1
|
Tieu A, Stewart DJ, Chwastek D, Lansdell C, Burger D, Lalu MM. Biodistribution of mesenchymal stromal cell-derived extracellular vesicles administered during acute lung injury. Stem Cell Res Ther 2023; 14:250. [PMID: 37705086 PMCID: PMC10500845 DOI: 10.1186/s13287-023-03472-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) are a promising cell-free therapy for acute lung injury (ALI). To date, no studies have investigated their biodistribution in ALI or discerned the timing of administration for maximal lung targeting, which are crucial considerations for clinical translation. Our study aimed to characterize a mouse model of ALI and establish the distribution kinetics and optimal timing of MSC-EV delivery during lung injury. METHODS MSC-EVs were isolated by ultracentrifugation alone (U/C) or tangential flow filtration with ultracentrifugation (TFF-U/C) and characterized by nanoparticle tracking analysis and western blot. A lipopolysaccharide (LPS)-induced mouse model of ALI was established to study the inflammatory response over 72 h. ALI was assessed by histological lung injury score, bronchoalveolar lavage fluid cell count and inflammatory cytokines. For biodistribution studies, ALI mice were intravenously administered fluorescently labeled MSC-EVs to determine the optimal timing of administration and organ-specific biodistribution. Live in vivo and ex vivo fluorescence imaging was conducted at various timepoints post-EV injection. RESULTS EVs isolated by either ultracentrifugation alone or TFF-U/C displayed comparable size distribution (~ 50-350 nm) and EV marker expression (CD63/81). TFF-U/C generated a 5.4-fold higher particle concentration and 3.9-fold higher total protein when compared to ultracentrifugation alone. From the inflammatory time-course study, cell count and IL-1β peaked in bronchoalveolar lavage fluid at 24 h after ALI induction. MSC-EVs delivered at 24 h (as opposed to 0.5 h, 5 h or 10 h) after disease induction resulted in a 2.7-4.4-fold higher lung uptake of EVs. Biodistribution studies comparing organ-specific MSC-EV uptake showed progressive lung accumulation up to 48 h post-delivery (threefold higher than the spleen/liver), with a decline at 72 h. Importantly, lung EV fluorescence at 48 h in ALI mice was significantly elevated as compared to control mice. The lung tropism of MSC-EVs was further validated as therapeutically inert EVs derived from HEK293T cells accumulated mainly to the spleen and liver with a 5.5-fold lower distribution to the lungs as compared to MSC-EVs. CONCLUSION MSC-EVs exhibit maximal lung accumulation when administered during heightened inflammation at 24 h after ALI induction. This lung tropism suggests that MSC-EVs may serve as a practical rescue treatment for acute inflammatory respiratory conditions.
Collapse
Affiliation(s)
- Alvin Tieu
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Department of Anesthesiology and Pain Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Clinical Epidemiology Program, BLUEPRINT Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Duncan J Stewart
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Damian Chwastek
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Clinical Epidemiology Program, BLUEPRINT Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Casey Lansdell
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Clinical Epidemiology Program, BLUEPRINT Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Dylan Burger
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Chronic Disease Program, Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- School of Pharmaceutical Sciences, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Manoj M Lalu
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
- Department of Anesthesiology and Pain Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
- Clinical Epidemiology Program, BLUEPRINT Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
2
|
Dias IX, Cordeiro A, Guimarães JAM, Silva KR. Potential and Limitations of Induced Pluripotent Stem Cells-Derived Mesenchymal Stem Cells in Musculoskeletal Disorders Treatment. Biomolecules 2023; 13:1342. [PMID: 37759742 PMCID: PMC10526864 DOI: 10.3390/biom13091342] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 09/29/2023] Open
Abstract
The burden of musculoskeletal disorders (MSK) is increasing worldwide. It affects millions of people worldwide, decreases their quality of life, and can cause mortality. The treatment of such conditions is challenging and often requires surgery. Thus, it is necessary to discuss new strategies. The therapeutic potential of mesenchymal stem cells (MSC) in several diseases has been investigated with relative success. However, this potential is hindered by their limited stemness and expansion ability in vitro and their high donor variability. MSC derived from induced pluripotent stem cells (iPSC) have emerged as an alternative treatment for MSK diseases. These cells present distinct features, such as a juvenile phenotype, in addition to higher stemness, proliferation, and differentiation potential than those of MSC. Here, we review the opportunities, challenges, and applications of iPSC as relevant clinical therapeutic cell sources for MSK disorders. We discuss iPSC sources from which to derive iMSC and the advantages and disadvantages of iMSC over MSC as a therapeutic approach. We further summarize the main preclinical and clinical studies exploring the therapeutic potential of iMSC in MSK disorders.
Collapse
Affiliation(s)
- Isabelle Xavier Dias
- Teaching and Research Division, National Institute of Traumatology and Orthopaedics, Rio de Janeiro 20940-070, Brazil; (A.C.); (J.A.M.G.)
| | - Aline Cordeiro
- Teaching and Research Division, National Institute of Traumatology and Orthopaedics, Rio de Janeiro 20940-070, Brazil; (A.C.); (J.A.M.G.)
| | - João Antonio Matheus Guimarães
- Teaching and Research Division, National Institute of Traumatology and Orthopaedics, Rio de Janeiro 20940-070, Brazil; (A.C.); (J.A.M.G.)
| | - Karina Ribeiro Silva
- Teaching and Research Division, National Institute of Traumatology and Orthopaedics, Rio de Janeiro 20940-070, Brazil; (A.C.); (J.A.M.G.)
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil
| |
Collapse
|
3
|
Parashar S, Pajai S, Tarang T. Recent Advancement in the Management of Intrauterine Adhesions Using Stem Cell Therapy: A Review Article. Cureus 2023; 15:e43553. [PMID: 37719487 PMCID: PMC10502444 DOI: 10.7759/cureus.43553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
Intrauterine adhesions (IUAs) are the formation of scar tissues in the endometrial cavity. The fibrous tissue in the uterus decreases the space inside the uterine cavity. It includes both endometrium and myometrium. It may lead to hypomenorrhea or amenorrhea, pain, difficulty in conceiving, and recurrent abortion. IUA is caused by uterine tissue damage mostly during surgical procedures such as dilatation and curettage. Other causes may include pregnancy-related complications, miscarriage, abnormal bleeding, infections, fibroid removal, and cesarean section (C-section). Patients generally do not have any symptoms and hence are unaware of the condition. The main therapeutic procedure presently used is hysteroscopic transcervical resection of adhesion (TCRA) with hormonal therapy and nondegradable stent as postoperative adjuvant therapy. It has some major limitations such as failure to prevent recurrence and preserve fertility along with difficulty in endometrial tissue repair due to its anatomical site. These limitations have forced the researchers to think about a better treatment modality. In recent times, a better treatment modality has evolved with stem cell therapy. Therefore, this review presents the recent and advanced therapeutic modalities for the treatment of IUAs.
Collapse
Affiliation(s)
- Shreya Parashar
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Wardha, IND
| | - Sandhya Pajai
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Wardha, IND
| | - Tanmay Tarang
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Wardha, IND
| |
Collapse
|
4
|
Zhang G, Li J, Wang D, Lou H, Zhang C, Liu W. The mechanisms related to fibroblasts in burn surface. Skin Res Technol 2023; 29:e13431. [PMID: 37632175 PMCID: PMC10407725 DOI: 10.1111/srt.13431] [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: 04/11/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023]
Abstract
PURPOSE Mesenchymal stem cells (MSCs) can promote burn wound healing, skin appearance, and function recovery by promoting the differentiation and migration of fibroblasts of a wound. The burn environment can activate the autophagy of MSCs. However, it is not clear whether this autophagy can affect the proliferation and migration of fibroblasts. METHODS In this study, pretreated MSCs with rapamycin and 3-methyladenine modulated autophagy and co-cultured with fibroblasts of burn. Cell migration was detected by immunofluorescence chemical staining. Western blot analysis and enzyme-linked immunosorbent assay were performed to detect 2,3-Dioxygenase (IDO), cytokine synthesis inhibitory factor 10 (IL-10), cytokine synthesis inhibitory factor 6 (IL-6), prostaglandin E2 (PGE2), transforming growth factor beta 1 (TGF-β1) proteins levels, and the autophagy proteins p62 and microtubule-associated protein LC3-II/I. RESULTS We demonstrated that autophagy regulates MSCs survival and proliferation in burn wound transplants and found that autophagy inhibition with 3-methyladenine reduced MSCs-mediated, fibroblast proliferation and migration in burn environment. However, rapamycin-induced autophagy had the opposite effect and increased the TGF-β1 expression. Therefore, we speculate that MSCs may promote fibroblast proliferation and migration by secreting TGF-β1 via the AKT/mTOR (RAC-alpha serine/threonine-protein kinase/mammalian target of rapamycin) pathway. CONCLUSION Autophagy of MSCs regulates burn wound fibroblast proliferation and migration by affecting TGF-β1 and prostaglandin E2 production adjacent to MSCs transplanted on the burn wound. The results of this study provide a potential strategy for promoting MSCs treatment of burns.
Collapse
Affiliation(s)
- Gaofei Zhang
- Department of burn and injurySecond Affiliated Hospital of Kunming Medical UniversityKunming CityChina
| | - Jiamei Li
- Department of burn and injurySecond Affiliated Hospital of Kunming Medical UniversityKunming CityChina
| | - Di Wang
- Department of burn and injurySecond Affiliated Hospital of Kunming Medical UniversityKunming CityChina
| | - Hanxiao Lou
- Department of burn and injurySecond Affiliated Hospital of Kunming Medical UniversityKunming CityChina
| | - Chenying Zhang
- Department of burn and injurySecond Affiliated Hospital of Kunming Medical UniversityKunming CityChina
| | - Wenjun Liu
- Department of burn and injurySecond Affiliated Hospital of Kunming Medical UniversityKunming CityChina
| |
Collapse
|
5
|
Yea JH, Kim Y, Jo CH. Comparison of mesenchymal stem cells from bone marrow, umbilical cord blood, and umbilical cord tissue in regeneration of a full-thickness tendon defect in vitro and in vivo. Biochem Biophys Rep 2023; 34:101486. [PMID: 37234487 PMCID: PMC10206173 DOI: 10.1016/j.bbrep.2023.101486] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/15/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Although mesenchymal stem cells (MSCs) can be obtained from various tissues such as bone marrow (BM), umbilical cord blood (UCB) and umbilical cord tissue (UC), the comparative efficacy of each MSC in tendon regeneration is unknown. Therefore, we investigated the efficacy of MSCs isolated from three different sources on tendon regeneration after injury. We evaluated the potential of BM-, UCB- and UC-MSC to differentiate into tendon-like cells in tensioned three-dimensional construct (T-3D) using gene and histological analysis. In animal experiments, full-thickness tendon defect (FTD) was created in supraspinatus of rats, and injected with Saline and BM-, UCB- and UC-MSC. After two and four weeks, histological evaluations were performed. After inducing tenogenic differentiation, the gene expression of scleraxis, mohawk, type I collagen and tenascin-C was upregulated by 3.12-, 5.92-, 6.01- and 1.61-fold respectively and formation of tendon-like matrix was increased 4.22-fold in UC-MSC compared to BM-MSC in T-3D. In animal experiments, the total degeneration score was lower in the UC-MSC group than in BM-MSC group at both weeks. In heterotopic matrix formation, glycosaminoglycan-rich area was reduced in the UC-MSC group, whereas area was larger in the BM-MSC group than in Saline group at four weeks. In conclusion, UC-MSC is superior to other MSCs in differentiating into tendon-like lineage cells and forming a well-organized tendon-like matrix under T-3D conditions. UC-MSC enhances regeneration of FTD in terms of histological properties compared to BM- and UCB-MSC.
