1
|
Cong M, Hu JJ, Yu Y, Li XL, Sun XT, Wang LT, Wu X, Zhu LJ, Yang XJ, He QR, Ding F, Shi HY. miRNA-21-5p is an important contributor to the promotion of injured peripheral nerve regeneration using hypoxia-pretreated bone marrow-derived neural crest cells. Neural Regen Res 2025; 20:277-290. [PMID: 38767492 PMCID: PMC11246143 DOI: 10.4103/1673-5374.390956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/06/2023] [Accepted: 09/26/2023] [Indexed: 05/22/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202501000-00035/figure1/v/2024-05-14T021156Z/r/image-tiff Our previous study found that rat bone marrow-derived neural crest cells (acting as Schwann cell progenitors) have the potential to promote long-distance nerve repair. Cell-based therapy can enhance peripheral nerve repair and regeneration through paracrine bioactive factors and intercellular communication. Nevertheless, the complex contributions of various types of soluble cytokines and extracellular vesicle cargos to the secretome remain unclear. To investigate the role of the secretome and extracellular vesicles in repairing damaged peripheral nerves, we collected conditioned culture medium from hypoxia-pretreated neural crest cells, and found that it significantly promoted the repair of sensory neurons damaged by oxygen-glucose deprivation. The mRNA expression of trophic factors was highly expressed in hypoxia-pretreated neural crest cells. We performed RNA sequencing and bioinformatics analysis and found that miR-21-5p was enriched in hypoxia-pretreated extracellular vesicles of neural crest cells. Subsequently, to further clarify the role of hypoxia-pretreated neural crest cell extracellular vesicles rich in miR-21-5p in axonal growth and regeneration of sensory neurons, we used a microfluidic axonal dissociation model of sensory neurons in vitro, and found that hypoxia-pretreated neural crest cell extracellular vesicles promoted axonal growth and regeneration of sensory neurons, which was greatly dependent on loaded miR-21-5p. Finally, we constructed a miR-21-5p-loaded neural conduit to repair the sciatic nerve defect in rats and found that the motor and sensory functions of injured rat hind limb, as well as muscle tissue morphology of the hind limbs, were obviously restored. These findings suggest that hypoxia-pretreated neural crest extracellular vesicles are natural nanoparticles rich in miRNA-21-5p. miRNA-21-5p is one of the main contributors to promoting nerve regeneration by the neural crest cell secretome. This helps to explain the mechanism of action of the secretome and extracellular vesicles of neural crest cells in repairing damaged peripheral nerves, and also promotes the application of miR-21-5p in tissue engineering regeneration medicine.
Collapse
Affiliation(s)
- Meng Cong
- School of Medicine, Nantong University, Nantong, Jiangsu Province, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Jing-Jing Hu
- School of Medicine, Nantong University, Nantong, Jiangsu Province, China
- Department of Physiology, Jiangsu Health Vocational College, Nanjing, Jiangsu Province, China
| | - Yan Yu
- School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Li Li
- School of Medicine, Nantong University, Nantong, Jiangsu Province, China
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Ting Sun
- School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Li-Ting Wang
- School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Xia Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Ling-Jie Zhu
- School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Jia Yang
- School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Qian-Ru He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Hai-Yan Shi
- School of Medicine, Nantong University, Nantong, Jiangsu Province, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
2
|
Zou X, Brigstock D. Extracellular Vesicles from Mesenchymal Stem Cells: Potential as Therapeutics in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Biomedicines 2024; 12:2848. [PMCID: PMC11673942 DOI: 10.3390/biomedicines12122848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/03/2025] Open
Abstract
Background/Objectives: Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by the accumulation of triglycerides within hepatocytes, which can progress to more severe conditions, such as metabolic dysfunction-associated steatohepatitis (MASH), which may include progressive fibrosis, leading to cirrhosis, cancer, and death. This goal of this review is to highlight recent research showing the potential of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) in reducing the key pathogenic pathways of MASLD or MASH. Methods: Relevant published studies were identified using PubMed with one or more of the following search terms: MASLD, MASH, NAFLD, NASH, exosome, extracellular vesicle (EV), therapy, and/or mesenchymal stem cells (MSC). The primary literature were subsequently downloaded and summarized. Results: Using in vitro or in vivo models, MSC-EVs have been found to counteract oxidative stress, a significant contributor to liver injury in MASH, and to suppress disease progression, including steatosis, inflammation, and, in a few instances, fibrosis. Some of these outcomes have been attributed to specific EV cargo components including microRNAs and proteins. Thus, MSC-EVs enriched with these types of molecules may have improved the therapeutic efficacy for MASLD/MASH and represent a novel approach to potentially halt or reverse the disease process. Conclusions: MSC-EVs are attractive therapeutic agents for treating MASLD/MASH. Further studies are necessary to validate the clinical applicability and efficacy of MSC-EVs in human MASH patients, focusing on optimizing delivery strategies and identifying the pathogenic pathways that are targeted by specific EV components.
Collapse
Affiliation(s)
- Xue Zou
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - David Brigstock
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43212, USA
| |
Collapse
|
3
|
Lee HK, Kim HK, Kim JY, Kim JS, Park J, Kim MS, Lee TY, Lim KH, Park H, Son DJ, Hong JT, Han SB. Ingenol-3-Angelate Enhances the B Cell Inhibitory Potential of Mesenchymal Stem Cells, Leading to Marked Alleviation of Lupus Symptoms in MRL. faslpr Mice. Int J Mol Sci 2024; 25:12625. [PMID: 39684336 DOI: 10.3390/ijms252312625] [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: 10/08/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by autoantibody production by hyper-activated B cells. Although mesenchymal stem cells (MSCs) relieve lupus symptoms by inhibiting mainly T cells, whether MSCs also inhibit B cells has been controversial. Here, we found that naïve MSCs inhibited IFN-γ production by T cells, but not IgM production by B cells. We used a chemical approach to prime MSCs to inhibit B cells. We found that ingenol-3-angelate (I3A), a non-tumor-promoting phorbol ester, activated MSCs to inhibit B cells in a TGF-β1-dependent manner. We also showed that IL-1β induced MSCs to continuously secrete TGF-β1, which directly inhibited IgM production by B cells, whereas IL-1β did not. I3A-treated MSCs were better than naïve MSCs at ameliorating SLE symptoms in MRL.faslpr mice. In summary, our data provide information on how to generate MSCs that are effective for the treatment of SLE characterized by excessive B cell activation.
Collapse
Affiliation(s)
- Hong Kyung Lee
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
- Bioengineering Institute, CorestemChemon Inc., Gyeonggi 13486, Republic of Korea
| | - Hwa Kyung Kim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Ji Yeon Kim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Ji Su Kim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - JinKyung Park
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
- Bioengineering Institute, CorestemChemon Inc., Gyeonggi 13486, Republic of Korea
| | - Min Sung Kim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
- Bioengineering Institute, CorestemChemon Inc., Gyeonggi 13486, Republic of Korea
| | - Tae Yong Lee
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
- Bioengineering Institute, CorestemChemon Inc., Gyeonggi 13486, Republic of Korea
| | - Key-Hwan Lim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Hanseul Park
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| |
Collapse
|
4
|
Testa AM, Vignozzi L, Corallo D, Aveic S, Viola A, Allegra M, Angioni R. Hypoxic Human Microglia Promote Angiogenesis Through Extracellular Vesicle Release. Int J Mol Sci 2024; 25:12508. [PMID: 39684220 DOI: 10.3390/ijms252312508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Microglia, the brain-resident immune cells, orchestrate neuroinflammatory responses and are crucial in the progression of neurological diseases, including ischemic stroke (IS), which accounts for approximately 85% of all strokes worldwide. Initially deemed detrimental, microglial activation has been shown to perform protective functions in the ischemic brain. Besides their effects on neurons, microglia play a role in promoting post-ischemic angiogenesis, a pivotal step for restoring oxygen and nutrient supply. However, the molecular mechanisms underlying microglia-endothelial cell interactions remain largely unresolved, particularly in humans. Using both in vitro and in vivo models, we investigated the angiogenic signature and properties of extracellular vesicles (EVs) released by human microglia upon hypoxia-reperfusion stimulation. EVs were isolated and characterized in terms of their size, concentration, and protein content. Their angiogenic potential was evaluated using endothelial cell assays and a zebrafish xenograft model. The in vivo effects were further assessed in a mouse model of ischemic stroke. Our findings identified key proteins orchestrating the pro-angiogenic functions of human microglial EVs under hypoxic conditions. In vitro assays demonstrated that hypoxic EVs (hypEVs) promoted endothelial cell migration and tube formation. In vivo, hypEVs induced vessel sprouting in zebrafish and increased microvessel density in the perilesional area of mice following ischemic stroke.
Collapse
Affiliation(s)
- Alessandra Maria Testa
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Laboratory of Immunity, Inflammation and Angiogenesis, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| | - Livia Vignozzi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Manuela Allegra
- Laboratory of Neuronal Circuits in Developmental Disorders, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
- Neuroscience Institute, National Research Council, 35131 Padua, Italy
| | - Roberta Angioni
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Laboratory of Immunity, Inflammation and Angiogenesis, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| |
Collapse
|
5
|
Liu C, Li Q, Ma JX, Lu B, Criswell T, Zhang Y. Exosome-mediated renal protection: Halting the progression of fibrosis. Genes Dis 2024; 11:101117. [PMID: 39263535 PMCID: PMC11388648 DOI: 10.1016/j.gendis.2023.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 09/13/2024] Open
Abstract
Renal fibrosis is a complex and multifactorial process that involves inflammation, cell proliferation, collagen, and fibronectin deposition in the kidney, ultimately leading to chronic kidney disease and even end-stage renal disease. The main goal of treatment is to slow down or halt the progression of fibrosis and to improve or preserve kidney function. Despite significant progress made in understanding the underlying mechanisms of renal fibrosis, current therapies have limited renal protection as the disease progresses. Exosomes derived from stem cells are a newer area of research for the treatment of renal fibrosis. Exosomes as nano-sized extracellular vesicles carry proteins, lipids, and nucleic acids, which can be taken up by local or distant cells, serving as mediators of intercellular communication and as drug delivery vehicles. Exosomes deliver molecules that reduce inflammation, renal fibrosis and extracellular matrix protein production, and promote tissue regeneration in animal models of kidney disease. Additionally, they have several advantages over stem cells, such as being non-immunogenic, having low risk of tumor formation, and being easier to produce and store. This review describes the use of natural and engineered exosomes containing therapeutic agents capable of mediating anti-inflammatory and anti-fibrotic processes during both acute kidney injury and chronic kidney disease. Exosome-based therapies will be compared with stem cell-based treatments for tissue regeneration, with a focus on renal protection. Finally, future directions and strategies for improving the therapeutic efficacy of exosomes are discussed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Baisong Lu
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Tracy Criswell
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
6
|
Tian H, Tian F, Ma D, Xiao B, Ding Z, Zhai X, Song L, Ma C. Priming and Combined Strategies for the Application of Mesenchymal Stem Cells in Ischemic Stroke: A Promising Approach. Mol Neurobiol 2024; 61:7127-7150. [PMID: 38366307 DOI: 10.1007/s12035-024-04012-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/31/2024] [Indexed: 02/18/2024]
Abstract
Ischemic stroke (IS) is a leading cause of death and disability worldwide. Tissue plasminogen activator (tPA) administration and mechanical thrombectomy are the main treatments but have a narrow time window. Mesenchymal stem cells (MSCs), which are easily scalable in vitro and lack ethical concerns, possess the potential to differentiate into various types of cells and secrete a great number of growth factors for neuroprotection and regeneration. Moreover, MSCs have low immunogenicity and tumorigenic properties, showing safety and preliminary efficacy both in preclinical studies and clinical trials of IS. However, it is unlikely that MSC treatment alone will be sufficient to maximize recovery due to the low survival rate of transplanted cells and various mechanisms of ischemic brain damage in the different stages of IS. Preconditioning was used to facilitate the homing, survival, and secretion ability of the grafted MSCs in the ischemic region, while combination therapies are alternatives that can maximize the treatment effects, focusing on multiple therapeutic targets to promote stroke recovery. In this case, the combination therapy can yield a synergistic effect. In this review, we summarize the type of MSCs, preconditioning methods, and combined strategies as well as their therapeutic mechanism in the treatment of IS to accelerate the transformation from basic research to clinical application.
Collapse
Affiliation(s)
- Hao Tian
- Experimental Management Center, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, No. 121, University Street, Higher Education Park, Jinzhong, 030619, China
| | - Feng Tian
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
| | - Dong Ma
- Department of Neurosurgery, The Key Laboratory of Prevention and Treatment of Neurological Disease of Shanxi Provincial Health Commission, Sinopharm Tongmei General Hospital, Datong, 037003, China
| | - Baoguo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Zhibin Ding
- Department of Neurology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030000, China
| | - Xiaoyan Zhai
- Experimental Management Center, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, No. 121, University Street, Higher Education Park, Jinzhong, 030619, China
- School of Basic Medicine of Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Lijuan Song
- Experimental Management Center, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, No. 121, University Street, Higher Education Park, Jinzhong, 030619, China.
| | - Cungen Ma
- Experimental Management Center, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, No. 121, University Street, Higher Education Park, Jinzhong, 030619, China.
