progenitor cell retention. This review will provide an overview of how optimization of the aforementioned scaffold parameters facilitates bone regenerative capabilities as well as a discussion of common osteoconductive scaffold materials.
Collapse
Affiliation(s)
- Viktor Tollemar
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL 60637, USA
| | - Zach J. Collier
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Maryam K. Mohammed
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guillermo A. Ameer
- Department of Surgery, Feinberg School of Medicine, Chicago, IL 60611, USA
- Biomedical Engineering Department, Northwestern University, Evanston, IL 60208, USA
| | - Russell R. Reid
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL 60637, USA
| |
Collapse
|
39
|
Walmsley GG, Senarath-Yapa K, Wearda TL, Menon S, Hu MS, Duscher D, Maan ZN, Tsai JM, Zielins ER, Weissman IL, Gurtner GC, Lorenz HP, Longaker MT. Surveillance of Stem Cell Fate and Function: A System for Assessing Cell Survival and Collagen Expression In Situ. Tissue Eng Part A 2015; 22:31-40. [PMID: 26486617 DOI: 10.1089/ten.tea.2015.0221] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell-based therapy is an emerging paradigm in skeletal regenerative medicine. However, the primary means by which transplanted cells contribute to bone repair and regeneration remain controversial. To gain an insight into the mechanisms of how both transplanted and endogenous cells mediate skeletal healing, we used a transgenic mouse strain expressing both the topaz variant of green fluorescent protein under the control of the collagen, type I, alpha 1 promoter/enhancer sequence (Col1a1(GFP)) and membrane-bound tomato red fluorescent protein constitutively in all cell types (R26(mTmG)). A comparison of healing in parietal versus frontal calvarial defects in these mice revealed that frontal osteoblasts express Col1a1 to a greater degree than parietal osteoblasts. Furthermore, the scaffold-based application of adipose-derived stromal cells (ASCs), bone marrow-derived mesenchymal stem cells (BM-MSCs), and osteoblasts derived from these mice to critical-sized calvarial defects allowed for investigation of cell survival and function following transplantation. We found that ASCs led to significantly faster rates of bone healing in comparison to BM-MSCs and osteoblasts. ASCs displayed both increased survival and increased Col1a1 expression compared to BM-MSCs and osteoblasts following calvarial defect transplantation, which may explain their superior regenerative capacity in the context of bone healing. Using this novel reporter system, we were able to elucidate how cell-based therapies impact bone healing and identify ASCs as an attractive candidate for cell-based skeletal regenerative therapy. These insights potentially influence stem cell selection in translational clinical trials evaluating cell-based therapeutics for osseous repair and regeneration.
Collapse
Affiliation(s)
- Graham G Walmsley
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California.,2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Kshemendra Senarath-Yapa
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Taylor L Wearda
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Siddharth Menon
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Michael S Hu
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California.,2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Dominik Duscher
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Zeshaan N Maan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Jonathan M Tsai
- 2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Elizabeth R Zielins
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Irving L Weissman
- 2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Geoffrey C Gurtner
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - H Peter Lorenz
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California.,2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
40
|
Ryu S, Yoo J, Jang Y, Han J, Yu SJ, Park J, Jung SY, Ahn KH, Im SG, Char K, Kim BS. Nanothin Coculture Membranes with Tunable Pore Architecture and Thermoresponsive Functionality for Transfer-Printable Stem Cell-Derived Cardiac Sheets. ACS NANO 2015; 9:10186-202. [PMID: 26361723 DOI: 10.1021/acsnano.5b03823] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Coculturing stem cells with the desired cell type is an effective method to promote the differentiation of stem cells. The features of the membrane used for coculturing are crucial to achieving the best outcome. Not only should the membrane act as a physical barrier that prevents the mixing of the cocultured cell populations, but it should also allow effective interactions between the cells. Unfortunately, conventional membranes used for coculture do not sufficiently meet these requirements. In addition, cell harvesting using proteolytic enzymes following coculture impairs cell viability and the extracellular matrix (ECM) produced by the cultured cells. To overcome these limitations, we developed nanothin and highly porous (NTHP) membranes, which are ∼20-fold thinner and ∼25-fold more porous than the conventional coculture membranes. The tunable pore size of NTHP membranes at the nanoscale level was found crucial for the formation of direct gap junctions-mediated contacts between the cocultured cells. Differentiation of the cocultured stem cells was dramatically enhanced with the pore size-customized NTHP membrane system compared to conventional coculture methods. This was likely due to effective physical contacts between the cocultured cells and the fast diffusion of bioactive molecules across the membrane. Also, the thermoresponsive functionality of the NTHP membranes enabled the efficient generation of homogeneous, ECM-preserved, highly viable, and transfer-printable sheets of cardiomyogenically differentiated cells. The coculture platform developed in this study would be effective for producing various types of therapeutic multilayered cell sheets that can be differentiated from stem cells.