Collapse
Affiliation(s)
- Ji-Hye Yea
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Yeasol Kim
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Chris H. Jo
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
6
|
Stage HJ, Trappe S, Söllig K, Trachsel DS, Kirsch K, Zieger C, Merle R, Aschenbach JR, Gehlen H. Multilineage Differentiation Potential of Equine Adipose-Derived Stromal/Stem Cells from Different Sources. Animals (Basel) 2023; 13:ani13081352. [PMID: 37106915 PMCID: PMC10135324 DOI: 10.3390/ani13081352] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The investigation of multipotent stem/stromal cells (MSCs) in vitro represents an important basis for translational studies in large animal models. The study's aim was to examine and compare clinically relevant in vitro properties of equine MSCs, which were isolated from abdominal (abd), retrobulbar (rb) and subcutaneous (sc) adipose tissue by collagenase digestion (ASCs-SVF) and an explant technique (ASCs-EXP). Firstly, we examined proliferation and trilineage differentiation and, secondly, the cardiomyogenic differentiation potential using activin A, bone morphogenetic protein-4 and Dickkopf-1. Fibroblast-like, plastic-adherent ASCs-SVF and ASCs-EXP were obtained from all sources. The proliferation and chondrogenic differentiation potential did not differ significantly between the isolation methods and localizations. However, abd-ASCs-EXP showed the highest adipogenic differentiation potential compared to rb- and sc-ASCs-EXP on day 7 and abd-ASCs-SVF a higher adipogenic potential compared to abd-ASCs-EXP on day 14. Osteogenic differentiation potential was comparable at day 14, but by day 21, abd-ASCs-EXP demonstrated a higher osteogenic potential compared to abd-ASCs-SVF and rb-ASCs-EXP. Cardiomyogenic differentiation could not be achieved. This study provides insight into the proliferation and multilineage differentiation potential of equine ASCs and is expected to provide a basis for future preclinical and clinical studies in horses.
Collapse
Affiliation(s)
- Hannah J Stage
- Equine Clinic, Surgery and Radiology, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Susanne Trappe
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Katharina Söllig
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Dagmar S Trachsel
- Clinical Unit of Equine Internal Medicine, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Katharina Kirsch
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Cornelia Zieger
- Institute of Veterinary Pathology Department of Veterinary Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 15, 14163 Berlin, Germany
| | - Roswitha Merle
- Institute for Veterinary Epidemiology and Biostatistics, Department of Veterinary Medicine, Freie Universität Berlin, Königsweg 67, 14163 Berlin, Germany
| | - Jörg R Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Heidrun Gehlen
- Equine Clinic, Surgery and Radiology, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| |
Collapse
|
7
|
Baouche M, Krawczenko A, Paprocka M, Klimczak A, Mermillod P, Locatelli Y, Ochota M, Niżański W. Feline umbilical cord mesenchymal stem cells: Isolation and in vitro characterization from distinct parts of the umbilical cord. Theriogenology 2023; 201:116-125. [PMID: 36889011 DOI: 10.1016/j.theriogenology.2022.11.049] [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: 10/07/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) are a particular population of cells that play an essential role in the regeneration potential of the body. As a source of MSCs, the umbilical cord (UC) has significant advantages, such as a no-risk procedure of tissue retrieval after birth and the easiness of MSCs isolation. In the presented study, the cells derived from the feline whole umbilical cord (WUC) and two separate parts of the UC tissue, including Wharton's jelly (WJ) and umbilical cord vessels (UCV), were investigated to check whether they exhibit MSCs characteristics. The cells were isolated and characterized based on their morphology, pluripotency, differentiation potential, and phenotype. In our study MSCs were successfully isolated and cultured from all UC parts; after one week of culture, the cells had a typical spindle shape consistent with MSCs morphology. Cells showed the ability to differentiate into chondrocytes, osteoblasts and adipocytes cells. Two markers typical of MSCs (CD44, CD90) and three pluripotency markers (Oct4, SOX2 and Nanog) were expressed in all cells cultures; but no expression of (CD34, MCH II) was evidenced by flow cytometry and RT-PCR. In addition, WJ-MSCs showed the highest ability of proliferation, more significant pluripotency gene expressions, and greater differentiation potential than the cells isolated from WUC and UCV. Finally, we conclude in this study that cat MSCs derived from all the parts are valuable cells that can be efficiently used in various fields of feline regenerative medicine, but cells from WJ can offer the best clinical utility.
Collapse
Affiliation(s)
- Meriem Baouche
- Wrocław University of Environmental and Life Sciences, Department of Reproduction and Clinic of Farm Animals, 50-366, Wrocław, Poland
| | - Agnieszka Krawczenko
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114, Wroclaw, Poland
| | - Maria Paprocka
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114, Wroclaw, Poland
| | - Aleksandra Klimczak
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114, Wroclaw, Poland
| | - Pascal Mermillod
- Physiology of Reproduction and Behaviors (PR China), UMR085, INRAE, CNRS, University of Tours, 37380, Nouzilly, France
| | - Yann Locatelli
- Physiology of Reproduction and Behaviors (PR China), UMR085, INRAE, CNRS, University of Tours, 37380, Nouzilly, France; Museum National d'Histoire Naturelle, Réserve Zoologique de la Haute Touche, 36290, Obterre, France
| | - Małgorzata Ochota
- Wrocław University of Environmental and Life Sciences, Department of Reproduction and Clinic of Farm Animals, 50-366, Wrocław, Poland.
| | - Wojciech Niżański
- Wrocław University of Environmental and Life Sciences, Department of Reproduction and Clinic of Farm Animals, 50-366, Wrocław, Poland.
| |
Collapse
|
8
|
Yu X, Liu P, Li Z, Zhang Z. Function and mechanism of mesenchymal stem cells in the healing of diabetic foot wounds. Front Endocrinol (Lausanne) 2023; 14:1099310. [PMID: 37008908 PMCID: PMC10061144 DOI: 10.3389/fendo.2023.1099310] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Diabetes has become a global public health problem. Diabetic foot is one of the most severe complications of diabetes, which often places a heavy economic burden on patients and seriously affects their quality of life. The current conventional treatment for the diabetic foot can only relieve the symptoms or delay the progression of the disease but cannot repair damaged blood vessels and nerves. An increasing number of studies have shown that mesenchymal stem cells (MSCs) can promote angiogenesis and re-epithelialization, participate in immune regulation, reduce inflammation, and finally repair diabetic foot ulcer (DFU), rendering it an effective means of treating diabetic foot disease. Currently, stem cells used in the treatment of diabetic foot are divided into two categories: autologous and allogeneic. They are mainly derived from the bone marrow, umbilical cord, adipose tissue, and placenta. MSCs from different sources have similar characteristics and subtle differences. Mastering their features to better select and use MSCs is the premise of improving the therapeutic effect of DFU. This article reviews the types and characteristics of MSCs and their molecular mechanisms and functions in treating DFU to provide innovative ideas for using MSCs to treat diabetic foot and promote wound healing.
Collapse
Affiliation(s)
- Xiaoping Yu
- School of Medicine and Nursing, Chengdu University, Chengdu, Sichuan, China
| | - Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zheng Li
- People’s Hospital of Jiulongpo District, Chongqing, China
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Dama G, Du J, Zhu X, Liu Y, Lin J. Bone marrow-derived mesenchymal stem cells: A promising therapeutic option for the treatment of diabetic foot ulcers. Diabetes Res Clin Pract 2023; 195:110201. [PMID: 36493913 DOI: 10.1016/j.diabres.2022.110201] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 08/31/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Chronic wounds fail to heal through the three normal stages of healing (inflammatory, proliferative, and remodelling), resulting in a chronic tissue injury that is not repaired within the average time limit. Patients suffering from type 1 and type 2 diabetes are prone to develop diabetic foot ulcers (DFUs), which commonly develop into chronic wounds that are non treatable with conventional therapies. DFU develops due to various risk factors, such as peripheral neuropathy, peripheral vascular disease, arterial insufficiency, foot deformities, trauma and impaired resistance to infection. DFUs have gradually become a major problem in the health care system worldwide. In this review, we not only focus on the pathogenesis of DFU but also comprehensively summarize the outcomes of preclinical and clinical studies thus far and the potential therapeutic mechanism of bone marrow-derived mesenchymal stem cells (BMSCs) for the treatment of DFU. Based on the published results, BMSC transplantation can contribute to wound healing through growth factor secretion, anti-inflammation, differentiation into tissue-specific cells, neovascularization, re-epithelialization and angiogenesis in DFUs. Moreover, clinical trials showed that BMSC treatment in patients with diabetic ulcers improved ulcer healing and the ankle-brachial index, ameliorated pain scores, and enhanced claudication walking distances with no reported complications. In conclusion, although BMSC transplantation exhibits promising therapeutic potential in DFU treatment, additional studies should be performed to confirm their efficacy and long-term safety in DFU patients.
Collapse
Affiliation(s)
- Ganesh Dama
- Stem Cell and Biotherapy Engineering Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China; Department of Community Health, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Jiang Du
- Stem Cell and Biotherapy Engineering Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China; College of Medical Engineering, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China
| | - Xinxing Zhu
- Stem Cell and Biotherapy Engineering Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China; College of Medical Engineering, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China
| | - Yanli Liu
- Stem Cell and Biotherapy Engineering Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China; College of Life Sciences and Technology, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China.
| | - Juntang Lin
- Stem Cell and Biotherapy Engineering Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China; College of Medical Engineering, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China; College of Life Sciences and Technology, Xinxiang Medical University, East of JinSui Road #601, 453003 Xinxiang, China.
| |
Collapse
|
10
|
Baranovskii DS, Klabukov ID, Arguchinskaya NV, Yakimova AO, Kisel AA, Yatsenko EM, Ivanov SA, Shegay PV, Kaprin AD. Adverse events, side effects and complications in mesenchymal stromal cell-based therapies. Stem Cell Investig 2022; 9:7. [PMID: 36393919 PMCID: PMC9659480 DOI: 10.21037/sci-2022-025] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/28/2022] [Indexed: 07/22/2023]
Abstract
Numerous clinical studies have shown a wide clinical potential of mesenchymal stromal cells (MSCs) application. However, recent experience has accumulated numerous reports of adverse events and side effects associated with MSCs therapy. Furthermore, the strategies and methods of MSCs therapy did not change significantly in recent decades despite the clinical impact and awareness of potential complications. An extended understanding of limitations could lead to a wider clinical implementation of safe cell therapies and avoid harmful approaches. Therefore, our objective was to summarize the possible negative effects observed during MSCs-based therapies. We were also aimed to discuss the risks caused by weaknesses in cell processing, including isolation, culturing, and storage. Cell processing and cell culture could dramatically influence cell population profile, change protein expression and cell differentiation paving the way for future negative effects. Long-term cell culture led to accumulation of chromosomal abnormalities. Overdosed antibiotics in culture media enhanced the risk of mycoplasma contamination. Clinical trials reported thromboembolism and fibrosis as the most common adverse events of MSCs therapy. Their delayed manifestation generally depends on the patient's individual phenotype and requires specific awareness during the clinical trials with obligatory inclusion in the patient' informed consents. Finally we prepared the safety checklist, recommended for clinical specialists before administration or planning of MSCs therapy.