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China.
| |
Collapse
|
7
|
Xue Z, Hu D, Tang H, Xue M, Zhu Y, Li Y, Liao Y. Mechanical force regulates the paracrine functions of ADSCs to assist skin expansion in rats. Stem Cell Res Ther 2024; 15:250. [PMID: 39135129 PMCID: PMC11321134 DOI: 10.1186/s13287-024-03822-0] [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: 04/11/2024] [Accepted: 06/30/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND In the repair of massive tissue defects using expanded large skin flaps, the incidence of complications increases with the size of the expanded area. Currently, stem cell therapy has limitations to solve this problem. We hypothesized that conditioned medium of adipose-derived stem cells (ADSC-CM) collected following mechanical pretreatment can assist skin expansion. METHODS Rat aortic endothelial cells and fibroblasts were cultured with ADSC-CM collected under 0%, 10%, 12%, and 15% stretching force. Ten-milliliter cylindrical soft tissue expanders were subcutaneously implanted into the backs of 36 Sprague-Dawley rats. The 0% and 10% stretch groups were injected with ADSC-CM collected under 0% and 10% stretching force, respectively, while the control group was not injected. After 3, 7, 14, and 30 days of expansion, expanded skin tissue was harvested for staining and qPCR analyses. RESULTS Endothelial cells had the best lumen formation and highest migration rate, and fibroblasts secreted the most collagen upon culture with ADSC-CM collected under 10% stretching force. The skin expansion rate was significantly increased in the 10% stretch group. After 7 days of expansion, the number of blood vessels in the expanded area, expression of the angiogenesis-associated proteins vascular endothelial growth factor, basic fibroblast growth factor, and hepatocyte growth factor, and collagen deposition were significantly increased in the 10% stretch group. CONCLUSIONS The optimal mechanical force upregulates specific paracrine proteins in ADSCs to increase angiogenesis and collagen secretion, and thereby promote skin regeneration and expansion. This study provides a new auxiliary method to expand large skin flaps.
Collapse
Grants
- 202312121015, S202312121094, 202312121227, 202312121313, 202312121314, 202312121317, 202312121321 College Students' Innovative Entrepreneurial Training Plan Program
- 82202474, 82360615 National Natural Science Foundation of China
- 2022CR007 Clinical Program of Nanfang Hospital, Southern Medical University
- KHYJ-2023-5-02,2023-KHRCBZ-B14 First People's Hospital of Yunnan Province
- 2021A1515110440 Basic and Applied Basic Research Foundation of Guangdong Province
- 2024A04J5192, 2023A04J2350, 2023A04J2349, 2023A04J2347, 2023A04J2271 Science and Technology Projects in Guangzhou
- College Students’ Innovative Entrepreneurial Training Plan Program
- First People’s Hospital of Yunnan Province
Collapse
Affiliation(s)
- Zhixin Xue
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, P. R. China
| | - Delin Hu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, P. R. China
| | - Haojing Tang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, P. R. China
| | - Mingheng Xue
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, P. R. China
| | - Yufan Zhu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, P. R. China
| | - Ye Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, P. R. China.
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, P. R. China.
| |
Collapse
|
8
|
Bobis-Wozowicz S, Paw M, Sarna M, Kędracka-Krok S, Nit K, Błażowska N, Dobosz A, Hammad R, Cathomen T, Zuba-Surma E, Tyszka-Czochara M, Madeja Z. Hypoxic extracellular vesicles from hiPSCs protect cardiomyocytes from oxidative damage by transferring antioxidant proteins and enhancing Akt/Erk/NRF2 signaling. Cell Commun Signal 2024; 22:356. [PMID: 38982464 PMCID: PMC11232324 DOI: 10.1186/s12964-024-01722-7] [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: 02/25/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Stem cell-derived extracellular vesicles (EVs) are an emerging class of therapeutics with excellent biocompatibility, bioactivity and pro-regenerative capacity. One of the potential targets for EV-based medicines are cardiovascular diseases (CVD). In this work we used EVs derived from human induced pluripotent stem cells (hiPSCs; hiPS-EVs) cultured under different oxygen concentrations (21, 5 and 3% O2) to dissect the molecular mechanisms responsible for cardioprotection. METHODS EVs were isolated by ultrafiltration combined with size exclusion chromatography (UF + SEC), followed by characterization by nanoparticle tracking analysis, atomic force microscopy (AFM) and Western blot methods. Liquid chromatography and tandem mass spectrometry coupled with bioinformatic analyses were used to identify differentially enriched proteins in various oxygen conditions. We directly compared the cardioprotective effects of these EVs in an oxygen-glucose deprivation/reoxygenation (OGD/R) model of cardiomyocyte (CM) injury. Using advanced molecular biology, fluorescence microscopy, atomic force spectroscopy and bioinformatics techniques, we investigated intracellular signaling pathways involved in the regulation of cell survival, apoptosis and antioxidant response. The direct effect of EVs on NRF2-regulated signaling was evaluated in CMs following NRF2 inhibition with ML385. RESULTS We demonstrate that hiPS-EVs derived from physiological hypoxia at 5% O2 (EV-H5) exert enhanced cytoprotective function towards damaged CMs compared to EVs derived from other tested oxygen conditions (normoxia; EV-N and hypoxia 3% O2; EV-H3). This resulted from higher phosphorylation rates of Akt kinase in the recipient cells after transfer, modulation of AMPK activity and reduced apoptosis. Furthermore, we provide direct evidence for improved calcium signaling and sustained contractility in CMs treated with EV-H5 using AFM measurements. Mechanistically, our mass spectrometry and bioinformatics analyses revealed differentially enriched proteins in EV-H5 associated with the antioxidant pathway regulated by NRF2. In this regard, EV-H5 increased the nuclear translocation of NRF2 protein and enhanced its transcription in CMs upon OGD/R. In contrast, inhibition of NRF2 with ML385 abolished the protective effect of EVs on CMs. CONCLUSIONS In this work, we demonstrate a superior cardioprotective function of EV-H5 compared to EV-N and EV-H3. Such EVs were most effective in restoring redox balance in stressed CMs, preserving their contractile function and preventing cell death. Our data support the potential use of hiPS-EVs derived from physiological hypoxia, as cell-free therapeutics with regenerative properties for the treatment of cardiac diseases.
Collapse
Affiliation(s)
- Sylwia Bobis-Wozowicz
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland.
| | - Milena Paw
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland
| | - Michał Sarna
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics, Jagiellonian University, Krakow, Poland
| | - Sylwia Kędracka-Krok
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Physical Biochemistry, Jagiellonian University, Krakow, Poland
| | - Kinga Nit
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland
| | - Natalia Błażowska
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland
| | - Anna Dobosz
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland
| | - Ruba Hammad
- Freiburg iPS Core Facility, Institute for Transfusion Medicine and Gene Therapy, Medical Center- University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany
| | - Toni Cathomen
- Freiburg iPS Core Facility, Institute for Transfusion Medicine and Gene Therapy, Medical Center- University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany
| | - Ewa Zuba-Surma
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland
| | - Małgorzata Tyszka-Czochara
- Faculty of Pharmacy, Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, Kraków, Poland
| | - Zbigniew Madeja
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
9
|
Haider KH. Priming mesenchymal stem cells to develop "super stem cells". World J Stem Cells 2024; 16:623-640. [PMID: 38948094 PMCID: PMC11212549 DOI: 10.4252/wjsc.v16.i6.623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/04/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
The stem cell pre-treatment approaches at cellular and sub-cellular levels encompass physical manipulation of stem cells to growth factor treatment, genetic manipulation, and chemical and pharmacological treatment, each strategy having advantages and limitations. Most of these pre-treatment protocols are non-combinative. This editorial is a continuum of Li et al's published article and Wan et al's editorial focusing on the significance of pre-treatment strategies to enhance their stemness, immunoregulatory, and immunosuppressive properties. They have elaborated on the intricacies of the combinative pre-treatment protocol using pro-inflammatory cytokines and hypoxia. Applying a well-defined multi-pronged combinatorial strategy of mesenchymal stem cells (MSCs), pre-treatment based on the mechanistic understanding is expected to develop "Super MSCs", which will create a transformative shift in MSC-based therapies in clinical settings, potentially revolutionizing the field. Once optimized, the standardized protocols may be used with slight modifications to pre-treat different stem cells to develop "super stem cells" with augmented stemness, functionality, and reparability for diverse clinical applications with better outcomes.
Collapse
Affiliation(s)
- Khawaja Husnain Haider
- Department of Basic Sciences, Sulaiman AlRajhi University, AlQaseem 52736, Saudi Arabia.
| |
Collapse
|
10
|
Ding Z, Greenberg ZF, Serafim MF, Ali S, Jamieson JC, Traktuev DO, March K, He M. Understanding molecular characteristics of extracellular vesicles derived from different types of mesenchymal stem cells for therapeutic translation. EXTRACELLULAR VESICLE 2024; 3:100034. [PMID: 38957857 PMCID: PMC11218754 DOI: 10.1016/j.vesic.2024.100034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Mesenchymal stem cells (MSCs) have been studied for decades as candidates for cellular therapy, and their secretome, including secreted extracellular vesicles (EVs), has been identified to contribute significantly to regenerative and reparative functions. Emerging evidence has suggested that MSC-EVs alone, could be used as therapeutics that emulate the biological function of MSCs. However, just as with MSCs, MSC-EVs have been shown to vary in composition, depending on the tissue source of the MSCs as well as the protocols employed in culturing the MSCs and obtaining the EVs. Therefore, the importance of careful choice of cell sources and culture environments is receiving increasing attention. Many factors contribute to the therapeutic potential of MSC-EVs, including the source tissue, isolation technique, and culturing conditions. This review illustrates the molecular landscape of EVs derived from different types of MSC cells along with culture strategies. A thorough analysis of publicly available omic datasets was performed to advance the precision understanding of MSC-EVs with unique tissue source-dependent molecular characteristics. The tissue-specific protein and miRNA-driven Reactome ontology analysis was used to reveal distinct patterns of top Reactome ontology pathways across adipose, bone marrow, and umbilical MSC-EVs. Moreover, a meta-analysis assisted by an AI technique was used to analyze the published literature, providing insights into the therapeutic translation of MSC-EVs based on their source tissues.
Collapse
Affiliation(s)
- Zuo Ding
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Zachary F. Greenberg
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Maria Fernanda Serafim
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Samantha Ali
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Julia C. Jamieson
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Dmitry O. Traktuev
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Keith March
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Mei He
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
11
|
Heydari P, Zargar Kharazi A, Shariati L. Enhanced wound regeneration by PGS/PLA fiber dressing containing platelet-rich plasma: an in vitro study. Sci Rep 2024; 14:12019. [PMID: 38797743 PMCID: PMC11128439 DOI: 10.1038/s41598-024-62855-w] [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: 09/05/2023] [Accepted: 05/22/2024] [Indexed: 05/29/2024] Open
Abstract
Novel wound dressings with therapeutic effects are being continually designed to improve the wound healing process. In this study, the structural, chemical, physical, and biological properties of an electrospun poly glycerol sebacate/poly lactide acid/platelet-rich plasma (PGS/PLA-PRP) nanofibers were evaluated to determine its impacts on in vitro wound healing. Results revealed desirable cell viability in the Fibroblast (L929) and macrophage (RAW-264.7) cell lines as well as human umbilical vein endothelial cells (HUVEC). Cell migration was evident in the scratch assay (L929 cell line) so that it promoted scratch contraction to accelerate in vitro wound healing. Moreover, addition of PRP to the fiber structure led to enhanced collagen deposition (~ 2 times) in comparison with PGS/PLA scaffolds. While by addition PRP to PGS/PLA fibers not only decreased the expression levels of pro-inflammatory cytokines (IL-6 and TNF-α) in RAW-264.7 cells but also led to significantly increased levels of cytokine (IL-10) and the growth factor (TGF-β), which are related to the anti-inflammatory phase (M2 phenotype). Finally, PGS/PLA-PRP was found to induce a significant level of angiogenesis by forming branching points, loops, and tubes. Based on the results obtained, the PGS/PLA-PRP dressing developed might be a promising evolution in skin tissue engineering ensuring improved wound healing and tissue regeneration.