Collapse
Affiliation(s)
| | | | | | | | - Seung Jung Yu
- Department of Chemical and Biomolecular Engineering & KI for Nano Century, Korea Advanced Institute of Science and Technology , Daejeon 305-701, Republic of Korea
| | | | | | | | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering & KI for Nano Century, Korea Advanced Institute of Science and Technology , Daejeon 305-701, Republic of Korea
| | | | | |
Collapse
|
41
|
Padda J, Sequiera GL, Sareen N, Dhingra S. Stem cell therapy for cardiac regeneration: hits and misses. Can J Physiol Pharmacol 2015; 93:835-41. [DOI: 10.1139/cjpp-2014-0468] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiac injury and loss of cardiomyocytes is a causative as well as a resultant condition of cardiovascular disorders, which are the leading cause of death throughout the world. This loss of cardiomyocytes cannot be completely addressed through the currently available drugs being administered, which mainly function only in relieving the symptoms. There is a huge potential being investigated for regenerative and cell replacement therapies through recruiting stem cells of various origins namely embryonic, reprogramming/induction, and adult tissue. These sources are being actively studied for translation to clinical scenarios. In this review, we attempt to discuss some of these promising scenarios, including the clinical trials and the obstacles that need to be overcome, and hope to address the direction in which stem cell therapy is heading.
Collapse
Affiliation(s)
- Jagjit Padda
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Regenerative Medicine Program, College of Medicine, Faculty of Health Sciences, University of Manitoba, R 3028-2, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Regenerative Medicine Program, College of Medicine, Faculty of Health Sciences, University of Manitoba, R 3028-2, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Glen Lester Sequiera
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Regenerative Medicine Program, College of Medicine, Faculty of Health Sciences, University of Manitoba, R 3028-2, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Regenerative Medicine Program, College of Medicine, Faculty of Health Sciences, University of Manitoba, R 3028-2, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Niketa Sareen
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Regenerative Medicine Program, College of Medicine, Faculty of Health Sciences, University of Manitoba, R 3028-2, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Regenerative Medicine Program, College of Medicine, Faculty of Health Sciences, University of Manitoba, R 3028-2, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Sanjiv Dhingra
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Regenerative Medicine Program, College of Medicine, Faculty of Health Sciences, University of Manitoba, R 3028-2, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Regenerative Medicine Program, College of Medicine, Faculty of Health Sciences, University of Manitoba, R 3028-2, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
42
|
Extracellular Matrix can Recover the Downregulation of Adhesion Molecules after Cell Detachment and Enhance Endothelial Cell Engraftment. Sci Rep 2015; 5:10902. [PMID: 26039874 PMCID: PMC4454140 DOI: 10.1038/srep10902] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 05/05/2015] [Indexed: 12/13/2022] Open
Abstract
The low cell engraftment after transplantation limits the successful application of stem cell therapy and the exact pathway leading to acute donor cell death following transplantation is still unknown. Here we investigated if processes involved in cell preparation could initiate downregulation of adhesion-related survival signals, and further affect cell engraftment after transplantation. Human embryonic stem cell-derived endothelial cells (hESC-ECs) were suspended in PBS or Matrigel and kept at 4 °C. Quantitative RT-PCR analysis was used to test the adhesion and apoptosis genes’ expression of hESC-ECs. We demonstrated that cell detachment can cause downregulation of cell adhesion and extracellular matrix (ECM) molecules, but no obvious cell anoikis, a form of apoptosis after cell detachment, was observed. The downregulation of adhesion and ECM molecules could be regained in the presence of Matrigel. Finally, we transplanted hESC-ECs into a mouse myocardial ischemia model. When transplanted with Matrigel, the long-term engraftment of hESC-ECs was increased through promoting angiogenesis and inhibiting apoptosis, and this was confirmed by bioluminescence imaging. In conclusion, ECM could rescue the functional genes expression after cell detached from culture dish, and this finding highlights the importance of increasing stem cell engraftment by mimicking stem cell niches through ECM application.