Collapse
Affiliation(s)
- Denis S. Baranovskii
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | - Ilya D. Klabukov
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering of the National Research Nuclear University MEPhI, Obninsk, Russia
| | - Nadezhda V. Arguchinskaya
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Anna O. Yakimova
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Anastas A. Kisel
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Elena M. Yatsenko
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Sergei A. Ivanov
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Peter V. Shegay
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
11
|
Thanaskody K, Jusop AS, Tye GJ, Wan Kamarul Zaman WS, Dass SA, Nordin F. MSCs vs. iPSCs: Potential in therapeutic applications. Front Cell Dev Biol 2022; 10:1005926. [PMID: 36407112 PMCID: PMC9666898 DOI: 10.3389/fcell.2022.1005926] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/21/2022] [Indexed: 01/24/2023] Open
Abstract
Over the past 2 decades, mesenchymal stem cells (MSCs) have attracted a lot of interest as a unique therapeutic approach for a variety of diseases. MSCs are capable of self-renewal and multilineage differentiation capacity, immunomodulatory, and anti-inflammatory properties allowing it to play a role in regenerative medicine. Furthermore, MSCs are low in tumorigenicity and immune privileged, which permits the use of allogeneic MSCs for therapies that eliminate the need to collect MSCs directly from patients. Induced pluripotent stem cells (iPSCs) can be generated from adult cells through gene reprogramming with ectopic expression of specific pluripotency factors. Advancement in iPS technology avoids the destruction of embryos to make pluripotent cells, making it free of ethical concerns. iPSCs can self-renew and develop into a plethora of specialized cells making it a useful resource for regenerative medicine as they may be created from any human source. MSCs have also been used to treat individuals infected with the SARS-CoV-2 virus. MSCs have undergone more clinical trials than iPSCs due to high tumorigenicity, which can trigger oncogenic transformation. In this review, we discussed the overview of mesenchymal stem cells and induced pluripotent stem cells. We briefly present therapeutic approaches and COVID-19-related diseases using MSCs and iPSCs.
Collapse
Affiliation(s)
- Kalaiselvaan Thanaskody
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amirah Syamimi Jusop
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor, Malaysia
| | - Wan Safwani Wan Kamarul Zaman
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia,Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Sylvia Annabel Dass
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor, Malaysia
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia,*Correspondence: Fazlina Nordin,
| |
Collapse
|
12
|
Lee AY, Jang KH, Jo CH. Minimal Cube Explant Provides Optimal Isolation Condition of Mesenchymal Stem Cells from Umbilical Cord. Tissue Eng Regen Med 2022; 19:793-807. [PMID: 35325405 PMCID: PMC9294096 DOI: 10.1007/s13770-022-00440-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: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Enzymatic digestion and explant method have been widely used for isolating umbilical cord-derived mesenchymal stem cells (UC MSCs), although there is still a strong need for robust protocols for optimal isolation for large-scale stem cell banks. This study aims to establish an explant method for clinical scale production of MSCs from human UC tissue and to characterize UC MSCs isolated and cultured with the explant method. METHODS UC MSCs were isolated by enzymatic digestion, minimal cube explant (MCE) 1-2, MCE 2-4, and MCE 10 and cultured, respectively. Also, human antibody array and basic fibroblast growth factor (bFGF) secretion in conditioned medium (CM) was analyzed. The cells were evaluated initial cell number, colony forming unit-fibroblast (CFU-F), proliferation capacity, CD marker expression, and multi-lineage differentiation. SA-β-gal assay as well as expression of p16, p21 and p53 was performed by RT-PCR. RESULTS MCE 2-4 is the most optimized method for isolation of small umbilical cord-derived fast proliferating cells (smumf cells) with the greatest number. MCE 2-4 had the highest secretion of various bioactive factors including bFGF. The MCE 2-4 provided significantly higher CD146 expression than enzymatic digestion, and that expression was maintained until P20. The gene expression of p16, p21, and p53 of smumf cells did not change until P10 and SA-β-gal activity did not increase until P14. CONCLUSION This study demonstrated that MCE 2-4 provided an optimal environment to isolate MSCs with quantity and quality from human whole UC tissue through secretion of various bioactive factors inherent to UC.
Collapse
Affiliation(s)
- Ah-Young Lee
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea
| | - Kwi-Hoon Jang
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea
| | - Chris Hyunchul Jo
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea.
- Department of Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
13
|
Cui X, Alcala-Orozco CR, Baer K, Li J, Murphy C, Durham M, Lindberg G, Hooper GJ, Lim K, Woodfield TBF. 3D bioassembly of cell-instructive chondrogenic and osteogenic hydrogel microspheres containing allogeneic stem cells for hybrid biofabrication of osteochondral constructs. Biofabrication 2022; 14. [PMID: 35344942 DOI: 10.1088/1758-5090/ac61a3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/28/2022] [Indexed: 12/21/2022]
Abstract
Recently developed modular bioassembly techniques hold tremendous potential in tissue engineering and regenerative medicine, due to their ability to recreate the complex microarchitecture of native tissue. Here, we developed a novel approach to fabricate hybrid tissue-engineered constructs adopting high-throughput microfluidic and 3D bioassembly strategies. Osteochondral tissue fabrication was adopted as an example in this study, because of the challenges in fabricating load bearing osteochondral tissue constructs with phenotypically distinct zonal architecture. By developing cell-instructive chondrogenic and osteogenic bioink microsphere modules in high-throughput, together with precise manipulation of the 3D bioassembly process, we successfully fabricated hybrid engineered osteochondral tissue in vitro with integrated but distinct cartilage and bone layers. Furthermore, by encapsulating allogeneic umbilical cord blood-derived mesenchymal stromal cells (UCB-MSCs), and demonstrating chondrogenic and osteogenic differentiation, the hybrid biofabrication of hydrogel microspheres in this 3D bioassembly model offers potential for an off-the-shelf, single-surgery strategy for osteochondral tissue repair.
Collapse
Affiliation(s)
- Xiaolin Cui
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Cesar R Alcala-Orozco
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Kenzie Baer
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Jun Li
- Dept. of Orthopaedic Surgery , University of Otago, 2 Riccarton Avenue, Christchurch, Christchurch, Canterbury, 8011, NEW ZEALAND
| | - Caroline Murphy
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Mitch Durham
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Gabriella Lindberg
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Gary J Hooper
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8041, NEW ZEALAND
| | - Khoon Lim
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Avenue, Christchurch, 8140, NEW ZEALAND
| | - Tim B F Woodfield
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| |
Collapse
|
14
|
Huang J, Li Q, Yuan X, Liu Q, Zhang W, Li P. Intrauterine infusion of clinically graded human umbilical cord-derived mesenchymal stem cells for the treatment of poor healing after uterine injury: a phase I clinical trial. Stem Cell Res Ther 2022; 13:85. [PMID: 35241151 PMCID: PMC8895869 DOI: 10.1186/s13287-022-02756-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/24/2021] [Indexed: 12/19/2022] Open
Abstract
Background Intrauterine adhesion and cesarean scar diverticulum are the main complications of poor healing after uterine injury. Human umbilical cord MSCs transplantation has been regarded as the most potential treatment in the clinic, the safety and efficacy of which in the clinic, however, remains unclear. Methods In this study, ten patients were enrolled: six with intrauterine adhesion and four with cesarean scar diverticulum. All the patients were injected with human umbilical cord MSCs twice into the uterus. Beside the chest X-ray, ECG and abdominal ultrasound, many laboratory tests including blood routine, liver and renal function, ovarian function, tumor biomarkers, and immune function were used to estimate the safe after stem cell transplanted. In addition, the efficacy of stem cell transplanted was shown by the endometrial thickness, the volume of the uterus, and cesarean scar diverticulum based on 3D ultrasound imaging. Results We found that all results of these laboratory tests were normal in these enrolled patients before and after cell injection. Meanwhile, the results of the chest X-ray and ECG were also normal in the treatment process. The abdominal ultrasound showed that the size of the left and right kidneys was inconsistent in one patient after cell therapy, while those of other patients were normal. In addition, endometrial thickness, the volume of the uterus, and cesarean scar diverticulum showed an improving tendency, but no significant difference was noted. Conclusion In summary, intrauterine injection of clinically graded human umbilical cord MSCs was safe for poor healing after uterus injury. Trial registration NCT03386708. Registered 27 December 2017, https://clinicaltrials.gov/ct2/show/NCT03386708?cond=CSD&cntry=CN&draw=2&rank=2 Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02756-9.
Collapse
Affiliation(s)
- Jingrui Huang
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, 410008, Hunan, People's Republic of China
| | - Qi Li
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Xiaohua Yuan
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Qiaoshu Liu
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Weishe Zhang
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, 410008, Hunan, People's Republic of China
| | - Ping Li
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China. .,Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
15
|
Treatment of Radiation Bone Injury with Transplanted hUCB-MSCs via Wnt/ β-Catenin. Stem Cells Int 2021; 2021:5660927. [PMID: 34876908 PMCID: PMC8645406 DOI: 10.1155/2021/5660927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/16/2021] [Indexed: 11/19/2022] Open
Abstract
Radiation-induced bone injury (RIBI) is one of the complications after radiotherapy for malignant tumors. However, there are no effective measures for the treatment of RIBI in clinical practice, and the mechanism of RIBI is unclear. We use a single high-dose ionizing radiation (6Gy) to analyze the effect of radiotherapy on osteoblast function. Human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) were cocultured with irradiated osteoblasts to examine their therapeutic effects and mechanisms on osteoblast injury. The hUCB-MSC transplantation mouse model is used to confirm the in vivo role of hUCB-MSC treatment in radiation bone injury. Western blot analysis, qRT-PCR, immunohistochemistry, and immunofluorescence staining were used to analyze gene expression and angiogenesis. The apoptosis and migration of osteoblasts were measured by Hoechst staining, scratch test, and transwell. The differentiation of osteoblasts was measured by ALP and Alizarin red staining and transmission electron microscopy. The bone-related parameters of mice were evaluated by micro-CT analysis. We found that radiation can damage the DNA of osteoblasts; induce apoptosis; reduce the differentiation, migration, and adhesion of osteoblasts, leading to lipogenesis of bone marrow mesenchymal stem cells (BMSCs) and reducing the source of osteoblasts; and increase the number of osteoclasts in bone tissue, while MSC treatment prevents these changes. Our results reveal the inhibitory effect of radiation on osteoblast function. hUCB-MSCs can be used as a therapeutic target for the development of new therapeutic strategies for radiotherapy of bone injury diseases.