Collapse
Affiliation(s)
- Parisa Heydari
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran
- Applied Physiology Research Center, Isfahan, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anousheh Zargar Kharazi
- Applied Physiology Research Center, Isfahan, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
- Biomaterials Nanotechnology and Tissue Engineering Faculty, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Laleh Shariati
- Applied Physiology Research Center, Isfahan, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
12
|
Phinney DG. Alexander Friedenstein, Mesenchymal Stem Cells, Shifting Paradigms and Euphemisms. Bioengineering (Basel) 2024; 11:534. [PMID: 38927770 PMCID: PMC11201071 DOI: 10.3390/bioengineering11060534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Six decades ago, Friedenstein and coworkers published a series of seminal papers identifying a cell population in bone marrow with osteogenic potential, now referred to as mesenchymal stem cells (MSCs). This work was also instrumental in establishing the identity of hematopoietic stem cell and the identification of skeletal stem/progenitor cell (SSPC) populations in various skeletal compartments. In recognition of the centenary year of Friedenstein's birth, I review key aspects of his work and discuss the evolving concept of the MSC and its various euphemisms indorsed by changing paradigms in the field. I also discuss the recent emphasis on MSC stromal quality attributes and how emerging data demonstrating a mechanistic link between stromal and stem/progenitor functions bring renewed relevance to Friedenstein's contributions and much needed unity to the field.
Collapse
Affiliation(s)
- Donald G Phinney
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL 33458, USA
| |
Collapse
|
13
|
Heydari P, Kharaziha M, Varshosaz J, Kharazi AZ, Javanmard SH. Co-release of nitric oxide and L-arginine from poly (β-amino ester)-based adhesive reprogram macrophages for accelerated wound healing and angiogenesis in vitro and in vivo. BIOMATERIALS ADVANCES 2024; 158:213762. [PMID: 38227989 DOI: 10.1016/j.bioadv.2024.213762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 12/06/2023] [Accepted: 01/06/2024] [Indexed: 01/18/2024]
Abstract
Recently, insufficient angiogenesis and prolonged inflammation are crucial challenges of chronic skin wound healing. The sustained release of L-Arginine (L-Arg) and nitric oxide (NO) production can control immune responses, improve angiogenesis, enhance re-epithelialization, and accelerate wound healing. Here, we aim to improve wound healing via the controlled release of NO and L-Arg from poly (β-amino ester) (PβAE). In this regard, PβAE is functionalized with methacrylate poly-L-Arg (PAMA), and the role of PAMA content (50, 66, and 75 wt%) on the adhesive properties, L-Arg, and NO release, as well as collagen deposition, inflammatory responses, and angiogenesis, is investigated in vitro and in vivo. Results show that the PAMA/ PβAE could provide suitable adhesive strength (~25 kPa) for wound healing application. In addition, increasing the PAMA content from 50 to 75 wt% results in an increased release of L-Arg (approximately 1.4-1.7 times) and enhanced NO production (approximately 2 times), promoting skin cell proliferation and migration. The in vitro studies also show that compared to PβAE hydrogel, incorporation of 66 wt% PAMA (PAMA 66 sample) reveals superior collagen I synthesis (~ 3-4 times) of fibroblasts, controlled pro-inflammatory and improved anti-inflammatory cytokines secretion of macrophages, and accelerated angiogenesis (~1.5-2 times). In vivo studies in a rat model with a full-thickness skin defect also demonstrate the PAMA66 sample could accelerate wound healing (~98 %) and angiogenesis, compared to control (untreated wound) and Tegaderm™ commercial wound dressing. In summary, the engineered multifunctional PAMA functionalized PβAE hydrogel with desired NO and L-Arg release, and adhesive properties can potentially reprogram macrophages and accelerate skin healing for chronic wound healing.
Collapse
Affiliation(s)
- Parisa Heydari
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran; Applied Physiology Research Center, Isfahan, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Isfahan, Iran.
| | - Anousheh Zargar Kharazi
- Applied Physiology Research Center, Isfahan, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran; Biomaterials Nanotechnology and Tissue Engineering Faculty, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Isfahan, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
14
|
Kolenc A, Maličev E. Current Methods for Analysing Mesenchymal Stem Cell-Derived Extracellular Vesicles. Int J Mol Sci 2024; 25:3439. [PMID: 38542411 PMCID: PMC10970230 DOI: 10.3390/ijms25063439] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 11/11/2024] Open
Abstract
The use of extracellular vesicles (EVs) generated by mesenchymal stem cells (MSCs) holds great promise as a novel therapeutic approach. Although their immunomodulatory and regeneration potential has been reported to be similar to that of MSCs, the use of MSC-derived EVs in clinical settings will require several problems to be resolved. It is necessary to develop a standardised and widely accepted isolation technology and to improve methods such as the quantification and characterisation of MSC-derived EVs. In this way, EV studies can be compared, the acquired knowledge can be safely transferred to clinical platforms and the clinical results can be evaluated appropriately. There are many procedures for the collection and analysis of vesicles derived from different cells; however, this review provides an overview of methods for the determination of the total protein amount, specific proteins, particle number, non-protein markers like lipids and RNA, microscopy and other methods focusing on MSC-derived EVs.
Collapse
Affiliation(s)
- Ana Kolenc
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
| | - Elvira Maličev
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000 Ljubljana, Slovenia
| |
Collapse
|
15
|
Noor Azlan NAB, Vitus V, Nor Rashid N, Nordin F, Tye GJ, Wan Kamarul Zaman WS. Human mesenchymal stem cell secretomes: Factors affecting profiling and challenges in clinical application. Cell Tissue Res 2024; 395:227-250. [PMID: 38244032 DOI: 10.1007/s00441-023-03857-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 12/21/2023] [Indexed: 01/22/2024]
Abstract
The promising field of regenerative medicine is thrilling as it can repair and restore organs for various debilitating diseases. Mesenchymal stem cells are one of the main components in regenerative medicine that work through the release of secretomes. By adopting the use of the secretome in cell-free-based therapy, we may be able to address the challenges faced in cell-based therapy. As one of the components of cell-free-based therapy, secretome has the advantage of a better safety and efficacy profile than mesenchymal stem cells. However, secretome has its challenges that need to be addressed, such as its bioprocessing methods that may impact the secretome content and its mechanisms of action in clinical settings. Effective and standardization of bioprocessing protocols are important to ensure the supply and sustainability of secretomes for clinical applications. This may eventually impact its commercialization and marketability. In this review, the bioprocessing methods and their impacts on the secretome profile and treatment are discussed. This improves understanding of its fundamental aspects leading to potential clinical applications.
Collapse
Affiliation(s)
| | - Vieralynda Vitus
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
- Centre for Innovation in Medical Engineering, Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nurshamimi Nor Rashid
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Minden, Pulau Pinang, Malaysia
| | - Wan Safwani Wan Kamarul Zaman
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
- Centre for Innovation in Medical Engineering, Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
16
|
Lammi MJ, Qu C. Regulation of Oxygen Tension as a Strategy to Control Chondrocytic Phenotype for Cartilage Tissue Engineering and Regeneration. Bioengineering (Basel) 2024; 11:211. [PMID: 38534484 DOI: 10.3390/bioengineering11030211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Cartilage defects and osteoarthritis are health problems which are major burdens on health care systems globally, especially in aging populations. Cartilage is a vulnerable tissue, which generally faces a progressive degenerative process when injured. This makes it the 11th most common cause of global disability. Conservative methods are used to treat the initial phases of the illness, while orthopedic management is the method used for more progressed phases. These include, for instance, arthroscopic shaving, microfracturing and mosaicplasty, and joint replacement as the final treatment. Cell-based implantation methods have also been developed. Despite reports of successful treatments, they often suffer from the non-optimal nature of chondrocyte phenotype in the repair tissue. Thus, improved strategies to control the phenotype of the regenerating cells are needed. Avascular tissue cartilage relies on diffusion for nutrients acquisition and the removal of metabolic waste products. A low oxygen content is also present in cartilage, and the chondrocytes are, in fact, well adapted to it. Therefore, this raises an idea that the regulation of oxygen tension could be a strategy to control the chondrocyte phenotype expression, important in cartilage tissue for regenerative purposes. This narrative review discusses the aspects related to oxygen tension in the metabolism and regulation of articular and growth plate chondrocytes and progenitor cell phenotypes, and the role of some microenvironmental factors as regulators of chondrocytes.
Collapse
Affiliation(s)
- Mikko J Lammi
- Department of Medical and Translational Biology, Umeå University, SE-90187 Umeå, Sweden
| | - Chengjuan Qu
- Department of Odontology, Umeå University, SE-90187 Umeå, Sweden
| |
Collapse
|
17
|
Ciferri MC, Bruno S, Rosenwasser N, Gorgun C, Reverberi D, Gagliani MC, Cortese K, Grange C, Bussolati B, Quarto R, Tasso R. Standardized Method to Functionalize Plasma-Extracellular Vesicles via Copper-Free Click Chemistry for Targeted Drug Delivery Strategies. ACS APPLIED BIO MATERIALS 2024; 7:827-838. [PMID: 38227342 DOI: 10.1021/acsabm.3c00822] [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: 01/17/2024]
Abstract
Extracellular vesicles (EVs) have emerged as potential vehicles for targeted drug delivery and diagnostic applications. However, achieving consistent and reliable functionalization of EV membranes remains a challenge. Copper-catalyzed click chemistry, commonly used for EV surface modification, poses limitations due to cytotoxicity and interference with biological systems. To overcome these limitations, we developed a standardized method for functionalizing an EV membrane via copper-free click chemistry. EVs derived from plasma hold immense potential as diagnostic and therapeutic agents. However, the isolation and functionalization of EVs from such a complex biofluid represent considerable challenges. We compared three different EV isolation methods to obtain an EV suspension with an optimal purity/yield ratio, and we identified sucrose cushion ultracentrifugation (sUC) as the ideal protocol. We then optimized the reaction conditions to successfully functionalize the plasma-EV surface through a copper-free click chemistry strategy with a fluorescently labeled azide, used as a proof-of-principle molecule. Click-EVs maintained their identity, size, and, more importantly, capacity to be efficiently taken up by responder tumor cells. Moreover, once internalized, click EVs partially followed the endosomal recycling route. The optimized reaction conditions and characterization techniques presented in this study offer a foundation for future investigations and applications of functionalized EVs in drug delivery, diagnostics, and therapeutics.
Collapse
Affiliation(s)
- Maria Chiara Ciferri
- Department of Experimental Medicine, University of Genova, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Silvia Bruno
- Department of Experimental Medicine, University of Genova, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Nicole Rosenwasser
- Department of Experimental Medicine, University of Genova, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Cansu Gorgun
- Department of Experimental Medicine, University of Genova, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Daniele Reverberi
- UO Molecular Pathology, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Maria Cristina Gagliani
- Department of Experimental Medicine, University of Genova, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Katia Cortese
- Department of Experimental Medicine, University of Genova, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Cristina Grange
- Department of Medical Sciences, University of Torino, Via Nizza 52, Torino 10126, Italy
| | - Benedetta Bussolati
- UO Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Rodolfo Quarto
- Department of Experimental Medicine, University of Genova, Largo Rosanna Benzi 10, Genova 16132, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Roberta Tasso
- Department of Experimental Medicine, University of Genova, Largo Rosanna Benzi 10, Genova 16132, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| |
Collapse
|
18
|
Wong C, Stoilova I, Gazeau F, Herbeuval JP, Fourniols T. Mesenchymal stromal cell derived extracellular vesicles as a therapeutic tool: immune regulation, MSC priming, and applications to SLE. Front Immunol 2024; 15:1355845. [PMID: 38390327 PMCID: PMC10881725 DOI: 10.3389/fimmu.2024.1355845] [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: 12/14/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by a dysfunction of the immune system. Mesenchymal stromal cell (MSCs) derived extracellular vesicles (EVs) are nanometer-sized particles carrying a diverse range of bioactive molecules, such as proteins, miRNAs, and lipids. Despite the methodological disparities, recent works on MSC-EVs have highlighted their broad immunosuppressive effect, thus driving forwards the potential of MSC-EVs in the treatment of chronic diseases. Nonetheless, their mechanism of action is still unclear, and better understanding is needed for clinical application. Therefore, we describe in this review the diverse range of bioactive molecules mediating their immunomodulatory effect, the techniques and possibilities for enhancing their immune activity, and finally the potential application to SLE.