Collapse
|
43
|
Ikuta Y, Kamei N, Ishikawa M, Adachi N, Ochi M. In Vivo Kinetics of Mesenchymal Stem Cells Transplanted into the Knee Joint in a Rat Model Using a Novel Magnetic Method of Localization. Clin Transl Sci 2015; 8:467-74. [PMID: 25963065 DOI: 10.1111/cts.12284] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
We have developed a magnetic system for targeting cells in minimally invasive cell transplantation. Magnetically labeled MSCs (m-MSCs) with nanoscale iron particles can be guided into the desired region by magnetic force from an extracorporeal device. We reported that magnetic targeting of m-MSCs enhances cartilage repair in a mini-pig model. However, the detailed kinetics of these magnetically targeted m-MSCs remain unknown. For clinical use, this aspect should be clarified from a safety standpoint. We therefore investigated the spatial and temporal distribution of the fluorescently-labeled m-MSCs transplanted into the knee joint using in vivo fluorescence combined with three-dimensional computed tomographic imaging in a rat model. Although the intraarticularly injected m-MSCs were spread throughout the joint cavity in the absence of magnetic force, the magnetic force caused the injected m-MSCs to accumulate around the chondral lesion. Further examinations including ex vivo imaging, histological assessments and reverse transcription polymerase chain reaction revealed that transplanted MSCs were not present in any major organs after intraarticular administration, regardless of magnetic targeting. Our data suggest that m-MSCs can be accumulated efficiently into a chondral lesion using our magnetic targeting system, while none of the intraarticularly transplanted MSCs migrate to other major organs.
Collapse
Affiliation(s)
- Yasunari Ikuta
- Department of Orthopedic Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Naosuke Kamei
- Department of Orthopedic Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Masakazu Ishikawa
- Department of Orthopedic Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Nobuo Adachi
- Department of Orthopedic Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Mitsuo Ochi
- Department of Orthopedic Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
44
|
Li KC, Hu YC. Cartilage tissue engineering: recent advances and perspectives from gene regulation/therapy. Adv Healthc Mater 2015; 4:948-68. [PMID: 25656682 DOI: 10.1002/adhm.201400773] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/10/2015] [Indexed: 12/16/2022]
Abstract
Diseases in articular cartilages affect millions of people. Despite the relatively simple biochemical and cellular composition of articular cartilages, the self-repair ability of cartilage is limited. Successful cartilage tissue engineering requires intricately coordinated interactions between matrerials, cells, biological factors, and phycial/mechanical factors, and still faces a multitude of challenges. This article presents an overview of the cartilage biology, current treatments, recent advances in the materials, biological factors, and cells used in cartilage tissue engineering/regeneration, with strong emphasis on the perspectives of gene regulation (e.g., microRNA) and gene therapy.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| | - Yu-Chen Hu
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| |
Collapse
|
45
|
Blázquez R, Sánchez-Margallo FM, Crisóstomo V, Báez C, Maestre J, García-Lindo M, Usón A, Álvarez V, Casado JG. Intrapericardial administration of mesenchymal stem cells in a large animal model: a bio-distribution analysis. PLoS One 2015; 10:e0122377. [PMID: 25816232 PMCID: PMC4376786 DOI: 10.1371/journal.pone.0122377] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 02/15/2015] [Indexed: 12/18/2022] Open
Abstract
The appropriate administration route for cardiovascular cell therapy is essential to ensure the viability, proliferative potential, homing capacity and implantation of transferred cells. At the present, the intrapericardial administration of pharmacological agents is considered an efficient method for the treatment of cardiovascular diseases. However, only a few reports have addressed the question whether the intrapericardial delivery of Mesenchymal Stem Cells (MSCs) could be an optimal administration route. This work firstly aimed to analyze the pericardial fluid as a cell-delivery vehicle. Moreover, the in vivo biodistribution pattern of intrapericardially administered MSCs was evaluated in a clinically relevant large animal model. Our in vitro results firstly showed that, MSCs viability, proliferative behavior and phenotypic profile were unaffected by exposure to pericardial fluid. Secondly, in vivo cell tracking by magnetic resonance imaging, histological examination and Y-chromosome amplification clearly demonstrated the presence of MSCs in pericardium, ventricles (left and right) and atrium (left and right) when MSCs were administered into the pericardial space. In conclusion, here we demonstrate that pericardial fluid is a suitable vehicle for MSCs and intrapericardial route provides an optimal retention and implantation of MSCs.