Collapse
|
16
|
Horie S, Gonzalez H, Brady J, Devaney J, Scully M, O’Toole D, Laffey JG. Fresh and Cryopreserved Human Umbilical-Cord-Derived Mesenchymal Stromal Cells Attenuate Injury and Enhance Resolution and Repair following Ventilation-Induced Lung Injury. Int J Mol Sci 2021; 22:ijms222312842. [PMID: 34884645 PMCID: PMC8657992 DOI: 10.3390/ijms222312842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Ventilator-induced lung injury (VILI) frequently worsens acute respiratory distress syndrome (ARDS) severity. Human mesenchymal stem/stromal cells (MSCs) offer considerable therapeutic promise, but the key impediments of clinical translation stem from limitations due to cell source and availability, and concerns regarding the loss of efficacy following cryopreservation. These experiments compared the efficacy of umbilical-cord-derived MSCs (UC-MSCs), a readily available and homogenous tissue source, to the previously more widely utilised bone-marrow-derived MSCs (BM-MSCs). We assessed their capacity to limit inflammation, resolve injury and enhance repair in relevant lung mechanical stretch models, and the impact of cryopreservation on therapeutic efficacy. Methods: In series 1, confluent alveolar epithelial layers were subjected to cyclic mechanical stretch (22% equibiaxial strain) and wound injury, and the potential of the secretome from BM- and UC-derived MSCs to attenuate epithelial inflammation and cell death, and enhance wound repair was determined. In series 2, anesthetized rats underwent VILI, and later received, in a randomised manner, 1 × 107 MSCs/kg intravenously, that were: (i) fresh BM-MSCs, (ii) fresh UC-MSCs or (iii) cryopreserved UC-MSCs. Control animals received a vehicle (PBS). The extent of the resolution of inflammation and injury, and repair was measured at 24 h. Results: Conditioned medium from BM-MSCs and UC-MSCs comparably decreased stretch-induced pulmonary epithelial inflammation and cell death. BM-MSCs and UC-MSCs comparably enhanced wound resolution. In animals subjected to VILI, both fresh BM-MSCs and UC-MSCs enhanced injury resolution and repair, while cryopreserved UC-MSCs comparably retained their efficacy. Conclusions: Cryopreserved UC-MSCs can reduce stretch-induced inflammation and cell death, enhance wound resolution, and enhance injury resolution and repair following VILI. Cryopreserved UC-MSCs represent a more abundant, cost-efficient, less variable and equally efficacious source of therapeutic MSC product.
Collapse
Affiliation(s)
- Shahd Horie
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
| | - Hector Gonzalez
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
| | - Jack Brady
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
| | - James Devaney
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
| | - Michael Scully
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Medicine, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland
| | - Daniel O’Toole
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
- Correspondence: (D.O.); (J.G.L.)
| | - John G. Laffey
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland; (S.H.); (H.G.); (J.B.); (J.D.); (M.S.)
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Biomedical Sciences Building, H91 TK33 Galway, Ireland
- Medicine, School of Medicine, Clinical Sciences Institute, National University of Ireland, H91 TK33 Galway, Ireland
- Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group, H91 YR71 Galway, Ireland
- Correspondence: (D.O.); (J.G.L.)
| |
Collapse
|
17
|
Fang W, Sun Z, Chen X, Han B, Vangsness CT. Synovial Fluid Mesenchymal Stem Cells for Knee Arthritis and Cartilage Defects: A Review of the Literature. J Knee Surg 2021; 34:1476-1485. [PMID: 32403148 DOI: 10.1055/s-0040-1710366] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells that have the ability to self-renew and differentiate into several cell lineages including adipocytes, chondrocytes, tenocytes, bones, and myoblasts. These properties make the cell a promising candidate for regenerative medicine applications, especially when dealing with sports injuries in the knee. MSCs can be isolated from almost every type of adult tissue. However, most of the current research focuses on MSCs derived from bone marrow, adipose, and placenta derived products. Synovial fluid-derived MSCs (SF-MSCs) are relatively overlooked but have demonstrated promising therapeutic properties including possessing higher chondrogenic proliferation capabilities than other types of MSCs. Interestingly, SF-MSC population has shown to increase exponentially in patients with joint injury or disease, pointing to a potential use as a biomarker or as a treatment of some orthopaedic disorders. In this review, we go over the current literature on synovial fluid-derived MSCs including the characterization, the animal studies, and discuss future perspectives.
Collapse
Affiliation(s)
- William Fang
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - ZhiTao Sun
- Department of Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangzhou, China
| | - Xiao Chen
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Bo Han
- Department of Surgery, USC Keck School of Medicine, Los Angeles, California
| | - C Thomas Vangsness
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
18
|
Tieu A, Hu K, Gnyra C, Montroy J, Fergusson DA, Allan DS, Stewart DJ, Thébaud B, Lalu MM. Mesenchymal stromal cell extracellular vesicles as therapy for acute and chronic respiratory diseases: A meta-analysis. J Extracell Vesicles 2021; 10:e12141. [PMID: 34596349 PMCID: PMC8485337 DOI: 10.1002/jev2.12141] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/30/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
Preclinical studies suggest mesenchymal stromal cell extracellular vesicles (MSC-EVs) reduce inflammation and improve organ function in lung diseases; however, an objective analysis of all available data is needed prior to translation. Using rigorous meta-research methods, we determined the effectiveness of MSC-EVs for preclinical respiratory diseases and identified experimental conditions that may further refine this therapy. A systematic search of MEDLINE/Embase identified 1167 records. After screening, 52 articles were included for data extraction and evaluated for risk of bias and quality of reporting in study design. A random effects meta-analysis was conducted for acute lung injury (ALI; N = 23), bronchopulmonary dysplasia (BPD; N = 8) and pulmonary arterial hypertension (PAH; N = 7). Subgroup analyses identified EV methods/characteristics that may be associated with improved efficacy. Data is presented as standardized mean differences (SMD) or risk ratios (RR) with 95% confidence intervals (CI). For ALI, MSC-EVs markedly reduced lung injury (SMD -4.33, CI -5.73 to -2.92), vascular permeability (SMD -2.43, CI -3.05 to -1.82), and mortality (RR 0.39, CI 0.22 to 0.68). Small EVs were more consistently effective than large EVs whereas no differences were observed between tissue sources, immunocompatibility or isolation techniques. For BPD, alveolarization was improved by MSC-EVs (SMD -1.45, CI -2.08 to -0.82) with small EVs more consistently beneficial then small/large EVs. In PAH, right ventricular systolic pressure (SMD -4.16, CI -5.68 to -2.64) and hypertrophy (SMD -2.80, CI -3.68 to -1.91) were significantly attenuated by EVs. In BPD and PAH, EVs isolated by ultracentrifugation demonstrated therapeutic benefit whereas tangential flow filtration (N = 2) displayed minimal efficacy. Lastly, risk of bias and quality of reporting for experimental design were consistently unclear across all studies. Our findings demonstrate clear potential of MSC-EVs to be developed as therapy for acute and chronic lung diseases. However, greater transparency in research design and direct comparisons of isolation technique and EV subtypes are needed to generate robust evidence to guide clinical translation. Protocol Registration: PROSPERO CRD42020145334.
Collapse
Affiliation(s)
- Alvin Tieu
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Kevin Hu
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Catherine Gnyra
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Joshua Montroy
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Dean A. Fergusson
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
- Department of MedicineThe Ottawa HospitalOttawaOntarioCanada
| | - David S. Allan
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Department of MedicineThe Ottawa HospitalOttawaOntarioCanada
| | - Duncan J. Stewart
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Department of MedicineThe Ottawa HospitalOttawaOntarioCanada
| | - Bernard Thébaud
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Division of NeonatologyDepartment of PediatricsChildren's Hospital of Eastern OntarioOttawaOntarioCanada
| | - Manoj M. Lalu
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Departments of Anesthesiology and Pain Medicine, The Ottawa HospitalOttawaOntarioCanada
| |
Collapse
|
19
|
Takayama Y, Kusamori K, Nishikawa M. Mesenchymal stem/stromal cells as next-generation drug delivery vehicles for cancer therapeutics. Expert Opin Drug Deliv 2021; 18:1627-1642. [PMID: 34311638 DOI: 10.1080/17425247.2021.1960309] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Drug delivery to solid tumors remains a significant therapeutic challenge. Mesenchymal stem/stromal cells (MSCs) home to tumor tissues and can be employed as tumor targeted drug/gene delivery vehicles. Reportedly, therapeutic gene- or anti-cancer drug-loaded MSCs have shown remarkable anti-tumor effects in preclinical studies, and some clinical trials for assessing therapeutic MSCs in patients with cancer have been registered. AREAS COVERED In the present review, we first discuss the source and interdonor heterogeneity of MSCs, their tumor-homing mechanism, and the route of MSC administration in MSC-based cancer therapy. We then summarize the therapeutic applications of MSCs as a drug delivery vehicle for therapeutic genes or anti-cancer drugs and the drug delivery mechanism from drug-loaded MSCs to cancer cells. EXPERT OPINION Although numerous preclinical studies have revealed significant anti-tumor effects, several clinical trials assessing MSC-based cancer gene therapy have failed to demonstrate corroborative results, documenting limited therapeutic effects. Notably, a successful clinical outcome with MSC-based cancer therapy would require the interdonor heterogeneity of administered MSCs to be resolved, along with improved tumor-homing efficiency and optimized drug delivery efficiency from MSCs to cancer cells.
Collapse
Affiliation(s)
- Yukiya Takayama
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba Japan
| | - Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba Japan
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba Japan
| |
Collapse
|
20
|
García-Muñoz E, Vives J. Towards the standardization of methods of tissue processing for the isolation of mesenchymal stromal cells for clinical use. Cytotechnology 2021; 73:513-522. [PMID: 33994662 PMCID: PMC8109215 DOI: 10.1007/s10616-021-00474-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are currently the most extensively studied type of adult stem cells in advanced stages of development in the field of regenerative medicine. The biological properties of MSCs have generated great hope for their therapeutic use in degenerative and autoimmune conditions that, at present, lack effective treatment options. Over the last decades, MSCs have been typically obtained from adult bone marrow, but the extraction process is highly invasive and the quality and numbers of isolated cells is drastically influenced by patient age, medication and associated comorbidities. Therefore, there is currently an open discussion on the convenience of allogeneic over autologous treatments, despite potential disadvantages such as rejection by the host. This shift to the allogeneic setting entails the need for high production of MSCs to ensure availability of sufficient cell numbers for transplantation, and therefore making the search for alternative tissue sources of highly proliferative MSC cultures with low levels of senescence occurrence, which is one of the greatest current challenges in the scale up of therapeutic cell bioprocessing. Herein we (i) present the main isolation protocols of MSCs from bone marrow, adipose tissue and Wharton’s jelly of the umbilical cord; and (ii) compare their qualities from a bioprocess standpoint, addressing both quality and regulatory aspects, in view of their anticipated clinical use.