Collapse
Affiliation(s)
- Christophe Wong
- EVerZom, Paris, France
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8601, Université Paris Cité, Paris, France
- Chemistry and Biology, Modeling and Immunology for Therapy (CBMIT), Université Paris Cité, Paris, France
| | - Ivana Stoilova
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8601, Université Paris Cité, Paris, France
- Chemistry and Biology, Modeling and Immunology for Therapy (CBMIT), Université Paris Cité, Paris, France
| | - Florence Gazeau
- Matière et Systèmes Complexes (MSC) UMR CNRS 7057, Université Paris Cité, Paris, France
| | - Jean-Philippe Herbeuval
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8601, Université Paris Cité, Paris, France
- Chemistry and Biology, Modeling and Immunology for Therapy (CBMIT), Université Paris Cité, Paris, France
| | | |
Collapse
|
19
|
Zomer HD, de Souza Lima VJ, Bion MC, Brito KNL, Rode M, Stimamiglio MA, Jeremias TDS, Trentin AG. Evaluation of secretomes derived from human dermal and adipose tissue mesenchymal stem/stromal cells for skin wound healing: not as effective as cells. Stem Cell Res Ther 2024; 15:15. [PMID: 38229157 PMCID: PMC10792854 DOI: 10.1186/s13287-023-03630-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/27/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Although the paracrine effects of mesenchymal stem/stromal cells (MSCs) have been recognized as crucial mediators of their regenerative effects on tissue repair, the potential of MSC secretomes as effective substitutes for cellular therapies remains underexplored. METHODS In this study, we compared MSCs from the human dermis (DSCs) and adipose tissue (ASCs) with their secretomes regarding their efficacy for skin wound healing using a translationally relevant murine model. RESULTS Proteomic analysis revealed that while there was a substantial overlap in protein composition between DSC and ASC secretomes, specific proteins associated with wound healing and angiogenesis were differentially expressed. Despite a similar angiogenic potential in vivo, DSC and ASC secretomes were found to be less effective than cells in accelerating wound closure and promoting tissue remodeling. CONCLUSIONS Overall, secretome-treated groups showed intermediary results between cells- and control-treated (empty scaffold) groups. These findings highlight that although secretomes possess therapeutic potential, their efficacy might be limited compared to cellular therapies. This study contributes to the growing understanding of MSC secretomes, emphasizes the need for further protocol optimization, and offers insights into their potential applications in regenerative medicine.
Collapse
Affiliation(s)
- Helena Debiazi Zomer
- Department of Physiological Sciences, University of Florida, Gainesville, USA.
- Department of Cell Biology, Embryology, and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil.
| | - Victor Juan de Souza Lima
- Department of Cell Biology, Embryology, and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Monique Coelho Bion
- Department of Cell Biology, Embryology, and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
- National Institute of Translational Neuroscience, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karynne Nazare Lins Brito
- Department of Cell Biology, Embryology, and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Michele Rode
- Department of Cell Biology, Embryology, and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Marco Augusto Stimamiglio
- Laboratory for Stem Cells Basic Biology, Carlos Chagas Institute, FIOCRUZ/PR, Curitiba, Paraná, Brazil
| | - Talita da Silva Jeremias
- Department of Cell Biology, Embryology, and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Andrea Gonçalves Trentin
- Department of Cell Biology, Embryology, and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Olcar HN, Isildar B, Ozkan S, Ercin M, Gezginci-Oktayoglu S, Koyuturk M. Investigation of conditioned medium properties obtained from human umbilical cord mesenchymal stem/stromal cells preconditioned with dimethyloxalylglycine in a correlation with ultrastructural changes. Microsc Res Tech 2024; 87:159-171. [PMID: 37728208 DOI: 10.1002/jemt.24420] [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: 05/02/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) hold significant therapeutic value due to their regeneration abilities, migration capacity, and immunosuppressive and immunomodulatory properties. These cells secrete soluble and insoluble factors, and this complex secretome contributes to their therapeutic effect. Furthermore, stimulation of cells by various external stimuli lead to secretome modifications that can increase the therapeutic efficacy. So, this study examined the effect of dimethyloxalylglycine (DMOG), a hypoxia-mimetic agent, on secretome profiles and exosome secretions of MSCs by evaluating conditioned medium (CM) and ultrastructural morphologies of the cells in comparison with unpreconditioned MSCs. The appropriate dose and duration of the use of DMOG were determined as 1000 μM and 24 h by evaluating the HIF-1α expression. DMOG-CM and N-CM were collected from MSCs incubated in serum-free medium with/without DMOG for 24 h, respectively. The content analysis of conditioned mediums (CMs) revealed that VEGF, NGF, and IL-4 levels were increased in DMOG-CM. Subsequently, exosomes were isolated from the CMs and were shown by transmission electron microscopy and Western blot analysis in both groups. The effects of CMs on proliferation and migration were determined by in vitro wound healing tests; both CMs increased the fibroblast's migratory and proliferative capacities. According to the ultrastructural evaluation, autophagosome, autolysosome, myelin figure, and microvesicular body structures were abundant in DMOG-preconditioned MSCs. Consistent with the high number of autophagic vacuoles, Beclin-1 expression was increased in those cells. These findings suggested that DMOG could alter MSCs' secretion profile, modify their ultrastructural morphology accordingly, and make the CM a more potent therapeutic tool. RESEARCH HIGHLIGHTS: Preconditioning mesenchymal stem/stromal cells with dimethyloxalylglycine, a hypoxia-mimetic agent, could modify cellular metabolism. Hypoxic mechanisms lead to alterations in the ultrastructural characteristics of mesenchymal stromal/stem cells. Preconditioning with dimethyloxalylglycine leads to ultrastructural and metabolic changes of mesenchymal stromal/stem cells along with modifications in their secretome profiles. Preconditioning of mesenchymal stromal/stem cells could render them a more potent therapeutic tool.
Collapse
Affiliation(s)
- Hanife Nurdan Olcar
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Basak Isildar
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Serbay Ozkan
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Merve Ercin
- Department of Biology, Molecular Biology Section, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Selda Gezginci-Oktayoglu
- Department of Biology, Molecular Biology Section, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Meral Koyuturk
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
21
|
Zheng L, Gong H, Zhang J, Guo L, Zhai Z, Xia S, Hu Z, Chang J, Jiang Y, Huang X, Ge J, Zhang B, Yan M. Strategies to improve the therapeutic efficacy of mesenchymal stem cell-derived extracellular vesicle (MSC-EV): a promising cell-free therapy for liver disease. Front Bioeng Biotechnol 2023; 11:1322514. [PMID: 38155924 PMCID: PMC10753838 DOI: 10.3389/fbioe.2023.1322514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Liver disease has emerged as a significant worldwide health challenge due to its diverse causative factors and therapeutic complexities. The majority of liver diseases ultimately progress to end-stage liver disease and liver transplantation remains the only effective therapy with the limitations of donor organ shortage, lifelong immunosuppressants and expensive treatment costs. Numerous pre-clinical studies have revealed that extracellular vesicles released by mesenchymal stem cells (MSC-EV) exhibited considerable potential in treating liver diseases. Although natural MSC-EV has many potential advantages, some characteristics of MSC-EV, such as heterogeneity, uneven therapeutic effect, and rapid clearance in vivo constrain its clinical translation. In recent years, researchers have explored plenty of ways to improve the therapeutic efficacy and rotation rate of MSC-EV in the treatment of liver disease. In this review, we summarized current strategies to enhance the therapeutic potency of MSC-EV, mainly including optimization culture conditions in MSC or modifications of MSC-EV, aiming to facilitate the development and clinical application of MSC-EV in treating liver disease.
Collapse
Affiliation(s)
- Lijuan Zheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Hui Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Jing Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Linna Guo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Zhuofan Zhai
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shuang Xia
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Zhiyu Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Jing Chang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yizhu Jiang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xinran Huang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jingyi Ge
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| |
Collapse
|
22
|
Rovere M, Reverberi D, Arnaldi P, Palamà MEF, Gentili C. Spheroid size influences cellular senescence and angiogenic potential of mesenchymal stromal cell-derived soluble factors and extracellular vesicles. Front Bioeng Biotechnol 2023; 11:1297644. [PMID: 38162179 PMCID: PMC10756914 DOI: 10.3389/fbioe.2023.1297644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction: The secretome of mesenchymal stromal cells (MSCs) serves as an innovative tool employed in the regenerative medicine approach. In this particular context, three-dimensional (3D) culture systems are widely utilized to better replicate in vivo conditions and facilitate prolonged cell maintenance during culture. The use of spheroids enables the preservation of the classical phenotypical characteristics of MSCs. However, the distinct microenvironment within the spheroid may impact the secretome, thereby enhancing the angiogenic properties of adult MSCs that typically possess a reduced angiogenic potential compared to MSCs derived from perinatal tissues due to the hypoxia created in the internal region of the spheroid. Methods: In this study, large spheroids (2,600 cells, ∼300 μm diameter) and small spheroids (1,000 cells, ∼200 μm diameter) were used to examine the role of spheroid diameter in the generation of nutrients and oxygen gradients, cellular senescence, and the angiogenic potential of secreted factors and extracellular vesicles (EVs). Results: In this study, we demonstrate that large spheroids showed increased senescence and a secretome enriched in pro-angiogenic factors, as well as pro-inflammatory and anti-angiogenic cytokines, while small spheroids exhibited decreased senescence and a secretome enriched in pro-angiogenic molecules. We also demonstrated that 3D culture led to a higher secretion of EVs with classical phenotypic characteristics. Soluble factors and EVs from small spheroids exhibited higher angiogenic potential in a human umbilical vein endothelial cell (HUVEC) angiogenic assay. Discussion: These findings highlighted the necessity of choosing the appropriate culture system for obtaining soluble factors and EVs for specific therapeutic applications.
Collapse
Affiliation(s)
- Matteo Rovere
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | | | - Pietro Arnaldi
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | | | - Chiara Gentili
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
23
|
Li H, Ji XQ, Zhang SM, Bi RH. Hypoxia and inflammatory factor preconditioning enhances the immunosuppressive properties of human umbilical cord mesenchymal stem cells. World J Stem Cells 2023; 15:999-1016. [PMID: 38058960 PMCID: PMC10696190 DOI: 10.4252/wjsc.v15.i11.999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/28/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have great potential for the treatment of various immune diseases due to their unique immunomodulatory properties. However, MSCs exposed to the harsh inflammatory environment of damaged tissue after intravenous transplantation cannot exert their biological effects, and therefore, their therapeutic efficacy is reduced. In this challenging context, an in vitro preconditioning method is necessary for the development of MSC-based therapies with increased immunomodulatory capacity and transplantation efficacy. AIM To determine whether hypoxia and inflammatory factor preconditioning increases the immunosuppressive properties of MSCs without affecting their biological characteristics. METHODS Umbilical cord MSCs (UC-MSCs) were pretreated with hypoxia (2% O2) exposure and inflammatory factors (interleukin-1β, tumor necrosis factor-α, interferon-γ) for 24 h. Flow cytometry, polymerase chain reaction, enzyme-linked immunosorbent assay and other experimental methods were used to evaluate the biological characteristics of pretreated UC-MSCs and to determine whether pretreatment affected the immunosuppressive ability of UC-MSCs in coculture with immune cells. RESULTS Pretreatment with hypoxia and inflammatory factors caused UC-MSCs to be elongated but did not affect their viability, proliferation or size. In addition, pretreatment significantly decreased the expression of coagulation-related tissue factors but did not affect the expression of other surface markers. Similarly, mitochondrial function and integrity were retained. Although pretreatment promoted UC-MSC apoptosis and senescence, it increased the expression of genes and proteins related to immune regulation. Pretreatment increased peripheral blood mononuclear cell and natural killer (NK) cell proliferation rates and inhibited NK cell-induced toxicity to varying degrees. CONCLUSION In summary, hypoxia and inflammatory factor preconditioning led to higher immunosuppressive effects of MSCs without damaging their biological characteristics.
Collapse
Affiliation(s)
- Hang Li
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Xiao-Qing Ji
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Shu-Ming Zhang
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Ri-Hui Bi
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Hospital Affiliated to Shanxi Medical University, The Third Hospital of Shanxi Medical University, Taiyuan 030002, Shanxi Province, China.
| |
Collapse
|
24
|
Hu Z, Wang D, Gong J, Li Y, Ma Z, Luo T, Jia X, Shi Y, Song Z. MSCs Deliver Hypoxia-Treated Mitochondria Reprogramming Acinar Metabolism to Alleviate Severe Acute Pancreatitis Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207691. [PMID: 37409821 PMCID: PMC10477874 DOI: 10.1002/advs.202207691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/18/2023] [Indexed: 07/07/2023]
Abstract
Mitochondrial function impairment due to abnormal opening of the mitochondrial permeability transition pore (MPTP) is considered the central event in acute pancreatitis; however, therapeutic choices for this condition remain controversial. Mesenchymal stem cells (MSCs) are a family member of stem cells with immunomodulatory and anti-inflammatory capabilities that can mitigate damage in experimental pancreatitis. Here, it is shown that MSCs deliver hypoxia-treated functional mitochondria to damaged pancreatic acinar cells (PACs) via extracellular vesicles (EVs), which reverse the metabolic function of PACs, maintain ATP supply, and exhibit an excellent injury-inhibiting effect. Mechanistically, hypoxia inhibits superoxide accumulation in the mitochondria of MSCs and upregulates the membrane potential, which is internalized into PACs via EVs, thus, remodeling the metabolic state. In addition, cargocytes constructed via stem cell denucleation as mitochondrial vectors are shown to exert similar therapeutic effects to MSCs. These findings reveal an important mechanism underlying the role of mitochondria in MSC therapy and offer the possibility of applying mitochondrial therapy to patients with severe acute pancreatitis.