Collapse
Affiliation(s)
- Rebeca Blázquez
- Stem Cell Therapy Unit, Minimally Invasive Surgery Centre, Caceres, Spain
| | | | - Verónica Crisóstomo
- Endoluminal Therapy and Diagnosis, Minimally Invasive Surgery Centre, Caceres, Spain
| | - Claudia Báez
- Endoluminal Therapy and Diagnosis, Minimally Invasive Surgery Centre, Caceres, Spain
| | - Juan Maestre
- Endoluminal Therapy and Diagnosis, Minimally Invasive Surgery Centre, Caceres, Spain
| | | | - Alejandra Usón
- Stem Cell Therapy Unit, Minimally Invasive Surgery Centre, Caceres, Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, Minimally Invasive Surgery Centre, Caceres, Spain
| | - Javier G. Casado
- Stem Cell Therapy Unit, Minimally Invasive Surgery Centre, Caceres, Spain
- * E-mail:
| |
Collapse
|
46
|
Nejadnik H, Ye D, Lenkov OD, Donig J, Martin JE, Castillo R, Derugin N, Sennino B, Rao J, Daldrup-Link HE. Magnetic resonance imaging of stem cell apoptosis in arthritic joints with a caspase activatable contrast agent. ACS NANO 2015; 9:1150-60. [PMID: 25597243 PMCID: PMC4441518 DOI: 10.1021/nn504494c] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
About 43 million individuals in the U.S. encounter cartilage injuries due to trauma or osteoarthritis, leading to joint pain and functional disability. Matrix-associated stem cell implants (MASI) represent a promising approach for repair of cartilage defects. However, limited survival of MASI creates a significant bottleneck for successful cartilage regeneration outcomes and functional reconstitution. We report an approach for noninvasive detection of stem cell apoptosis with magnetic resonance imaging (MRI), based on a caspase-3-sensitive nanoaggregation MRI probe (C-SNAM). C-SNAM self-assembles into nanoparticles after hydrolysis by caspase-3, leading to 90% amplification of (1)H MR signal and prolonged in vivo retention. Following intra-articular injection, C-SNAM causes significant MR signal enhancement in apoptotic MASI compared to viable MASI. Our results indicate that C-SNAM functions as an imaging probe for stem cell apoptosis in MASI. This concept could be applied to a broad range of cell transplants and target sites.
Collapse
Affiliation(s)
- Hossein Nejadnik
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA
| | - Deju Ye
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA
| | - Olga D. Lenkov
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA
| | - Jessica Donig
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA
| | - John E. Martin
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA
| | - Rostislav Castillo
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA
| | - Nikita Derugin
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA
| | - Barbara Sennino
- Comprehensive Cancer Center, Cardiovascular Research Institute and Department of Anatomy, University of California San Francisco, San Francisco, CA
| | - Jianghong Rao
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA
| | - Heike E. Daldrup-Link
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA
| |
Collapse
|
47
|
Tang YH, Ma YY, Zhang ZJ, Wang YT, Yang GY. Opportunities and challenges: stem cell-based therapy for the treatment of ischemic stroke. CNS Neurosci Ther 2015; 21:337-47. [PMID: 25676164 DOI: 10.1111/cns.12386] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 01/09/2015] [Accepted: 01/09/2015] [Indexed: 01/01/2023] Open
Abstract
Stem cell-based therapy for ischemic stroke has been widely explored in animal models and provides strong evidence of benefits. In this review, we summarize the types of stem cells, various delivery routes, and tracking tools for stem cell therapy of ischemic stroke. MSCs, EPCs, and NSCs are the most explored cell types for ischemic stroke treatment. Although the mechanisms of stem cell-based therapies are not fully understood, the most possible functions of the transplanted cells are releasing growth factors and regulating microenvironment through paracrine mechanism. Clinical application of stem cell-based therapy is still in its infancy. The next decade of stem cell research in stroke field needs to focus on combining different stem cells and different imaging modalities to fully explore the potential of this therapeutic avenue: from bench to bedside and vice versa.