Collapse
Affiliation(s)
- Elisabeth García-Muñoz
- Banc de Sang iTeixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Joaquim Vives
- Banc de Sang iTeixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall D'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035 Barcelona, Spain.,Departament de Medicina, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035 Barcelona, Spain
| |
Collapse
|
21
|
Lindsay SL, Barnett SC. Therapeutic Potential of Niche-Specific Mesenchymal Stromal Cells for Spinal Cord Injury Repair. Cells 2021; 10:cells10040901. [PMID: 33919910 PMCID: PMC8070966 DOI: 10.3390/cells10040901] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
The use of mesenchymal stem/stromal cells (MSCs) for transplant-mediated repair represents an important and promising therapeutic strategy after spinal cord injury (SCI). The appeal of MSCs has been fuelled by their ease of isolation, immunosuppressive properties, and low immunogenicity, alongside the large variety of available tissue sources. However, despite reported similarities in vitro, MSCs sourced from distinct tissues may not have comparable biological properties in vivo. There is accumulating evidence that stemness, plasticity, immunogenicity, and adaptability of stem cells is largely controlled by tissue niche. The extrinsic impact of cellular niche for MSC repair potential is therefore important, not least because of its impact on ex vivo expansion for therapeutic purposes. It is likely certain niche-targeted MSCs are more suited for SCI transplant-mediated repair due to their intrinsic capabilities, such as inherent neurogenic properties. In addition, the various MSC anatomical locations means that differences in harvest and culture procedures can make cross-comparison of pre-clinical data difficult. Since a clinical grade MSC product is inextricably linked with its manufacture, it is imperative that cells can be made relatively easily using appropriate materials. We discuss these issues and highlight the importance of identifying the appropriate niche-specific MSC type for SCI repair.
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW One aim in bone tissue engineering is to develop human cell-based, 3D in vitro bone models to study bone physiology and pathology. Due to the heterogeneity of cells among patients, patient's own cells are needed to be obtained, ideally, from one single cell source. This review attempts to identify the appropriate cell sources for development of such models. RECENT FINDINGS Bone marrow and peripheral blood are considered as suitable sources for extraction of osteoblast/osteocyte and osteoclast progenitor cells. Recent studies on these cell sources have shown no significant differences between isolated progenitor cells. However, various parameters such as medium composition affect the cell's proliferation and differentiation potential which could make the peripheral blood-derived stem cells superior to the ones from bone marrow. Peripheral blood can be considered a suitable source for osteoblast/osteocyte and osteoclast progenitor cells, being less invasive for the patient. However, more investigations are needed focusing on extraction and differentiation of both cell types from the same donor sample of peripheral blood.
Collapse
Affiliation(s)
- Sana Ansari
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands.
| |
Collapse
|
23
|
Gao Y, Wu G, Xu Y, Zhao D, Zheng L. Stem Cell-Based Therapy for Asherman Syndrome: Promises and Challenges. Cell Transplant 2021; 30:9636897211020734. [PMID: 34105392 PMCID: PMC8193648 DOI: 10.1177/09636897211020734] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/23/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
Asherman syndrome (AS) has an adverse effect on reproductive health and fertility by affecting endometrial regeneration. Stem cell-based therapies hold promise for future use in activating non-functional endometrium and reconstructing the endometrium in vivo. It has been postulated that various endometrial stem cells (EnSCs) are responsible for endometrial regeneration. Numerous studies have focused on bone marrow-derived stem cells (BMDSCs), which may provide new ideas for repairing endometrial lesions and reconstructing the endometrium. Other sources of stem cells, such as menstrual blood, umbilical cord, and amniotic membrane, have also attracted much attention as candidates for transplantation in AS. This review discusses the features and specific biomarkers among four types of resident endometrial stem cells, applications of four different sources of exogenous stem cells in AS, and development of stem cell therapy using biomaterials and exosomes.
Collapse
Affiliation(s)
- Yiyin Gao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Guijie Wu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Xu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Donghai Zhao
- Department of Pathology, Jilin Medical University, Jilin, Jilin, China
| | - Lianwen Zheng
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
24
|
Gorman E, Millar J, McAuley D, O'Kane C. Mesenchymal stromal cells for acute respiratory distress syndrome (ARDS), sepsis, and COVID-19 infection: optimizing the therapeutic potential. Expert Rev Respir Med 2020; 15:301-324. [PMID: 33172313 DOI: 10.1080/17476348.2021.1848555] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Mesenchymal stromal (stem) cell (MSC) therapies are emerging as a promising therapeutic intervention in patients with Acute Respiratory Distress Syndrome (ARDS) and sepsis due to their reparative, immunomodulatory, and antimicrobial properties.Areas covered: This review provides an overview of Mesenchymal stromal cells (MSCs) and their mechanisms of effect in ARDS and sepsis. The preclinical and clinical evidence to support MSC therapy in ARDS and sepsis is discussed. The potential for MSC therapy in COVID-19 ARDS is discussed with insights from respiratory viral models and early clinical reports of MSC therapy in COVID-19. Strategies to optimize the therapeutic potential of MSCs in ARDS and sepsis are considered including preconditioning, altered gene expression, and alternative cell-free MSC-derived products, such as extracellular vesicles and conditioned medium.Expert opinion: MSC products present considerable therapeutic promise for ARDS and sepsis. Preclinical investigations report significant benefits and early phase clinical studies have not highlighted safety concerns. Optimization of MSC function in preclinical models of ARDS and sepsis has enhanced their beneficial effects. MSC-derived products, as cell-free alternatives, may provide further advantages in this field. These strategies present opportunity for the clinical development of MSCs and MSC-derived products with enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Ellen Gorman
- School of Medicine Dentistry and Biomedical Science, Queen's University Belfast, UK
| | - Jonathan Millar
- Division of Functional Genetics and Development, Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Danny McAuley
- School of Medicine Dentistry and Biomedical Science, Queen's University Belfast, UK
| | - Cecilia O'Kane
- School of Medicine Dentistry and Biomedical Science, Queen's University Belfast, UK
| |
Collapse
|
25
|
Yang DW, Moon JS, Ko HM, Shin YK, Fukumoto S, Kim SH, Kim MS. Direct reprogramming of fibroblasts into diverse lineage cells by DNA demethylation followed by differentiating cultures. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:463-472. [PMID: 33093268 PMCID: PMC7585590 DOI: 10.4196/kjpp.2020.24.6.463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/24/2020] [Accepted: 09/26/2020] [Indexed: 11/15/2022]
Abstract
Direct reprogramming, also known as a trans-differentiation, is a technique to allow mature cells to be converted into other types of cells without inducing a pluripotent stage. It has been suggested as a major strategy to acquire the desired type of cells in cell-based therapies to repair damaged tissues. Studies related to switching the fate of cells through epigenetic modification have been progressing and they can bypass safety issues raised by the virus-based transfection methods. In this study, a protocol was established to directly convert fully differentiated fibroblasts into diverse mesenchymal-lineage cells, such as osteoblasts, adipocytes, chondrocytes, and ectodermal cells, including neurons, by means of DNA demethylation, immediately followed by culturing in various differentiating media. First, 24 h exposure of 5-azacytidine (5-aza-CN), a well-characterized DNA methyl transferase inhibitor, to NIH-3T3 murine fibroblast cells induced the expression of stem-cell markers, that is, increasing cell plasticity. Next, 5-aza-CN treated fibroblasts were cultured in osteogenic, adipogenic, chondrogenic, and neurogenic media with or without bone morphogenetic protein 2 for a designated period. Differentiation of each desired type of cell was verified by quantitative reverse transcriptase-polymerase chain reaction/western blot assays for appropriate marker expression and by various staining methods, such as alkaline phosphatase/alizarin red S/oil red O/alcian blue. These proposed procedures allowed easier acquisition of the desired cells without any transgenic modification, using direct reprogramming technology, and thus may help make it more available in the clinical fields of regenerative medicine.
Collapse
Affiliation(s)
- Dong-Wook Yang
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Jung-Sun Moon
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Hyun-Mi Ko
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Yeo-Kyeong Shin
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Satoshi Fukumoto
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Sun-Hun Kim
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Min-Seok Kim
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
26
|
Puig-Pijuan T, de Godoy MA, Pinheiro Carvalho LR, Bodart-Santos V, Lindoso RS, Pimentel-Coelho PM, Mendez-Otero R. Human Wharton's jelly mesenchymal stem cells protect neural cells from oxidative stress through paracrine mechanisms. Future Sci OA 2020; 6:FSO627. [PMID: 33235812 PMCID: PMC7668126 DOI: 10.2144/fsoa-2020-0036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aim Mesenchymal stem cells (MSCs) have neuroprotective and immunomodulatory properties, which are partly mediated by extracellular vesicles (EVs) secretion. We aimed to evaluate the effects of human Wharton's jelly-derived MSCs (WJ-MSCs) and their EVs on rat hippocampal cultures subjected to hydrogen peroxide (H2O2). Materials & methods Hippocampal dissociated cultures were either co-cultured with WJ-MSCs or treated with their EVs prior to H2O2 exposure and reactive oxygen species levels and cell viability were evaluated. Results Coculture with WJ-MSCs or pre-incubation with EVs prior to the insult reduced reactive oxygen species after H2O2 exposure. Cell viability was improved only when coculture was maintained following the insult, while EVs did not significantly improve cell viability. Conclusion WJ-MSCs have potential antioxidant and neuroprotective effects on hippocampal cultures which might be partially mediated by EVs.
Collapse
Affiliation(s)
- Teresa Puig-Pijuan
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Mariana A de Godoy
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Luiza Rachel Pinheiro Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Victor Bodart-Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Rafael Soares Lindoso
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil.,Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Moreno Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Guo M, Wu L, Song Z, Yang B. Enhancement of Neural Stem Cell Proliferation in Rats with Spinal Cord Injury by a Combination of Repetitive Transcranial Magnetic Stimulation (rTMS) and Human Umbilical Cord Blood Mesenchymal Stem Cells (hUCB-MSCs). Med Sci Monit 2020; 26:e924445. [PMID: 32814758 PMCID: PMC7453755 DOI: 10.12659/msm.924445] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND This study was designed to explore the combined effects of repetitive transcranial magnetic stimulation (rTMS) and human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) transplantation on neural stem cell proliferation in rats with spinal cord injury (SCI). MATERIAL AND METHODS SCI was induced in 90 rats by laminectomy at T10. Fifteen rats each were treated with 0.5 Hz rTMS or 10 Hz rTMS or underwent hUCB-MSC transplantation; 15 each were treated with 0.5 Hz rTMS+hUCB-MSCs or 10 Hz rTMS+hUCB-MSCs; and 15 were untreated (control group). The Basso, Beattie, and Bresnahan (BBB) scores and motor evoked potentials (MEPs) were measured, and all rats underwent biotin dextran-amine (BDA) tracing of the corticospinal tract (CST). The levels of expression of neural stem cell proliferation related proteins, including BrdU, nestin, Tuj1, Ng2+ and GFAP, were measured, and the levels of bFGF and EGF determined by Western blotting. RESULTS BBB scores and MEPs were increased after rTMS and hUCB-MSC transplantation, while histologically determined SCI-induced neuron apoptosis was attenuated. The numbers of BDA-positive fibers and Brdu-, nestin- and Tuj1-positive cells were markedly increased and the numbers of Ng2+- and GFAP-positive cells were markedly decreased following treatment with rTMS alone or rTMS plus hUCB-MSC transplantation. The levels of expression of bFGF and EGF were significantly upregulated following rTMS treatment and hUCB-MSC transplantation. Higher performance was observed after combined treatment with rTMS and hUCB-MSC transplantation than after either alone. CONCLUSIONS The combination of rTMS treatment and hUCB-MSC transplantation could attenuate SCI-induced neural stem cell apoptosis and motor dysfunction in rats.