Collapse
Affiliation(s)
- Zhengyu Hu
- Department of Hepatopancreatobiliary SurgeryShanghai Fourth People's HospitalSchool of MedicineTongji UniversityShanghai200434China
- Department of General SurgeryShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230032China
| | - Dongyan Wang
- Department of GastroenterologyShanghai Pudong New Area Gongli HospitalShanghai200135China
| | - Jian Gong
- Department of General SurgeryShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Yan Li
- Department of GastroenterologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Zhilong Ma
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
| | - Tingyi Luo
- Department of General SurgeryShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Xuyang Jia
- Department of General SurgeryShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Yihai Shi
- Department of GastroenterologyShanghai Pudong New Area Gongli HospitalShanghai200135China
| | - Zhenshun Song
- Department of Hepatopancreatobiliary SurgeryShanghai Fourth People's HospitalSchool of MedicineTongji UniversityShanghai200434China
| |
Collapse
|
25
|
Yang Y, Wu Y, Yang D, Neo SH, Kadir ND, Goh D, Tan JX, Denslin V, Lee EH, Yang Z. Secretive derived from hypoxia preconditioned mesenchymal stem cells promote cartilage regeneration and mitigate joint inflammation via extracellular vesicles. Bioact Mater 2023; 27:98-112. [PMID: 37006826 PMCID: PMC10063382 DOI: 10.1016/j.bioactmat.2023.03.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/23/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Secretome derived from mesenchymal stem cells (MSCs) have profound effects on tissue regeneration, which could become the basis of future MSCs therapies. Hypoxia, as the physiologic environment of MSCs, has great potential to enhance MSCs paracrine therapeutic effect. In our study, the paracrine effects of secretome derived from MSCs preconditioned in normoxia and hypoxia was compared through both in vitro functional assays and an in vivo rat osteochondral defect model. Specifically, the paracrine effect of total EVs were compared to that of soluble factors to characterize the predominant active components in the hypoxic secretome. We demonstrated that hypoxia conditioned medium, as well as the corresponding EVs, at a relatively low dosage, were efficient in promoting the repair of critical-sized osteochondral defects and mitigated the joint inflammation in a rat osteochondral defect model, relative to their normoxia counterpart. In vitro functional test shows enhancement through chondrocyte proliferation, migration, and matrix deposition, while inhibit IL-1β-induced chondrocytes senescence, inflammation, matrix degradation, and pro-inflammatory macrophage activity. Multiple functional proteins, as well as a change in EVs' size profile, with enrichment of specific EV-miRNAs were detected with hypoxia preconditioning, implicating complex molecular pathways involved in hypoxia pre-conditioned MSCs secretome generated cartilage regeneration.
Collapse
|
26
|
Guo F, Pan Q, Chen T, Liao S, Li S, Li A, Chen S, Chen J, Xiao Z, Su H, Yang L, Yang C, Liu HF, Pan Q. hUC-MSC transplantation therapy effects on lupus-prone MRL/lpr mice at early disease stages. Stem Cell Res Ther 2023; 14:211. [PMID: 37605271 PMCID: PMC10441722 DOI: 10.1186/s13287-023-03432-2] [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: 01/04/2023] [Accepted: 07/26/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND The efficacy of human umbilical cord mesenchymal stem cell (hUC-MSC) transplantation in treating systemic lupus erythematosus (SLE) has been confirmed by small-scale clinical trials. However, these trials focused on severe or refractory SLE, while few studies focused on mild SLE. Therefore, this study focused on the therapeutic effects of hUC-MSC transplantation in early-stage or mild MRL/lpr lupus model mice. METHODS Commercially available hUC-MSCs were transplanted into 8-week-old MRL/lpr mice by tail vein injection. Flow cytometry was used to analyze B cells and their subsets in the peripheral blood. Further, plasma inflammatory factors, autoantibodies, and plasma biochemical indices were detected using protein chip technology and ELISA kits. In addition, pathological staining and immunofluorescence were performed to detect kidney injury in mice. RESULTS hUC-MSC transplantation did not affect the mice's body weight, and both middle and high dose hUC-MSC transplantation (MD and HD group) actually reduced spleen weight. hUC-MSC transplantation significantly decreased the proportion of plasmablasts (PB), IgG1- PB, IgG1+ PB, IgG1+ memory B (MB) cells, IgG1+ DN MB, and IgG1+ SP MB cells. The hUC-MSC transplantation had significantly reduced plasma levels of inflammatory factors, such as TNF-α, IFN-γ, IL-6, and IL-13. Pathological staining showed that the infiltration of glomerular inflammatory cells was significantly reduced and that the level of glomerular fibrosis was significantly alleviated in hUC-MSC-transplanted mice. Immunofluorescence assays showed that the deposition of IgG and IgM antibodies in the kidneys of hUC-MSC-transplanted mice was significantly lower than in the control. CONCLUSION hUC-MSC transplantation could inhibit the proliferation and differentiation of peripheral blood B cells in the early-stage of MRL/lpr mice, thereby alleviating the plasma inflammatory environment in mice, leading to kidney injury remission. The study provides a new and feasible strategy for SLE treatment.
Collapse
Affiliation(s)
- Fengbiao Guo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Quanren Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Shuzhen Liao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Shangmei Li
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Aifen Li
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Shuxian Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Jiaxuan Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Zengzhi Xiao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Hongyong Su
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Lawei Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Chen Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| | - Qingjun Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| |
Collapse
|
27
|
Roefs MT, Bauzá-Martinez J, van de Wakker SI, Qin J, Olijve WT, Tuinte R, Rozeboom M, Snijders Blok C, Mol EA, Wu W, Vader P, Sluijter JPG. Cardiac progenitor cell-derived extracellular vesicles promote angiogenesis through both associated- and co-isolated proteins. Commun Biol 2023; 6:800. [PMID: 37528162 PMCID: PMC10393955 DOI: 10.1038/s42003-023-05165-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 07/24/2023] [Indexed: 08/03/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived lipid bilayer-enclosed particles that play a role in intercellular communication. Cardiac progenitor cell (CPC)-derived EVs have been shown to protect the myocardium against ischemia-reperfusion injury via pro-angiogenic effects. However, the mechanisms underlying CPC-EV-induced angiogenesis remain elusive. Here, we discovered that the ability of CPC-EVs to induce in vitro angiogenesis and to stimulate pro-survival pathways was lost upon EV donor cell exposure to calcium ionophore. Proteomic comparison of active and non-active EV preparations together with phosphoproteomic analysis of activated endothelial cells identified the contribution of candidate protein PAPP-A and the IGF-R signaling pathway in EV-mediated cell activation, which was further validated using in vitro angiogenesis assays. Upon further purification using iodixanol gradient ultracentrifugation, EVs partly lost their activity, suggesting a co-stimulatory role of co-isolated proteins in recipient cell activation. Our increased understanding of the mechanisms of CPC-EV-mediated cell activation will pave the way to more efficient EV-based therapeutics.
Collapse
Affiliation(s)
- Marieke Theodora Roefs
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Julia Bauzá-Martinez
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - Jiabin Qin
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Willem Theodoor Olijve
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Robin Tuinte
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marjolein Rozeboom
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christian Snijders Blok
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Emma Alise Mol
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
- Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Singapore, Singapore.
| | - Pieter Vader
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | |
Collapse
|
28
|
de Pedro MÁ, López E, González-Nuño FM, Pulido M, Álvarez V, Marchena AM, Preußer C, Szymański W, Pogge von Strandmann E, Graumann J, Sánchez-Margallo FM, Casado JG, Gómez-Serrano M. Menstrual blood-derived mesenchymal stromal cells: impact of preconditioning on the cargo of extracellular vesicles as potential therapeutics. Stem Cell Res Ther 2023; 14:187. [PMID: 37507751 PMCID: PMC10386225 DOI: 10.1186/s13287-023-03413-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have been shown to exert their therapeutic effects through the secretion of broad spectrum of paracrine factors, including extracellular vesicles (EVs). Accordingly, EVs are being pursued as a promising alternative to cell-based therapies. Menstrual blood-derived stromal cells (MenSCs) are a type of MSC that, due to their immunomodulatory and regenerative properties, have emerged as an innovative source. Additionally, new strategies of cell priming may potentially alter the concentration and cargo of released EVs, leading to modification of their biological properties. In this study, we aimed to characterize the EVs released by MenSCs and compare their therapeutic potential under three different preconditioning conditions (proinflammatory stimuli, physioxia, and acute hypoxia). METHODS MenSCs were isolated from five healthy women. Following culturing to 80% confluence, MenSCs were exposed to different priming conditions: basal (21% O2), proinflammatory stimuli (IFNγ and TNFα, 21% O2), physioxia (1-2% O2), and acute hypoxia (< 1% O2) for 48-72 h. Conditioned media from MenSCs was collected after 48 h and EVs were isolated by a combination of ultra-filtration and differential centrifugation. An extensive characterization ranging from nano-flow cytometry (nFC) to quantitative high-throughput shotgun proteomics was performed. Bioinformatics analyses were used to derive hypotheses on their biological properties. RESULTS No differences in the morphology, size, or number of EVs released were detected between priming conditions. The proteome analysis associated with basal MenSC-EVs prominently revealed their immunomodulatory and regenerative capabilities. Furthermore, quantitative proteomic analysis of differentially produced MenSC-EVs provided sufficient evidence for the utility of the differential preconditioning in purpose-tailoring EVs for their therapeutic application: proinflammatory priming enhanced the anti-inflammatory, regenerative and immunomodulatory capacity in the innate response of EVs, physioxia priming also improves tissue regeneration, angiogenesis and their immunomodulatory capacity targeting on the adaptive response, while acute hypoxia priming, increased hemostasis and apoptotic processes regulation in MenSC-EVs, also by stimulating immunomodulation mainly through the adaptive response. CONCLUSIONS Priming of MenSCs under proinflammatory and hypoxic conditions affected the cargo proteome of EVs released, resulting in different therapeutic potential, and thus warrants experimental exploration with the aim to generate better-defined MSC-derived bioproducts.
Collapse
Affiliation(s)
- María Ángeles de Pedro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - Esther López
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain.
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain.
| | | | - María Pulido
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Ana María Marchena
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - Christian Preußer
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
| | - Witold Szymański
- Institute of Translational Proteomics, Biochemical/Pharmacological Center, Philipps University, 35043, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
| | - Johannes Graumann
- Institute of Translational Proteomics, Biochemical/Pharmacological Center, Philipps University, 35043, Marburg, Germany
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - Javier G Casado
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
- Immunology Unit, University of Extremadura, 10003, Cáceres, Spain
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003, Cáceres, Spain
| | - María Gómez-Serrano
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany.
| |
Collapse
|
29
|
Bakare OO, Gokul A, Niekerk LA, Aina O, Abiona A, Barker AM, Basson G, Nkomo M, Otomo L, Keyster M, Klein A. Recent Progress in the Characterization, Synthesis, Delivery Procedures, Treatment Strategies, and Precision of Antimicrobial Peptides. Int J Mol Sci 2023; 24:11864. [PMID: 37511621 PMCID: PMC10380191 DOI: 10.3390/ijms241411864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Infectious diseases are constantly evolving to bypass antibiotics or create resistance against them. There is a piercing alarm for the need to improve the design of new effective antimicrobial agents such as antimicrobial peptides which are less prone to resistance and possess high sensitivity. This would guard public health in combating and overcoming stubborn pathogens and mitigate incurable diseases; however, the emergence of antimicrobial peptides' shortcomings ranging from untimely degradation by enzymes to difficulty in the design against specific targets is a major bottleneck in achieving these objectives. This review is aimed at highlighting the recent progress in antimicrobial peptide development in the area of nanotechnology-based delivery, selectivity indices, synthesis and characterization, their doping and coating, and the shortfall of these approaches. This review will raise awareness of antimicrobial peptides as prospective therapeutic agents in the medical and pharmaceutical industries, such as the sensitive treatment of diseases and their utilization. The knowledge from this development would guide the future design of these novel peptides and allow the development of highly specific, sensitive, and accurate antimicrobial peptides to initiate treatment regimens in patients to enable them to have accommodating lifestyles.