Collapse
Affiliation(s)
- Yao-Hui Tang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | |
Collapse
|
48
|
Wang Y, Li C, Cheng K, Zhang R, Narsinh K, Li S, Li X, Qin X, Zhang R, Li C, Su T, Chen J, Cao F. Activation of liver X receptor improves viability of adipose-derived mesenchymal stem cells to attenuate myocardial ischemia injury through TLR4/NF-κB and Keap-1/Nrf-2 signaling pathways. Antioxid Redox Signal 2014; 21:2543-57. [PMID: 24915051 PMCID: PMC4245883 DOI: 10.1089/ars.2013.5683] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIMS Clinical application of cellular therapy for cardiac regeneration is significantly hampered by the low retention of engrafted cells, mainly attributable to the poor microenvironment dominated by inflammation and oxidative stress in the host's infarcted myocardium. This study aims at investigating whether liver X receptor (LXR) agonist T0901317 will improve survival of adipose-derived mesenchymal stem cells (AD-MSCs) after transplantation into infarcted hearts. RESULTS Noninvasive in vivo bioluminescence imaging and histological staining showed that LXR agonist T0901317 improved the retention and survival of intramyocardially injected AD-MSCs. Moreover, combined therapy of LXR agonist and AD-MSCs inhibited host cardiomyocyte apoptosis, reduced fibrosis, and improved cardiac function, while it concomitantly decreased inflammatory cytokines (e.g., tumor necrosis factor-α and interleukin-6) and increased growth factor (e.g., vascular endothelial growth factor and basic fibroblast growth factor) expression in infarct myocardium. To reveal possible mechanisms, AD-MSCs were subjected to hypoxia/serum deprivation (H/SD) injury to simulate ischemic conditions in vivo. The LXR agonist (10(-7) mM) improved AD-MSC survival under H/SD condition. Western blot revealed that the LXR agonist reduced TLR4, TRAF-6, and MyD88 protein expression, inhibited IκBα phosphorylation and NF-κB-p65 nuclear translocation, which resulted in accelerated Keap-1 protein degradation, enhanced Nrf-2 nuclear translocation, and increased HO-1 protein expression. INNOVATION AND CONCLUSION LXR agonist can enhance the functional survival of transplanted AD-MSCs in infarcted myocardium, at least partially, via modulation of the TLR4/NF-κB and Keap-1/Nrf-2 signaling pathways. Moreover, combined therapy of LXR agonist and AD-MSCs has a synergetic effect on cardiac repair and functional improvement after infarction.
Collapse
Affiliation(s)
- Yabin Wang
- 1 Department of Cardiology, PLA General Hospital , Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Tevlin R, McArdle A, Atashroo D, Walmsley GG, Senarath-Yapa K, Zielins ER, Paik KJ, Longaker MT, Wan DC. Biomaterials for craniofacial bone engineering. J Dent Res 2014; 93:1187-95. [PMID: 25139365 DOI: 10.1177/0022034514547271] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Conditions such as congenital anomalies, cancers, and trauma can all result in devastating deficits of bone in the craniofacial skeleton. This can lead to significant alteration in function and appearance that may have significant implications for patients. In addition, large bone defects in this area can pose serious clinical dilemmas, which prove difficult to remedy, even with current gold standard surgical treatments. The craniofacial skeleton is complex and serves important functional demands. The necessity to develop new approaches for craniofacial reconstruction arises from the fact that traditional therapeutic modalities, such as autologous bone grafting, present myriad limitations and carry with them the potential for significant complications. While the optimal bone construct for tissue regeneration remains to be elucidated, much progress has been made in the past decade. Advances in tissue engineering have led to innovative scaffold design, complemented by progress in the understanding of stem cell-based therapy and growth factor enhancement of the healing cascade. This review focuses on the role of biomaterials for craniofacial bone engineering, highlighting key advances in scaffold design and development.