Collapse
Affiliation(s)
- Mengguo Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Lixin Wu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Zhenyu Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| |
Collapse
|
28
|
Davies LB, Jones RH, Thornton CA. Maternal Serum, an Isolation and Expansion Tool for Umbilical Cord Matrix Mesenchymal Stromal Cells. Tissue Eng Part C Methods 2020; 25:213-221. [PMID: 30794105 DOI: 10.1089/ten.tec.2019.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The umbilical cord offers a source of readily available mesenchymal stromal cells (MSCs) for use in research and ultimately therapeutic application. However, methods of isolating these cells vary between investigators, and no standard method has been adopted. The aims of this work were to (i) develop a methodology for the isolation of umbilical cord matrix cells without the use of enzymatic digestion or complicated dissection; (ii) investigate the use of pooled maternal serum (MS) as a media supplement; and (iii) demonstrate that the cells isolated were MSCs. We have demonstrated that incubating tissue explants of less than 2 mm3 in serum for an hour, followed by the gradual addition of serum containing culture medium can increase cell yield compared to incubation in serum containing culture medium alone. More importantly, our method demonstrated that the use of pooled serum from women >37 weeks pregnant (pooled MS) yields higher cell numbers than the use of fetal bovine serum or pooled umbilical cord serum. Irrespective of the type of serum used, the isolated cells were MSCs according to the minimal criteria set out by the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy. In conclusion, MS has the potential to be used as an alternative to fetal bovine serum for isolation and expansion of umbilical cord MSCs for clinical purposes.
Collapse
Affiliation(s)
- Lleucu B Davies
- Institute of Life Science, School of Medicine, Swansea University, Swansea, United Kingdom
| | - Ruth H Jones
- Institute of Life Science, School of Medicine, Swansea University, Swansea, United Kingdom
| | - Catherine A Thornton
- Institute of Life Science, School of Medicine, Swansea University, Swansea, United Kingdom
| |
Collapse
|
29
|
Identifying the Therapeutic Significance of Mesenchymal Stem Cells. Cells 2020; 9:cells9051145. [PMID: 32384763 PMCID: PMC7291143 DOI: 10.3390/cells9051145] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
The pleiotropic behavior of mesenchymal stem cells (MSCs) has gained global attention due to their immense potential for immunosuppression and their therapeutic role in immune disorders. MSCs migrate towards inflamed microenvironments, produce anti-inflammatory cytokines and conceal themselves from the innate immune system. These signatures are the reason for the uprising in the sciences of cellular therapy in the last decades. Irrespective of their therapeutic role in immune disorders, some factors limit beneficial effects such as inconsistency of cell characteristics, erratic protocols, deviating dosages, and diverse transfusion patterns. Conclusive protocols for cell culture, differentiation, expansion, and cryopreservation of MSCs are of the utmost importance for a better understanding of MSCs in therapeutic applications. In this review, we address the immunomodulatory properties and immunosuppressive actions of MSCs. Also, we sum up the results of the enhancement, utilization, and therapeutic responses of MSCs in treating inflammatory diseases, metabolic disorders and diabetes.
Collapse
|
30
|
Horie S, Masterson C, Brady J, Loftus P, Horan E, O'Flynn L, Elliman S, Barry F, O'Brien T, Laffey JG, O'Toole D. Umbilical cord-derived CD362 + mesenchymal stromal cells for E. coli pneumonia: impact of dose regimen, passage, cryopreservation, and antibiotic therapy. Stem Cell Res Ther 2020; 11:116. [PMID: 32169108 PMCID: PMC7071745 DOI: 10.1186/s13287-020-01624-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/18/2020] [Accepted: 02/27/2020] [Indexed: 01/31/2023] Open
Abstract
Background Mesenchymal stromal cells (MSCs) demonstrate considerable promise for acute respiratory distress syndrome (ARDS) and sepsis. However, standard approaches to MSC isolation generate highly heterogeneous cell populations, while bone marrow (BM) constitutes a limited and difficult to access MSC source. Furthermore, a range of cell manufacturing considerations and clinical setting practicalities remain to be explored. Methods Adult male rats were subject to E. coli-induced pneumonia and administered CD362+ umbilical cord (UC)-hMSCs using a variety of cell production and clinical relevance considerations. In series 1, animals were instilled with E. coli and randomized to receive heterogeneous BM or UC-hMSCs or CD362+ UC-hMSCs. Subsequent series examined the impact of concomitant antibiotic therapy, MSC therapeutic cryopreservation (cryopreserved vs fresh CD362+ UC-hMSCs), impact of cell passage on efficacy (passages 3 vs 5 vs 7 vs 10), and delay of administration of cell therapy (0 h vs 6 h post-injury vs 6 h + 12 h) following E. coli installation. Results CD362+ UC-hMSCs were as effective as heterogonous MSCs in reducing E. coli-induced acute lung injury, improving oxygenation, decreasing bacterial load, reducing histologic injury, and ameliorating inflammatory marker levels. Cryopreserved CD362+ UC-hMSCs recapitulated this efficacy, attenuating E. coli-induced injury, but therapeutic relevance did not extend beyond passage 3 for all indices. CD362+ UC-hMSCs maintained efficacy in the presence of antibiotic therapy and rescued the animal from E. coli injury when delivered at 6 h + 12 h, following E. coli instillation. Conclusions These translational studies demonstrated the efficacy of CD362+ UC-hMSCs, where they decreased the severity of E. coli-induced pneumonia, maintained efficacy following cryopreservation, were more effective at early passage, were effective in the presence of antibiotic therapy, and could continue to provide benefit at later time points following E. coli injury.
Collapse
Affiliation(s)
- Shahd Horie
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Claire Masterson
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Jack Brady
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Paul Loftus
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Emma Horan
- Orbsen Therapeutics Ltd., Galway, Ireland
| | | | | | - Frank Barry
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland.,Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland.,Medicine, School of Medicine, National University of Ireland, Galway, Ireland
| | - John G Laffey
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Daniel O'Toole
- Anaesthesia, School of Medicine, National University of Ireland, Galway, Ireland. .,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland.
| |
Collapse
|
31
|
Couto PS, Shatirishvili G, Bersenev A, Verter F. First decade of clinical trials and published studies with mesenchymal stromal cells from umbilical cord tissue. Regen Med 2019; 14:309-319. [DOI: 10.2217/rme-2018-0171] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aim: This is the first analysis of both clinical trials and published studies that employ umbilical cord mesenchymal stromal cells, for the decade 2007–2017. Materials & methods: Searching international databases, we found 178 registered trials and 98 publications. Results: Among the registered clinical trials, 20% have resulted in publications so far. Among the publications, 18% report safety and 74% report some form of improvement. Between 36 and 45% of the publications do not report aspects of the cell manufacturing, including isolation method, culture medium or number of culture passages. Conclusion: Analyses that link trials with publications can elucidate factors that promote study completion and publication. More full disclosure of cell manufacturing is needed to evaluate the efficacy of mesenchymal stromal cell isolated from umbilical cord tissue (UC-MSC) products.
Collapse
Affiliation(s)
- Pedro S Couto
- Parent's Guide to Cord Blood Foundation, Brookeville, MD 20833, USA
- Department of Biochemical Engineering, University College London, London WC1E 7JE, UK
| | | | - Alexey Bersenev
- Advanced Cell Therapy Laboratory at Yale – New Haven Hospital, Yale University, New Haven, CT 06520, USA
| | - Frances Verter
- Parent's Guide to Cord Blood Foundation, Brookeville, MD 20833, USA
| |
Collapse
|
32
|
Ma Z, Song G, Liu D, Qian D, Wang Y, Zhou J, Gong J, Meng H, Zhou B, Yang T, Song Z. N-Acetylcysteine enhances the therapeutic efficacy of bone marrow-derived mesenchymal stem cell transplantation in rats with severe acute pancreatitis. Pancreatology 2019; 19:258-265. [PMID: 30660392 DOI: 10.1016/j.pan.2019.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Severe acute pancreatitis (SAP) is a high mortality disease, for which there is a lack of effective therapies. Previous research has demonstrated that bone marrow-derived mesenchymal stem cells (BMSCs), which have immunomodulatory and antioxidant properties, have potential for the treatment of SAP. It remains unclear, however, whether the free radical scavenger N-acetylcysteine (NAC) can enhance the therapeutic efficacy of BMSC transplantation in SAP. In this study, we investigated the effect of combining treatment with NAC and BMSCs in a rat model of SAP. METHODS SAP was induced by injection of sodium taurocholate into the pancreatic duct and, after successful induction of SAP, the rats were treated with BMSCs and NAC, either singly or in combination. RESULTS After 3 days, serum levels of amylase, proinflammatory factors, malondialdehyde, and reactive oxygen species were significantly decreased in animals treated with BMSCs or NAC, compared with vehicle-treated animals. In contrast, total glutathione, superoxide dismutase and catalase were markedly increased after treatment with BMSCs or NAC. However, oxidative stress markers and inflammatory factors were significantly improved in the SAP + BMSCs + NAC group compared with those in the SAP + NAC group and the SAP + BMSCs group. CONCLUSIONS Combined NAC and BMSC therapy was found to alleviate oxidative stress damage to the pancreas and to inhibit the inflammatory response to a significantly greater extent than single therapy with either BMSCs or NAC. Because NAC enhances the therapeutic efficacy of BMSC transplantation in a rat model of SAP, combined therapy may provide a promising new approach for the treatment of SAP.