Collapse
Affiliation(s)
- Olalekan Olanrewaju Bakare
- Department of Biochemistry, Faculty of Basic Medical Sciences, Olabisi Onabanjo University, Sagamu 2002, Nigeria
| | - Arun Gokul
- Department of Plant Sciences, Qwaqwa Campus, University of the Free State, Phuthadithjaba 9866, South Africa
| | - Lee-Ann Niekerk
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa
| | - Omolola Aina
- Plant Omics Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa
| | - Ademola Abiona
- Department of Biochemistry, Faculty of Basic Medical Sciences, Olabisi Onabanjo University, Sagamu 2002, Nigeria
| | - Adele Mariska Barker
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa
| | - Gerhard Basson
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa
| | - Mbukeni Nkomo
- Department of Botany, H13 Botany Building, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa
| | - Laetitia Otomo
- Department of Plant Sciences, Qwaqwa Campus, University of the Free State, Phuthadithjaba 9866, South Africa
| | - Marshall Keyster
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa
| | - Ashwil Klein
- Plant Omics Laboratory, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa
| |
Collapse
|
30
|
Zhu Y, Liao ZF, Mo MH, Xiong XD. Mesenchymal Stromal Cell-Derived Extracellular Vesicles for Vasculopathies and Angiogenesis: Therapeutic Applications and Optimization. Biomolecules 2023; 13:1109. [PMID: 37509145 PMCID: PMC10377109 DOI: 10.3390/biom13071109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Extracellular vesicles (EVs), as part of the cellular secretome, have emerged as essential cell-cell communication regulators in multiple physiological and pathological processes. Previous studies have widely reported that mesenchymal stromal cell-derived EVs (MSC-EVs) have potential therapeutic applications in ischemic diseases or regenerative medicine by accelerating angiogenesis. MSC-EVs also exert beneficial effects on other vasculopathies, including atherosclerosis, aneurysm, vascular restenosis, vascular calcification, vascular leakage, pulmonary hypertension, and diabetic retinopathy. Consequently, the potential of MSC-EVs in regulating vascular homeostasis is attracting increasing interest. In addition to native or naked MSC-EVs, modified MSC-EVs and appropriate biomaterials for delivering MSC-EVs can be introduced to this area to further promote their therapeutic applications. Herein, we outline the functional roles of MSC-EVs in different vasculopathies and angiogenesis to elucidate how MSC-EVs contribute to maintaining vascular system homeostasis. We also discuss the current strategies to optimize their therapeutic effects, which depend on the superior bioactivity, high yield, efficient delivery, and controlled release of MSC-EVs to the desired regions, as well as the challenges that need to be overcome to allow their broad clinical translation.
Collapse
Affiliation(s)
- Ying Zhu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Zhao-Fu Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Miao-Hua Mo
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Xing-Dong Xiong
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
31
|
Zheng D, Ruan H, Chen W, Zhang Y, Cui W, Chen H, Shen H. Advances in extracellular vesicle functionalization strategies for tissue regeneration. Bioact Mater 2023; 25:500-526. [PMID: 37056271 PMCID: PMC10087114 DOI: 10.1016/j.bioactmat.2022.07.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/06/2022] [Accepted: 07/19/2022] [Indexed: 11/02/2022] Open
Abstract
Extracellular vesicles (EVs) are nano-scale vesicles derived by cell secretion with unique advantages such as promoting cell proliferation, anti-inflammation, promoting blood vessels and regulating cell differentiation, which benefit their wide applications in regenerative medicine. However, the in vivo therapeutic effect of EVs still greatly restricted by several obstacles, including the off-targetability, rapid blood clearance, and undesired release. To address these issues, biomedical engineering techniques are vastly explored. This review summarizes different strategies to enhance EV functions from the perspective of drug loading, modification, and combination of biomaterials, and emphatically introduces the latest developments of functionalized EV-loaded biomaterials in different diseases, including cardio-vascular system diseases, osteochondral disorders, wound healing, nerve injuries. Challenges and future directions of EVs are also discussed.
Collapse
Affiliation(s)
- Dandan Zheng
- Department of Spine Surgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, 160 Pujian Road, Shanghai, 200127, PR China
| | - Huitong Ruan
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Wei Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Yuhui Zhang
- Department of Spine Surgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, 160 Pujian Road, Shanghai, 200127, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Hao Chen
- Department of Spine Surgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, 160 Pujian Road, Shanghai, 200127, PR China
| | - Hongxing Shen
- Department of Spine Surgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, 160 Pujian Road, Shanghai, 200127, PR China
| |
Collapse
|
32
|
Lee BW, Kwok SK. Mesenchymal Stem/Stromal Cell-Based Therapies in Systemic Rheumatic Disease: From Challenges to New Approaches for Overcoming Restrictions. Int J Mol Sci 2023; 24:10161. [PMID: 37373308 PMCID: PMC10299481 DOI: 10.3390/ijms241210161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Systemic rheumatic diseases, such as rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis, are chronic autoimmune diseases affecting multiple organs and tissues. Despite recent advances in treatment, patients still experience significant morbidity and disability. Mesenchymal stem/stromal cell (MSC)-based therapy is promising for treating systemic rheumatic diseases due to the regenerative and immunomodulatory properties of MSCs. However, several challenges need to be overcome to use MSCs in clinical practice effectively. These challenges include MSC sourcing, characterization, standardization, safety, and efficacy issues. In this review, we provide an overview of the current state of MSC-based therapies in systemic rheumatic diseases, highlighting the challenges and limitations associated with their use. We also discuss emerging strategies and novel approaches that can help overcome the limitations. Finally, we provide insights into the future directions of MSC-based therapies for systemic rheumatic diseases and their potential clinical applications.
Collapse
Affiliation(s)
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
33
|
Phinney DG, Hwa Lee R, Boregowda SV. Revisiting the Mesenchymal "Stem vs. Stromal" Cell Dichotomy and Its Implications for Development of Improved Potency Metrics. Stem Cells 2023; 41:444-452. [PMID: 36891977 PMCID: PMC10183967 DOI: 10.1093/stmcls/sxad019] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/21/2023] [Indexed: 03/10/2023]
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapies have been evaluated in over 1500 human clinical trials for a diverse array of disease indication, but outcomes remain unpredictable due to knowledge gaps in the quality attributes that confer therapeutic potency onto cells and their mode of action in vivo. Based on accumulated evidence from pre-clinical models, MSCs exert therapeutic effects by repressing inflammatory and immune-mediated response via paracrine action following reprogramming by the host injury microenvironment, and by polarization of tissue resident macrophages following phagocytosis to an alternatively activated (M2) state. An important tenet of this existing paradigm is that well-established stem/progenitor functions of MSCs are independent of paracrine function and dispensable for their anti-inflammatory and immune suppressive functions. Herein, we review evidence that stem/progenitor and paracrine functions of MSCs are mechanistically linked and organized hierarchically and describe how this link may be exploited to develop metrics that predict MSC potency across a spectrum of activities and regenerative medicine applications.
Collapse
Affiliation(s)
- Donald G Phinney
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| | - Ryang Hwa Lee
- Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Siddaraju V Boregowda
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| |
Collapse
|
34
|
Su Y, Ai S, Shen Y, Cheng W, Xu C, Sui L, Zhao Y. Regulatory Effects of Three-Dimensional Cultured Lipopolysaccharide-Pretreated Periodontal Ligament Stem Cell-Derived Secretome on Macrophages. Int J Mol Sci 2023; 24:ijms24086981. [PMID: 37108145 PMCID: PMC10139044 DOI: 10.3390/ijms24086981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Phenotypic transformation of macrophages plays important immune response roles in the occurrence, development and regression of periodontitis. Under inflammation or other environmental stimulation, mesenchymal stem cells (MSCs) exert immunomodulatory effects through their secretome. It has been found that secretome derived from lipopolysaccharide (LPS)-pretreated or three-dimensional (3D)-cultured MSCs significantly reduced inflammatory responses in inflammatory diseases, including periodontitis, by inducing M2 macrophage polarization. In this study, periodontal ligament stem cells (PDLSCs) pretreated with LPS were 3D cultured in hydrogel (termed SupraGel) for a certain period of time and the secretome was collected to explore its regulatory effects on macrophages. Expression changes of immune cytokines in the secretome were also examined to speculate on the regulatory mechanisms in macrophages. The results indicated that PDLSCs showed good viability in SupraGel and could be separated from the gel by adding PBS and centrifuging. The secretome derived from LPS-pretreated and/or 3D-cultured PDLSCs all inhibited the polarization of M1 macrophages, while the secretome derived from LPS-pretreated PDLSCs (regardless of 3D culture) had the ability to promote the polarization of M1 to M2 macrophages and the migration of macrophages. Cytokines involved in the production, migration and polarization of macrophages, as well as multiple growth factors, increased in the PDLSC-derived secretome after LPS pretreatment and/or 3D culture, which suggested that the secretome had the potential to regulate macrophages and promote tissue regeneration, and that it could be used in the treatment of inflammation-related diseases such as periodontitis in the future.
Collapse
Affiliation(s)
- Yuran Su
- Department of Prosthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Sifan Ai
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Youqing Shen
- Department of Orthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Wen Cheng
- Department of Orthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Chenyu Xu
- Department of Orthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Lei Sui
- Department of Prosthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Yanhong Zhao
- Department of Orthodontics, School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
35
|
Liu M, Shang Y, Liu N, Zhen Y, Chen Y, An Y. Strategies to Improve AFT Volume Retention After Fat Grafting. Aesthetic Plast Surg 2023; 47:808-824. [PMID: 36316460 DOI: 10.1007/s00266-022-03088-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/28/2022] [Indexed: 11/01/2022]
Abstract
BACKGROUND Autologous fat grafting has gained increasing popularity used in plastic surgery as a strategy to improve functional and aesthetic outcome. However, variable augmentation results have concerned surgeons in that volume loss of grafted fat reported fluctuates unsteadily. AIM An optimal technique that clinically maximizes the long-term survival rate of transplantation is in urgent need to be identified. METHOD The PubMed/MEDLINE database was queried to search for animal and human studies published through March of 2022 with search terms related to adipose grafting encompassing liposuction, adipose graft viability, processing technique, adipose-derived stem cell, SVF and others. RESULTS 45 in vivo studies met inclusion criteria. The principal of ideal processing technique is effective purification of fat and protection of tissue viability, such as gauze rolling and washing-filtration devices. Cell-assisted lipotransfer including SVF, SVF-gel and ADSCs significantly promotes graft retention via differentiation potential and paracrine manner. ADSCs induce polarization of macrophages to regulate inflammatory response, mediate extracellular matrix remodeling and promote endothelial cell migration and sprouting, and differentiate into adipocytes to replace necrotic cells, providing powerful evidence for the benefits and efficacy of cell-assisted lipotransfer. CONCLUSION Based on the current evidence, the best strategy can not be decided. Cell-assisted lipotransfer has great potential for use in regenerative medicine. But so far mechanically prepared SVF-gel is conducive to clinical promotion. PRP as endogenous growth factor sustained-release material shows great feasibility. LEVEL OF EVIDENCE IV This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Meiling Liu
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Yujia Shang
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Na Liu
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Youbai Chen
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
36
|
Lu Y, Mai Z, Cui L, Zhao X. Engineering exosomes and biomaterial-assisted exosomes as therapeutic carriers for bone regeneration. Stem Cell Res Ther 2023; 14:55. [PMID: 36978165 PMCID: PMC10053084 DOI: 10.1186/s13287-023-03275-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Mesenchymal stem cell-based therapy has become an effective therapeutic approach for bone regeneration. However, there are still limitations in successful clinical translation. Recently, the secretome of mesenchymal stem cells, especially exosome, plays a critical role in promoting bone repair and regeneration. Exosomes are nanosized, lipid bilayer-enclosed structures carrying proteins, lipids, RNAs, metabolites, growth factors, and cytokines and have attracted great attention for their potential application in bone regenerative medicine. In addition, preconditioning of parental cells and exosome engineering can enhance the regenerative potential of exosomes for treating bone defects. Moreover, with recent advancements in various biomaterials to enhance the therapeutic functions of exosomes, biomaterial-assisted exosomes have become a promising strategy for bone regeneration. This review discusses different insights regarding the roles of exosomes in bone regeneration and summarizes the applications of engineering exosomes and biomaterial-assisted exosomes as safe and versatile bone regeneration agent delivery platforms. The current hurdles of transitioning exosomes from bench to bedside are also discussed.
Collapse
Affiliation(s)
- Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, 510280, Guangzhou, China
| | - Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, 510280, Guangzhou, China
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, 510280, Guangzhou, China.
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, 510280, Guangzhou, China.
| |
Collapse
|
37
|
Prognostic Role of Soluble and Extracellular Vesicle-Associated PD-L1, B7-H3 and B7-H4 in Non-Small Cell Lung Cancer Patients Treated with Immune Checkpoint Inhibitors. Cells 2023; 12:cells12060832. [PMID: 36980174 PMCID: PMC10047289 DOI: 10.3390/cells12060832] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
The treatment of non-small cell lung cancer (NSCLC) has changed dramatically with the advent of immune checkpoint inhibitors (ICIs). Despite encouraging results, their efficacy remains limited to a subgroup of patients. Circulating immune checkpoints in soluble (s) form and associated with extracellular vesicles (EVs) represent promising markers, especially in ICI-based therapeutic settings. We evaluated the prognostic role of PD-L1 and of two B7 family members (B7-H3, B7-H4), both soluble and EV-associated, in a cohort of advanced NSCLC patients treated with first- (n = 56) or second-line (n = 126) ICIs. In treatment-naïve patients, high baseline concentrations of sPD-L1 (>24.2 pg/mL) were linked to worse survival, whereas high levels of sB7-H3 (>0.5 ng/mL) and sB7-H4 (>63.9 pg/mL) were associated with better outcomes. EV characterization confirmed the presence of EVs positive for PD-L1 and B7-H3, while only a small portion of EVs expressed B7-H4. The comparison between biomarker levels at the baseline and in the first radiological assessment under ICI-based treatment showed a significant decrease in EV-PD-L1 and an increase in EV-B7H3 in patients in the disease response to ICIs. Our study shows that sPD-L1, sB7-H3 and sB7-H4 levels are emerging prognostic markers in patients with advanced NSCLC treated with ICIs and suggests potential EV involvement in the disease response to ICIs.