Collapse
Affiliation(s)
- R Tevlin
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - A McArdle
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - D Atashroo
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - G G Walmsley
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - K Senarath-Yapa
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - E R Zielins
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - K J Paik
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - M T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, California, USA Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - D C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
50
|
Ceccaldi C, Bushkalova R, Alfarano C, Lairez O, Calise D, Bourin P, Frugier C, Rouzaud-Laborde C, Cussac D, Parini A, Sallerin B, Fullana SG. Evaluation of polyelectrolyte complex-based scaffolds for mesenchymal stem cell therapy in cardiac ischemia treatment. Acta Biomater 2014; 10:901-11. [PMID: 24211733 DOI: 10.1016/j.actbio.2013.10.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/20/2013] [Accepted: 10/25/2013] [Indexed: 11/15/2022]
Abstract
Three-dimensional (3D) scaffolds hold great potential for stem cell-based therapies. Indeed, recent results have shown that biomimetic scaffolds may enhance cell survival and promote an increase in the concentration of therapeutic cells at the injury site. The aim of this work was to engineer an original polymeric scaffold based on the respective beneficial effects of alginate and chitosan. Formulations were made from various alginate/chitosan ratios to form opposite-charge polyelectrolyte complexes (PECs). After freeze-drying, the resultant matrices presented a highly interconnected porous microstructure and mechanical properties suitable for cell culture. In vitro evaluation demonstrated their compatibility with mesenchymal stell cell (MSC) proliferation and their ability to maintain paracrine activity. Finally, the in vivo performance of seeded 3D PEC scaffolds with a polymeric ratio of 40/60 was evaluated after an acute myocardial infarction provoked in a rat model. Evaluation of cardiac function showed a significant increase in the ejection fraction, improved neovascularization, attenuated fibrosis as well as less left ventricular dilatation as compared to an animal control group. These results provide evidence that 3D PEC scaffolds prepared from alginate and chitosan offer an efficient environment for 3D culturing of MSCs and represent an innovative solution for tissue engineering.
Collapse
Affiliation(s)
- Caroline Ceccaldi
- Université de Toulouse, CIRIMAT, UPS-INPT-CNRS, Faculté de Pharmacie, F-31062 Toulouse, France; INSERM, UMR 1048, F-31432 Toulouse, France.
| | - Raya Bushkalova
- Université de Toulouse, CIRIMAT, UPS-INPT-CNRS, Faculté de Pharmacie, F-31062 Toulouse, France; INSERM, UMR 1048, F-31432 Toulouse, France
| | | | | | | | - Philippe Bourin
- EFS, Laboratoire de thérapie cellulaire, F-31027 Toulouse, France
| | | | - Charlotte Rouzaud-Laborde
- INSERM, UMR 1048, F-31432 Toulouse, France; CHU Toulouse, Service de Pharmacie Hospitalière, F-31432 Toulouse, France
| | - Daniel Cussac
- INSERM, UMR 1048, F-31432 Toulouse, France; Université de Toulouse, UPS, Faculté des Sciences Pharmaceutiques, F-31062 Toulouse, France
| | - Angelo Parini
- INSERM, UMR 1048, F-31432 Toulouse, France; Université de Toulouse, UPS, Faculté des Sciences Pharmaceutiques, F-31062 Toulouse, France; CHU Toulouse, Service de Pharmacie Hospitalière, F-31432 Toulouse, France
| | - Brigitte Sallerin
- INSERM, UMR 1048, F-31432 Toulouse, France; Université de Toulouse, UPS, Faculté des Sciences Pharmaceutiques, F-31062 Toulouse, France; CHU Toulouse, Service de Pharmacie Hospitalière, F-31432 Toulouse, France
| | - Sophie Girod Fullana
- Université de Toulouse, CIRIMAT, UPS-INPT-CNRS, Faculté de Pharmacie, F-31062 Toulouse, France
| |
Collapse
|