Collapse
Affiliation(s)
- Zhilong Ma
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Guodong Song
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Dalu Liu
- Shanghai Clinical Medical College of Anhui Medical University, Hefei, 230032, China
| | - Daohai Qian
- Department of Hepatobiliary Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, 241001, China
| | - Yuxiang Wang
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jia Zhou
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jian Gong
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hongbo Meng
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Bo Zhou
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Tingsong Yang
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Zhenshun Song
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
33
|
Rossetti D, Di Angelo Antonio S, Lukanović D, Kunic T, Certelli C, Vascone C, Sleiman Z. Human umbilical cord-derived mesenchymal stem cells: Current trends and future perspectives. ASIAN PACIFIC JOURNAL OF REPRODUCTION 2019. [DOI: 10.4103/2305-0500.259166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
34
|
Bone marrow-derived mesenchymal stromal cells ameliorate severe acute pancreatitis in rats via hemeoxygenase-1-mediated anti-oxidant and anti-inflammatory effects. Cytotherapy 2018; 21:162-174. [PMID: 30600195 DOI: 10.1016/j.jcyt.2018.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 11/09/2018] [Accepted: 11/20/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND AIMS It has been previously verified that mesenchymal stromal cells (MSCs) have a good therapeutic effect on severe acute pancreatitis (SAP) and the potential for regeneration of damaged pancreatic tissue, but the exact molecular mechanism remains unclear. In this study, we demonstrated the therapeutic effect of bone morrow MSCs (BMSCs) on SAP, probably by targeting heme oxygenase-1 (HO-1). METHODS Six hours after SAP induction, either phosphate-buffered saline (PBS) or BMSCs were transfused into the caudal vein of rats, zinc protoporphyrin (ZnPP) was administered intraperitoneally. Pancreatic pathological scoring, serum levels of amylase and inflammatory factors, as well as levels of reactive oxygen species (ROS), malondialdehyde (MDA) and myeloperoxidase (MPO), superoxide dismutase (SOD) and catalase (CAT) activity in the pancreas were evaluated. RESULTS Our data showed that BMSCs significantly reduce inflammation and oxidative stress, reduce apoptosis and promote angiogenesis of damaged pancreas. Moreover, BMSCs increased the level of HO-1 in the serum and pancreatic tissue in rats with SAP. In addition, the protective effect of BMSCs was partially neutralized by the HO-1 activity inhibitor ZnPP, suggesting a key role of HO-1 in the therapeutic effect of BMSCs on SAP. CONCLUSIONS BMSCs ameliorated SAP, probably by inducing expression of HO-1, which can exert anti-inflammatory and anti-oxidant effects, reduce apoptosis and promote angiogenesis.
Collapse
|
35
|
Mu Y, Wu X, Hao Z. Comparative evaluation of mesenchymal stromal cells from umbilical cord and amniotic membrane in xeno-free conditions. BMC Cell Biol 2018; 19:27. [PMID: 30545286 PMCID: PMC6293527 DOI: 10.1186/s12860-018-0178-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 11/28/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Within the past years, umbilical cord (UC) and amniotic membrane (AM) expanded in human platelet lysate (PL) have been found to become increasingly candidate of mesenchymal stromal cells (MSCs) in preclinical and clinical studies. Different sources of MSCs have different properties, and lead to different therapeutic applications. However, the similarity and differences between the AMMSCs and UCMSCs in PL remain unclear. RESULTS In this study, we conduct a direct head-to-head comparison with regard to biological characteristics (morphology, immunophenotype, self-renewal capacity, and trilineage differentiation potential) and immunosuppression effects of AMMSCs and UCMSCs expanded in PL. Our results indicated that AMMSCs showed similar morphology, immunophenotype, proliferative capacity and colony efficiency with UCMSCs. Moreover, no significantly differences in osteogenic, chondrogenic and adipogenic differentiation potential were observed between the two types of cells. However, AMMSCs exhibited higher PGE2 expression and IDO activity compared with UCMSCs when primed by IFN-γ and (or) TNF-α induction, and AMMSCs showed a higher inhibitory effect on PBMCs proliferation than UCMSCs. CONCLUSION The results suggest that AMMSCs expanded in PL showed similar morphology, immunophenotype, self-renewal capacity, and trilineage differentiation potential with UCMSCs. However, AMMSCs possessed superior immunosuppression effects in comparison with UCMSCs. These results suggest that AMMSCs in PL might be more suitable than UCMSCs for treatment of immune diseases. This work provides a novel insight into choosing the appropriate source of MSCs for treatment of immune diseases.
Collapse
Affiliation(s)
- Yongxu Mu
- Department of Rheumatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Pvovince, China.,Department of Interventional Treatment, the First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Xiaoyun Wu
- Department of Technology, Stem Cell Medicine Engineering & Technology Research Center of Inner Mongolia, Huhhot, Inner Mongolia, China.,Department of Research and Development, Beijing Jingmeng Stem Cell Technology. Co. Ltd., Beijing, China
| | - Zhiming Hao
- Department of Rheumatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Pvovince, China.
| |
Collapse
|
36
|
Balgi-Agarwal S, Winter C, Corral A, Mustafa SB, Hornsby P, Moreira A. Comparison of Preterm and Term Wharton's Jelly-Derived Mesenchymal Stem Cell Properties in Different Oxygen Tensions. Cells Tissues Organs 2018; 205:137-150. [PMID: 29949803 PMCID: PMC6117836 DOI: 10.1159/000489256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 04/15/2018] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have shown promise as therapeutic agents in treating morbidities associated with premature birth. MSCs derived from the human umbilical cord are easy to isolate and have low immunogenicity and a robust ability to secrete paracrine factors. To date, there are no studies evaluating preterm versus term umbilical cord tissue-derived MSCs. Therefore, our aim was twofold: (1) to compare stem cell properties in preterm versus term MSCs and (2) to examine the impact of oxygen tension on stem cell behavior. Umbilical cord tissue was obtained from 5 preterm and 5 term neonates. The cells were isolated and characterized as MSCs in accordance with the International Society for Cellular Therapy. We exposed MSCs to different oxygen tensions to examine the impact of environmental factors on cell performance. We studied the following stem cell properties: (i) motility, (ii) proliferation, (iii) senescence, (iv) cell viability, (v) colony-forming unit efficiency, and (vi) inflammatory cytokine expression. Under normoxia (21% O2), cells from preterm and term infants had similar properties. Under hypoxic conditions (1% O2), term MSCs had better cell proliferation; however, cells exposed to hyperoxia (90% O2) had the slowest motility and lowest cell viability (p < 0.05). There was no difference in the expression of senescence or cytokine expression between the groups. The term cells demonstrated more colony-forming efficiency than the preterm cells. In sum, our preliminary findings suggest that MSCs derived from term and preterm umbilical cords have similar characteristics, offering the potential of future autologous/allogeneic MSC transplants in neonates.
Collapse
Affiliation(s)
- Saloni Balgi-Agarwal
- Division of Neonatology MC-7812, Department of Pediatrics, University of Texas Health-San Antonio, San Antonio, Texas, USA
| | - Caitlyn Winter
- Division of Neonatology MC-7812, Department of Pediatrics, University of Texas Health-San Antonio, San Antonio, Texas, USA
| | - Alexis Corral
- Division of Neonatology MC-7812, Department of Pediatrics, University of Texas Health-San Antonio, San Antonio, Texas, USA
| | - Shamimunisa B Mustafa
- Division of Neonatology MC-7812, Department of Pediatrics, University of Texas Health-San Antonio, San Antonio, Texas, USA
| | - Peter Hornsby
- Department of Cellular and Integrative Physiology, University of Texas Health-San Antonio, San Antonio, Texas, USA
| | - Alvaro Moreira
- Division of Neonatology MC-7812, Department of Pediatrics, University of Texas Health-San Antonio, San Antonio, Texas, USA
| |
Collapse
|
37
|
Skiles ML, Brown KS, Tatz W, Swingle K, Brown HL. Quantitative analysis of composite umbilical cord tissue health using a standardized explant approach and an assay of metabolic activity. Cytotherapy 2018; 20:564-575. [PMID: 29429941 DOI: 10.1016/j.jcyt.2018.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 12/07/2017] [Accepted: 01/04/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Umbilical cord (UC) tissue can be collected in a noninvasive procedure and is enriched in progenitor cells with potential therapeutic value. Mesenchymal stromal cells (MSCs) can be reliably harvested from fresh or cryopreserved UC tissue by explant outgrowth with no apparent impact on functionality. A number of stem cell banks offer cryopreservation of UC tissue, alongside cord blood, for future cell-based applications. In this setting, measuring and monitoring UC quality is critical. MATERIALS AND METHODS UC explants were evaluated using a plating and scoring system accounting for cell attachment and proliferation. Explant scores for fresh and cryopreserved-then-thawed tissue from the same UC were compared. Metabolic activity of composite UC tissue was also assayed after exposure of the tissue to conditions anticipated to affect UC quality and compared with explant scores within the same UC. RESULTS All fresh and cryopreserved tissues yielded MSC-like cells, and cryopreservation of the tissue did not prevent the ability to isolate MSCs by the explant method. Thawed UC tissue scores were 91% (±0.6%; P = 0.0009) that of the fresh, biologically identical tissue. Within the same UC, explant scores correlated well to both cell yield (R2 = 0.85) and tissue metabolic activity (R2 = 0.69). DISCUSSION A uniform explant scoring assay can provide information about the quality of composite UC tissue. Such quantitative measurement is useful for analysis of tissue variability and process monitoring. Additionally, a metabolic assay of UC tissue health provides results that correlate well to explant scoring results.
Collapse
Affiliation(s)
- Matthew L Skiles
- Scientific and Medical Affairs, Cbr Systems, Inc., South San Francisco, California, USA.
| | - Katherine S Brown
- Scientific and Medical Affairs, Cbr Systems, Inc., South San Francisco, California, USA
| | - William Tatz
- Laboratory Operations, Cbr Systems, Inc., Tucson, Arizona, USA
| | - Kristen Swingle
- Consumer Sales and Operations, Cbr Systems, Inc., Tucson, Arizona, USA
| | - Heather L Brown
- Scientific and Medical Affairs, Cbr Systems, Inc., South San Francisco, California, USA
| |
Collapse
|
38
|
Zhang L, Li Y, Guan CY, Tian S, Lv XD, Li JH, Ma X, Xia HF. Therapeutic effect of human umbilical cord-derived mesenchymal stem cells on injured rat endometrium during its chronic phase. Stem Cell Res Ther 2018; 9:36. [PMID: 29433563 PMCID: PMC5810045 DOI: 10.1186/s13287-018-0777-5] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/04/2017] [Accepted: 01/15/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Repair deficiency after endometrial injury is an important reason for intra-uterine adhesions, amenorrhea, and infertility in females. Bone marrow-derived mesenchymal stem cell (BMSC) transplantation is effective in repairing the damaged endometrium. However, the possibility of using umbilical cord-derived MSCs (UC-MSCs) to treat endometrial injury is rarely reported. METHODS Ethanol (95%) was injected into rat uterus to establish a model of endometrial injury. UC-MSCs were injected through the tail vein, either as a single, twice, or thrice administration. Functional restoration of the uterus was assessed by testing embryo implantation rates. Endometrial morphological alteration was observed by hematoxylin and eosin staining. Endometrial fibrosis, markers of epithelial and stromal cells of endometrium, cell proliferation and angiogenesis, and inflammatory factors were detected using immunohistochemistry, Western blotting, and quantitative reverse-transcription polymerase chain reaction. RESULTS Endometrial morphology and embryo implantation rates were significantly improved on day 8 of transplantation among single-, twice-, or thrice-administered rats. Moreover, UC-MSCs could alleviate fibrosis in general, and reduced the expression of fibrosis markers, α-smooth muscle actin (α-SMA) and transforming growth factor (TGF)-β. The cell proliferation marker Ki-67 had a positive expression in the injured endometrium after UC-MSC transplantation. The endometrial stromal marker vimentin and epithelial marker cytokeratin-19 (CK-19) expressions were visibly increased. The expression of vascular markers CD31, vascular endothelial growth factor (VEGF)A, and matrix metalloprotein (MMP)9 was generally upregulated. Proinflammatory factors interferon (IFN)-γ, tumor necrosis factor (TNF)-α, and interleukin (IL)-2 were significantly downregulated in the rats administered UC-MSCs twice and thrice. CONCLUSIONS UC-MSC transplantation contributed to the repair of endometrial injury and restoration of fertility, likely through the suppression of excessive fibrosis and inflammation, and enhancement of endometrial cell proliferation and vascular remodeling.