Collapse
|
38
|
Younis T, Jabeen F, Hussain A, Rasool B, Raza Ishaq A, Nawaz A, El-Nashar HAS, El-Shazly M. Antioxidant and Pulmonary Protective Potential of Fraxinus xanthoxyloides Bark Extract against CCl 4 -Induced Toxicity in Rats. Chem Biodivers 2023; 20:e202200755. [PMID: 36722706 DOI: 10.1002/cbdv.202200755] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/02/2023]
Abstract
Fraxinus xanthoxyloides is a perennial shrub belonging to family Oleaceae, traditionally used for malaria, jaundice, pneumonia, inflammation, and rheumatism. Our study is aimed to assess the total phenolics (TPC), flavonoids (TFC), terpenoids contents (TTC) and antioxidant profiling of F. xanthoxyloides methanol bark extract (FXBM) and its fractions, hexane, chloroform, ethyl acetate and aqueous, along with high-performance liquid chromatography with diode-array detection (HPLC-DAD). Further, the antioxidant and pulmonary protective potential was explored against carbon tetrachloride (CCl4 )-induced CCl4-induced pulmonary tissue damage in rats. The highest TPC, TFC and TTC were found in FXBM (133.29±4.19 mg/g), ethyl acetate fraction (279.55±10.35 mg/g), and chloroform fraction (0.79±0.06 mg/g), respectively. The most potent antioxidant capacity was depicted by FXBM (29.21±2.40 μg/mg) and ethyl acetate fraction (91.16±5.51 μg/mg). The HPLC-DAD analysis revealed the predominance of gallic, chlorogenic, vanillic and ferulic acid in FXBM. The administration of CCl4 induced oxidative stress, suppressed antioxidant enzymes' activities including catalase, peroxidase, superoxide dismutase, glutathione peroxidase, glutathione-S-transferase, and glutathione reductase. Further, it increased thiobarbituric acid reactive substances (TBARS) and H2 O2 levels, induced DNA injuries and reduced the total protein and glutathione content in lung tissues. The treatment of rats with FXBM restored these biochemical parameters to the normal level. Moreover, the histopathological studies of lung tissues demonstrated that FXBM protected rats' lung tissues from oxidative damage restoring normal lung functions. Thus, F. xanthoxyloides bark extract is recommended as adjuvant therapy as protective agent for patients with lung disorders.
Collapse
Affiliation(s)
- Tahira Younis
- Department of Biochemistry and Biotechnology, The Women University Multan, 60000, Multan, Pakistan
- Department of Biochemistry, Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Faiza Jabeen
- Department of Zoology, University of Education Bank road campus, 54000, Lahore, Pakistan
| | - Ali Hussain
- Department of Zoology, University of the Punjab, 54590, Lahore, Pakistan
| | - Bilal Rasool
- Department of Zoology, Government College University, 38000, Faisalabad, Pakistan
| | - Ali Raza Ishaq
- Department of Zoology, Government College University, 38000, Faisalabad, Pakistan
| | - Atif Nawaz
- Department of Zoology, Government College University, 38000, Faisalabad, Pakistan
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, 11566, Cairo, Egypt
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, 11566, Cairo, Egypt
- Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, 11835, Cairo, Egypt
| |
Collapse
|
39
|
Pretreated Mesenchymal Stem Cells and Their Secretome: Enhanced Immunotherapeutic Strategies. Int J Mol Sci 2023; 24:ijms24021277. [PMID: 36674790 PMCID: PMC9864323 DOI: 10.3390/ijms24021277] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
Mesenchymal stem cells (MSCs) with self-renewing, multilineage differentiation and immunomodulatory properties, have been extensively studied in the field of regenerative medicine and proved to have significant therapeutic potential in many different pathological conditions. The role of MSCs mainly depends on their paracrine components, namely secretome. However, the components of MSC-derived secretome are not constant and are affected by the stimulation MSCs are exposed to. Therefore, the content and composition of secretome can be regulated by the pretreatment of MSCs. We summarize the effects of different pretreatments on MSCs and their secretome, focusing on their immunomodulatory properties, in order to provide new insights for the therapeutic application of MSCs and their secretome in inflammatory immune diseases.
Collapse
|
40
|
Proteomics profile of mesenchymal stromal cells and extracellular vesicles in normoxic and hypoxic conditions. Cytotherapy 2022; 24:1211-1224. [PMID: 36192337 DOI: 10.1016/j.jcyt.2022.08.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/11/2022] [Accepted: 08/27/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AIMS Although bone marrow-derived mesenchymal stromal cells (MSCs) have demonstrated success in pre-clinical studies, they have shown only mild therapeutic effects in clinical trials. Hypoxia pre-conditioning may optimize the performance of bone marrow-derived MSCs because it better reflects the physiological conditions of their origin. It is not known whether changes in the protein profile caused by hypoxia in MSCs can be extended to the extracellular vesicles (EVs) released from them. The aim of this study was to evaluate the proteomics profile of MSCs and their EVs under normoxic and hypoxic conditions. METHODS Bone marrow-derived MSCs were isolated from six healthy male Wistar rats. After achieving 80% confluence, MSCs were subjected to normoxia (MSC-Norm) (21% oxygen, 5% carbon dioxide, 74% nitrogen) or hypoxia (MSC-Hyp) (1% oxygen, 5% carbon dioxide, 94% nitrogen) for 48 h. Cell viability and oxygen consumption rate were assessed. EVs were extracted from MSCs for each condition (EV-Norm and EV-Hyp) by ultracentrifugation. Total proteins were isolated from MSCs and EVs and prepared for mass spectrometry. EVs were characterized by nanoparticle tracking analysis. Proteomics data were analyzed by PatternLab 4.0, Search Tool for the Retrieval of Interacting Genes/Proteins, Gene Ontology, MetaboAnalyst and Reactome software. RESULTS Cell viability was higher in MSC-Hyp than MSC-Norm (P = 0.007). Basal respiration (P = 0.001), proton leak (P = 0.004) and maximal respiration (P = 0.014) were lower in MSC-Hyp than MSC-Norm, and no changes in adenosine triphosphate-linked and residual respiration were observed. The authors detected 2177 proteins in MSC-Hyp and MSC-Norm, of which 147 were identified in only MSC-Hyp and 512 were identified in only MSC-Norm. Furthermore, 718 proteins were identified in EV-Hyp and EV-Norm, of which 293 were detected in only EV-Hyp and 30 were detected in only EV-Norm. Both MSC-Hyp and EV-Hyp showed enrichment of pathways and biological processes related to glycolysis, the immune system and extracellular matrix organization. CONCLUSIONS MSCs subjected to hypoxia showed changes in their survival and metabolic activity. In addition, MSCs under hypoxia released more EVs, and their content was related to expression of regulatory proteins of the immune system and extracellular matrix organization. Because of the upregulation of proteins involved in glycolysis, gluconeogenesis and glucose uptake during hypoxia, production of reactive oxygen species and expression of immunosuppressive properties may be affected.
Collapse
|
41
|
Yuan Y, Sun J, You T, Shen W, Xu W, Dong Q, Cui M. Extracellular Vesicle-Based Therapeutics in Neurological Disorders. Pharmaceutics 2022; 14:pharmaceutics14122652. [PMID: 36559145 PMCID: PMC9783774 DOI: 10.3390/pharmaceutics14122652] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/26/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Neurological diseases remain some of the major causes of death and disability in the world. Few types of drugs and insufficient delivery across the blood-brain barrier limit the treatment of neurological disorders. The past two decades have seen the rapid development of extracellular vesicle-based therapeutics in many fields. As the physiological and pathophysiological roles of extracellular vesicles are recognized in neurological diseases, they have become promising therapeutics and targets for therapeutic interventions. Moreover, advanced nanomedicine technologies have explored the potential of extracellular vesicles as drug delivery systems in neurological diseases. In this review, we discussed the preclinical strategies for extracellular vesicle-based therapeutics in neurological disorders and the struggles involved in their clinical application.
Collapse
Affiliation(s)
- Yiwen Yuan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Jian Sun
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Tongyao You
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Weiwei Shen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Wenqing Xu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- Correspondence: (Q.D.); (M.C.)
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- Correspondence: (Q.D.); (M.C.)
| |
Collapse
|
42
|
Preconditioned Mesenchymal Stromal Cell-Derived Extracellular Vesicles (EVs) Counteract Inflammaging. Cells 2022; 11:cells11223695. [PMID: 36429124 PMCID: PMC9688039 DOI: 10.3390/cells11223695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Inflammaging is one of the evolutionarily conserved mechanisms underlying aging and is defined as the long-term consequence of the chronic stimulation of the innate immune system. As macrophages are intimately involved in initiating and regulating the inflammatory process, their dysregulation plays major roles in inflammaging. The paracrine factors, and in particular extracellular vesicles (EVs), released by mesenchymal stromal cells (MSCs) retain immunoregulatory effects on innate and adaptive immune responses. In this paper, we demonstrate that EVs derived from MSCs preconditioned with hypoxia inflammatory cytokines exerted an anti-inflammatory role in the context of inflammaging. In this study, macrophages isolated from aged mice presented elevated pro-inflammatory factor levels already in basal conditions compared to the young counterpart, and this pre-activation status increased when cells were challenged with IFN-γ. EVs were able to attenuate the age-associated inflammation, inducing a decrease in the expression of TNF-α, iNOS, and the NADase CD38. Moreover, we demonstrate that EVs counteracted the mitochondrial dysfunction that affected the macrophages, reducing lipid peroxidation and hindering the age-associated impairment of mitochondrial complex I activity, oxygen consumption, and ATP synthesis. These results indicate that preconditioned MSC-derived EVs might be exploited as new anti-aging therapies in a variety of age-related diseases.
Collapse
|
43
|
Wang Y, Fang J, Liu B, Shao C, Shi Y. Reciprocal regulation of mesenchymal stem cells and immune responses. Cell Stem Cell 2022; 29:1515-1530. [DOI: 10.1016/j.stem.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/19/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
|
44
|
Oct4 cooperates with c-Myc to improve mesenchymal-to-endothelial transition and myocardial repair of cardiac-resident mesenchymal stem cells. Stem Cell Res Ther 2022; 13:445. [PMID: 36056383 PMCID: PMC9438134 DOI: 10.1186/s13287-022-03120-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cardiac-resident mesenchymal stem cells (cMSCs) can exhibit fibrotic, proinflammatory, and proangiogenic phenotype in response to myocardial ischemia (Isch). How their phenotypic fate decisions are determined remains poorly understood. Here, we demonstrate that the cooperation of Oct4 and c-Myc in cMSCs creates a preferable mesenchymal-to-endothelial transition (MEndoT) to promote angiogenesis and consequent myocardial repair. METHODS We collected MSCs from cardiac and peripheral blood of rat with left ventricular Isch (LV Isch) 30 days after myocardial infarction (MI) or sham operation. After a comparison of characterization between cMSCs and peripheral blood MSCs (pbMSCs), we conducted transcriptome analysis and RNA sequencing of cMSCs. Using loss/gain-of-function approaches to understand the cooperation of c-Myc and Oct4 on MEndoT of cMSCs under hypoxic condition, we explored the mechanisms through transcriptome and functional experiment, and chromatin immunoprecipitation. Next, we transplanted male cMSCs with overexpression or inhibition of c-Myc/Oct4 into the infarcted myocardium of female rats and evaluated infarct size, cell retention, inflammation, remodeling, and function after 30 days. RESULTS LV Isch switched cMSCs toward both inflammatory and proangiogenic phenotypes, with increased secretion of inflammatory cytokines as well as decreased expression of proangiogenic factors. The effect of LV Isch on pbMSCs was less remarkable. Gene expression heatmap showed imbalance in expression of Oct4 and c-Myc regulating genes associated with remodeling of cMSCs. We provided evidence that cMSCs-specific c-Myc- versus Oct4-overexpression showed divergent genomic signatures, and their corresponding target genes play an important role in regulating cMSCs phenotypic changes. In particular, Oct4 accelerated angiogenesis induced by c-Myc overexpression in cMSCs and inhibited their phenotypic transition into inflammatory cells and fibroblast. Mechanistically, exogenous Oct4 caused c-Myc to translocate from the nucleus to the cytoplasm and activated some of its target signalings including VEGF signaling. Although transplantation of cMSCs alone did not improve LV remodeling and function, cMSCs co-transfected with c-Myc and Oct4 promoted a more positive effect in their survival and reparative properties, increased animal survival, reduced infarct size, decreased scar thickness, inhibited LV remodeling, and improved heart function 30 days after MI. Significantly, Oct4 promoted MEndoT ("Rescue me" signal) of cMSCs after both c-Myc stimulation in vitro and transplantation into the infarcted heart. CONCLUSIONS Myocardial Isch drives resident cMSCs toward multiple phenotypes. Oct4 interacts with c-Myc to promote MEndoT capacity of cMSCs and improve their survival and reparative effects through upregulation of angiogenesis-related signaling pathways. These findings may identify novel targets for stem cell therapy.
Collapse
|
45
|
Zhang S, Tuk B, van de Peppel J, Kremers GJ, Koedam M, Pesch GR, Rahman Z, Hoogenboezem RM, Bindels EMJ, van Neck JW, Boukany PE, van Leeuwen JPTM, van der Eerden BCJ. Microfluidic evidence of synergistic effects between mesenchymal stromal cell-derived biochemical factors and biomechanical forces to control endothelial cell function. Acta Biomater 2022; 151:346-359. [PMID: 35995408 DOI: 10.1016/j.actbio.2022.08.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/20/2022] [Accepted: 08/12/2022] [Indexed: 11/01/2022]
Abstract
A functional vascular system is a prerequisite for bone repair as disturbed angiogenesis often causes non-union. Paracrine factors released from human bone marrow derived mesenchymal stromal cells (BMSCs) have angiogenic effects on endothelial cells. However, whether these paracrine factors participate in blood flow dynamics within bone capillaries remains poorly understood. Here, we used two different microfluidic designs to investigate critical steps during angiogenesis and found pronounced effects of endothelial cell proliferation as well as chemotactic and mechanotactic migration induced by BMSC conditioned medium (CM). The application of BMSC-CM in dynamic cultures demonstrates that bioactive factors in combination with fluidic flow-induced biomechanical signals significantly enhanced endothelial cell migration. Transcriptional analyses of endothelial cells demonstrate the induction of a unique gene expression profile related to tricarboxylic acid cycle and energy metabolism by the combination of BMSC-CM factors and shear stress, which opens an interesting avenue to explore during fracture healing. Our results stress the importance of in vivo - like microenvironments simultaneously including biochemical, biomechanical and oxygen levels when investigating key events during vessel repair. STATEMENT OF SIGNIFICANCE: Our results demonstrate the importance of recapitulating in vivo - like microenvironments when investigating key events during vessel repair. Endothelial cells exhibit enhanced angiogenesis characteristics when simultaneous exposing them to hMSC-CM, mechanical forces and biochemical signals simultaneously. The improved angiogenesis may not only result from the direct effect of growth factors, but also by reprogramming of endothelial cell metabolism. Moreover, with this model we demonstrated a synergistic impact of mechanical forces and biochemical factors on endothelial cell behavior and the expression of genes involved in the TCA cycle and energy metabolism, which opens an interesting new avenue to stimulate angiogenesis during fracture healing.
Collapse
Affiliation(s)
- Shuang Zhang
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bastiaan Tuk
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Jeroen van de Peppel
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Gert-Jan Kremers
- Erasmus Optical Imaging Center, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Marijke Koedam
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Georg R Pesch
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Remco M Hoogenboezem
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Eric M J Bindels
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Johan W van Neck
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Johannes P T M van Leeuwen
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bram C J van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands.
| |
Collapse
|
46
|
Műzes G, Sipos F. Mesenchymal Stem Cell-Derived Secretome: A Potential Therapeutic Option for Autoimmune and Immune-Mediated Inflammatory Diseases. Cells 2022; 11:cells11152300. [PMID: 35892597 PMCID: PMC9367576 DOI: 10.3390/cells11152300] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/16/2022] [Accepted: 07/26/2022] [Indexed: 02/05/2023] Open
Abstract
Immune-mediated inflammatory diseases (IMIDs) encompass several entities such as "classic" autoimmune disorders or immune-mediated diseases with autoinflammatory characteristics. Adult stem cells including mesenchymal stem cells (MSCs) are by far the most commonly used type in clinical practice. However, due to the possible side effects of MSC-based treatments, there is an increase in interest in the MSC-secretome (containing large extracellular vesicles, microvesicles, and exosomes) as an alternative therapeutic option in IMIDs. A wide spectrum of MSC-secretome-related biological activities has been proven thus far including anti-inflammatory, anti-apoptotic, and immunomodulatory properties. In comparison with MSCs, the secretome is less immunogenic but exerts similar biological actions, so it can be considered as an ideal cell-free therapeutic alternative. Additionally, since the composition of the MSC-secretome can be engineered, for a future perspective, it could also be viewed as part of a potential delivery system within nanomedicine, allowing us to specifically target dysfunctional cells or tissues. Although many encouraging results from pre-clinical studies have recently been obtained that strongly support the application of the MSC-secretome in IMIDs, human studies with MSC-secretome administration are still in their infancy. This article reviews the immunomodulatory effects of the MSC-secretome in IMIDs and provides insight into the interpretation of its beneficial biological actions.
Collapse
|
47
|
Fernández-Santos ME, Garcia-Arranz M, Andreu EJ, García-Hernández AM, López-Parra M, Villarón E, Sepúlveda P, Fernández-Avilés F, García-Olmo D, Prosper F, Sánchez-Guijo F, Moraleda JM, Zapata AG. Optimization of Mesenchymal Stromal Cell (MSC) Manufacturing Processes for a Better Therapeutic Outcome. Front Immunol 2022; 13:918565. [PMID: 35812460 PMCID: PMC9261977 DOI: 10.3389/fimmu.2022.918565] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/10/2022] [Indexed: 12/20/2022] Open
Abstract
MSCs products as well as their derived extracellular vesicles, are currently being explored as advanced biologics in cell-based therapies with high expectations for their clinical use in the next few years. In recent years, various strategies designed for improving the therapeutic potential of mesenchymal stromal cells (MSCs), including pre-conditioning for enhanced cytokine production, improved cell homing and strengthening of immunomodulatory properties, have been developed but the manufacture and handling of these cells for their use as advanced therapy medicinal products (ATMPs) remains insufficiently studied, and available data are mainly related to non-industrial processes. In the present article, we will review this topic, analyzing current information on the specific regulations, the selection of living donors as well as MSCs from different sources (bone marrow, adipose tissue, umbilical cord, etc.), in-process quality controls for ensuring cell efficiency and safety during all stages of the manual and automatic (bioreactors) manufacturing process, including cryopreservation, the use of cell banks, handling medicines, transport systems of ATMPs, among other related aspects, according to European and US legislation. Our aim is to provide a guide for a better, homogeneous manufacturing of therapeutic cellular products with special reference to MSCs.
Collapse
Affiliation(s)
- Maria Eugenia Fernández-Santos
- Cardiology Department, HGU Gregorio Marañón. GMP-ATMPs Production Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM). Complutense University, CIBER Cardiovascular (CIBERCV), ISCIII, Madrid, Spain
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
| | - Mariano Garcia-Arranz
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD). Surgery Department, Autonoma University of Madrid, Madrid, Spain
| | - Enrique J. Andreu
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Hematology Department and Cell Therapy Area, Clínica Universidad de Navarra. CIBEROC and IDISNA, Pamplona, Spain
| | - Ana Maria García-Hernández
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Miriam López-Parra
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | - Eva Villarón
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | - Pilar Sepúlveda
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Francisco Fernández-Avilés
- Cardiology Department, HGU Gregorio Marañón. GMP-ATMPs Production Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM). Complutense University, CIBER Cardiovascular (CIBERCV), ISCIII, Madrid, Spain
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
| | - Damian García-Olmo
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD). Surgery Department, Autonoma University of Madrid, Madrid, Spain
| | - Felipe Prosper
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Hematology Department and Cell Therapy Area, Clínica Universidad de Navarra. CIBEROC and IDISNA, Pamplona, Spain
| | - Fermin Sánchez-Guijo
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | - Jose M. Moraleda
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Agustin G. Zapata
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Department of Cell Biology, Complutense University, Madrid, Spain
- *Correspondence: Maria Eugenia Fernández-Santos, ; Agustin G. Zapata,
| |
Collapse
|
48
|
Preconditioning and Engineering Strategies for Improving the Efficacy of Mesenchymal Stem Cell-Derived Exosomes in Cell-Free Therapy. Stem Cells Int 2022; 2022:1779346. [PMID: 35607400 PMCID: PMC9124131 DOI: 10.1155/2022/1779346] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/07/2022] [Accepted: 04/23/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely applied to regenerative medicine owing to their multiple differentiation, self-renewal, and immunomodulatory abilities. Exosomes are cell-secreted natural nanovesicles and thought to be mediators of intercellular communication and material transport. The therapeutic potential of MSCs can be largely attributed to MSC-derived exosomes (MSC-exosomes). Emerging evidence suggests that the therapeutic efficacy of MSC-exosomes is highly dependent on the status of MSCs, and optimization of the extracellular environment affects the exosomal content. Pretreatment methods including three-dimensional cultures, hypoxia, and other biochemical cues have been shown to potentially enhance the biological activity of MSC-exosomes while maintaining or enhancing their production. On the other hand, engineering means to enhance the desired function of MSC-exosomes has been rapidly gaining attention. In particular, biologically active molecule encapsulation and membrane modification can alter or enhance biological functions and targeting of MSC-exosomes. In this review, we summarize two possible strategies to improve the therapeutic activity of MSC-exosomes: preconditioning approaches and engineering exosomes. We also explore the underlying mechanisms of different strategies and discuss their advantages and limitations of the upcoming clinical applications.
Collapse
|
49
|
Tracy EP, Stielberg V, Rowe G, Benson D, Nunes SS, Hoying JB, Murfee WL, LeBlanc AJ. State of the field: cellular and exosomal therapeutic approaches in vascular regeneration. Am J Physiol Heart Circ Physiol 2022; 322:H647-H680. [PMID: 35179976 PMCID: PMC8957327 DOI: 10.1152/ajpheart.00674.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/19/2023]
Abstract
Pathologies of the vasculature including the microvasculature are often complex in nature, leading to loss of physiological homeostatic regulation of patency and adequate perfusion to match tissue metabolic demands. Microvascular dysfunction is a key underlying element in the majority of pathologies of failing organs and tissues. Contributing pathological factors to this dysfunction include oxidative stress, mitochondrial dysfunction, endoplasmic reticular (ER) stress, endothelial dysfunction, loss of angiogenic potential and vascular density, and greater senescence and apoptosis. In many clinical settings, current pharmacologic strategies use a single or narrow targeted approach to address symptoms of pathology rather than a comprehensive and multifaceted approach to address their root cause. To address this, efforts have been heavily focused on cellular therapies and cell-free therapies (e.g., exosomes) that can tackle the multifaceted etiology of vascular and microvascular dysfunction. In this review, we discuss 1) the state of the field in terms of common therapeutic cell population isolation techniques, their unique characteristics, and their advantages and disadvantages, 2) common molecular mechanisms of cell therapies to restore vascularization and/or vascular function, 3) arguments for and against allogeneic versus autologous applications of cell therapies, 4) emerging strategies to optimize and enhance cell therapies through priming and preconditioning, and, finally, 5) emerging strategies to bolster therapeutic effect. Relevant and recent clinical and animal studies using cellular therapies to restore vascular function or pathologic tissue health by way of improved vascularization are highlighted throughout these sections.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Virginia Stielberg
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Daniel Benson
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
| | - James B Hoying
- Advanced Solutions Life Sciences, Manchester, New Hampshire
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Amanda Jo LeBlanc
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
50
|
Wu X, Jin S, Ding C, Wang Y, He D, Liu Y. Mesenchymal Stem Cell-Derived Exosome Therapy of Microbial Diseases: From Bench to Bed. Front Microbiol 2022; 12:804813. [PMID: 35046923 PMCID: PMC8761948 DOI: 10.3389/fmicb.2021.804813] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Microbial diseases are a global health threat, leading to tremendous casualties and economic losses. The strategy to treat microbial diseases falls into two broad categories: pathogen-directed therapy (PDT) and host-directed therapy (HDT). As the typical PDT, antibiotics or antiviral drugs directly attack bacteria or viruses through discerning specific molecules. However, drug abuse could result in antimicrobial resistance and increase infectious disease morbidity. Recently, the exosome therapy, as a HDT, has attracted extensive attentions for its potential in limiting infectious complications and targeted drug delivery. Mesenchymal stem cell-derived exosomes (MSC-Exos) are the most broadly investigated. In this review, we mainly focus on the development and recent advances of the application of MSC-Exos on microbial diseases. The review starts with the difficulties and current strategies in antimicrobial treatments, followed by a comprehensive overview of exosomes in aspect of isolation, identification, contents, and applications. Then, the underlying mechanisms of the MSC-Exo therapy in microbial diseases are discussed in depth, mainly including immunomodulation, repression of excessive inflammation, and promotion of tissue regeneration. In addition, we highlight the latest progress in the clinical translation of the MSC-Exo therapy, by summarizing related clinical trials, routes of administration, and exosome modifications. This review will provide fundamental insights and future perspectives on MSC-Exo therapy in microbial diseases from bench to bedside.
Collapse
Affiliation(s)
| | | | | | | | | | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology and National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology and Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health and NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|