Collapse
Affiliation(s)
- Lu Zhang
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China.,Graduate School, Peking Union Medical College, Beijing, China
| | - Ying Li
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China.,Graduate School, Peking Union Medical College, Beijing, China
| | - Chun-Yi Guan
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China.,Graduate School, Peking Union Medical College, Beijing, China
| | - Shi Tian
- Haidian Maternal & Child Health Hospital, Beijing, China
| | - Xiao-Dan Lv
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China.,Graduate School, Peking Union Medical College, Beijing, China
| | - Jian-Hui Li
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China.,Graduate School, Peking Union Medical College, Beijing, China
| | - Xu Ma
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China. .,Graduate School, Peking Union Medical College, Beijing, China.
| | - Hong-Fei Xia
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China. .,Graduate School, Peking Union Medical College, Beijing, China.
| |
Collapse
|
39
|
Jia Z, Liang Y, Xu X, Li X, Liu Q, Ou Y, Duan L, Zhu W, Lu W, Xiong J, Wang D. Isolation and characterization of human mesenchymal stem cells derived from synovial fluid by magnetic-activated cell sorting (MACS). Cell Biol Int 2017; 42:262-271. [PMID: 29068101 DOI: 10.1002/cbin.10903] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 10/22/2017] [Indexed: 12/30/2022]
Abstract
Mesenchymal stem cells (MSCs) are the primary source of cells used for cell-based therapy in tissue engineering. MSCs are found in synovial fluid, a source that could be conveniently used for cartilage tissue engineering. However, the purification and characterization of SF-MSCs has been poorly documented in the literature. Here, we outline an easy-to-perform approach for the isolation and culture of MSCs derived from human synovial fluid (hSF-MSCs). We have successfully purified hSF-MSCs using magnetic-activated cell sorting (MACS) using the MSC surface marker, CD90. Purified SF-MSCs demonstrate significant renewal capacity following several passages in culture. Furthermore, we demonstrated that MACS-sorted CD90+ cells could differentiated into osteoblasts, adipocytes, and chondrocytes in vitro. In addition, we show that these cells can generate cartilage tissue in micromass culture as well. This study demonstrates that MACS is a useful tool that can be used for the purification of hSF-MSCs from synovial fluid. The proliferation properties and ability to differentiate into chondrocytes make these hSF-MSCs a promising source of stem cells for applications in cartilage repair.
Collapse
Affiliation(s)
- Zhaofeng Jia
- Guangzhou Medical University, Guangzhou 510095, Guangdong Province, China.,Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| | - Yujie Liang
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.,Shenzhen Key Laboratory for Psychological Healthcare, Shenzhen Institute of Mental Health, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong Province, China
| | - Xiao Xu
- Guangzhou Medical University, Guangzhou 510095, Guangdong Province, China.,Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| | - Xingfu Li
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| | - Qisong Liu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| | - Yangkan Ou
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| | - Li Duan
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| | - Weimin Zhu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| | - Wei Lu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| | - Jianyi Xiong
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| | - Daping Wang
- Guangzhou Medical University, Guangzhou 510095, Guangdong Province, China.,Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, Guangdong Province, China.,Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen 518035, Guangdong Province, China
| |
Collapse
|
40
|
Zhang B, Zhang J, Shi H, Mao F, Wang J, Yan Y, Zhang X, Qian H, Xu W. A novel method to isolate mesenchymal stem cells from mouse umbilical cord. Mol Med Rep 2017; 17:861-869. [PMID: 29115623 PMCID: PMC5780165 DOI: 10.3892/mmr.2017.7950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 05/11/2017] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs), derived from various tissues, are considered an ideal cell source for clinical use, among which MSCs from the umbilical cord exhibit advantages over those from adult tissues. In preclinical studies, mouse models and xenogeneic MSC treatment are most commonly used to imitate diseases and clinical practice, respectively. However, the efficiency of cross-species therapy remains controversial, making it difficult to elucidate the underlying mechanisms. Thus, allogeneic therapy may be more instructive and meaningful in clinical use. To confirm this hypothesis, the present study established a novel method for the isolation and expansion of MSCs from mouse umbilical cords (mUC-MSCs) to support in vivo experiments in mice. MSCs were isolated from mUCs and mouse bone marrow (mBM), and then identified by flow cytometry. The differences in mUC-MSCs and mBM-MSCs were analyzed using a growth curve and their differentiation ability. The results showed that the harvested cells exhibited general characteristics of MSCs and possessed the capacity for long-term culture. Despite having similar morphology and surface antigens to MSCs derived from mouse bone marrow, the mUC-MSCs showed differences in purification, proliferation, stem cell markers and differentiation. In addition to detailed characterization, the present study verified the presence of Toll-like receptor 3 (TLR3), an important component of immune responses, in mUC-MSCs. It was found that the activation of TLR3 upregulated the levels of stemness-related proteins, and enhanced the secretion and mRNA levels of inflammatory cytokines in the pre-treated mUC-MSCs. Collectively, the results of the present study provide further insight into the features of newly established mUC-MSCs, providing novel evidence for the selection of murine MSCs and their responses to TLR3 priming.
Collapse
Affiliation(s)
- Bin Zhang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jie Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Shi
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Juanjuan Wang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yongmin Yan
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hui Qian
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenrong Xu
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
41
|
Cryopreserved, Xeno-Free Human Umbilical Cord Mesenchymal Stromal Cells Reduce Lung Injury Severity and Bacterial Burden in Rodent Escherichia coli-Induced Acute Respiratory Distress Syndrome. Crit Care Med 2017; 45:e202-e212. [PMID: 27861182 DOI: 10.1097/ccm.0000000000002073] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Although mesenchymal stem/stromal cells represent a promising therapeutic strategy for acute respiratory distress syndrome, clinical translation faces challenges, including scarcity of bone marrow donors, and reliance on bovine serum during mesenchymal stem/stromal cell proliferation. We wished to compare mesenchymal stem/stromal cells from human umbilical cord, grown in xeno-free conditions, with mesenchymal stem/stromal cells from human bone marrow, in a rat model of Escherichia coli pneumonia. In addition, we wished to determine the potential for umbilical cord-mesenchymal stem/stromal cells to reduce E. coli-induced oxidant injury. DESIGN Randomized animal study. SETTING University research laboratory. SUBJECTS Male Sprague-Dawley rats. INTERVENTIONS Acute respiratory distress syndrome was induced in rats by intratracheal instillation of E. coli (1.5-2 × 10 CFU/kg). "Series 1" compared the effects of freshly thawed cryopreserved umbilical cord-mesenchymal stem/stromal cells with bone marrow-mesenchymal stem/stromal cells on physiologic indices of lung injury, cellular infiltration, and E. coli colony counts in bronchoalveolar lavage. "Series 2" examined the effects of cryopreserved umbilical cord-mesenchymal stem/stromal cells on survival, as well as measures of injury, inflammation and oxidant stress, including production of reactive oxidative species, reactive oxidative species scavenging by superoxide dismutase-1 and superoxide dismutase-2. MEASUREMENTS AND MAIN RESULTS In "Series 1," animals subjected to E. coli pneumonia who received umbilical cord-mesenchymal stem/stromal cells had improvements in oxygenation, respiratory static compliance, and wet-to-dry ratios comparable to bone marrow-mesenchymal stem/stromal cell treatment. E. coli colony-forming units in bronchoalveolar lavage were reduced in both cell therapy groups, despite a reduction in bronchoalveolar lavage neutrophils. In series 2, umbilical cord-mesenchymal stem/stromal cells enhanced animal survival and decreased alveolar protein and proinflammatory cytokine concentrations, whereas increasing interleukin-10 concentrations. Umbilical cord-mesenchymal stem/stromal cell therapy decreased nicotinamide adenine dinucleotide phosphate-oxidase 2 and inducible nitric oxide synthase and enhanced lung concentrations of superoxide dismutase-2, thereby reducing lung tissue reactive oxidative species concentrations. CONCLUSIONS Our results demonstrate that freshly thawed cryopreserved xeno-free human umbilical cord-mesenchymal stem/stromal cells reduce the severity of rodent E. coli-induced acute respiratory distress syndrome. Umbilical cord-mesenchymal stem/stromal cells, therefore, represent an attractive option for future clinical trials in acute respiratory distress syndrome.
Collapse
|
42
|
Umbilical cord tissue–derived mesenchymal stromal cells maintain immunomodulatory and angiogenic potencies after cryopreservation and subsequent thawing. Cytotherapy 2017; 19:360-370. [DOI: 10.1016/j.jcyt.2016.11.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/26/2016] [Accepted: 11/08/2016] [Indexed: 12/22/2022]
|
43
|
Cao Y, Gang X, Sun C, Wang G. Mesenchymal Stem Cells Improve Healing of Diabetic Foot Ulcer. J Diabetes Res 2017; 2017:9328347. [PMID: 28386568 PMCID: PMC5366201 DOI: 10.1155/2017/9328347] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 02/28/2017] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs), an ideal cell source for regenerative therapy with no ethical issues, play an important role in diabetic foot ulcer (DFU). Growing evidence has demonstrated that MSCs transplantation can accelerate wound closure, ameliorate clinical parameters, and avoid amputation. In this review, we clarify the mechanism of preclinical studies, as well as safety and efficacy of clinical trials in the treatment of DFU. Bone marrow-derived mesenchymal stem cells (BM-MSCs), compared with MSCs derived from other tissues, may be a suitable cell type that can provide easy, effective, and cost-efficient transplantation to treat DFU and protect patients from amputation.
Collapse
Affiliation(s)
- Yue Cao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Chenglin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
- *Guixia Wang:
| |
Collapse
|
44
|
de Soure AM, Fernandes-Platzgummer A, Moreira F, Lilaia C, Liu SH, Ku CP, Huang YF, Milligan W, Cabral JMS, da Silva CL. Integrated culture platform based on a human platelet lysate supplement for the isolation and scalable manufacturing of umbilical cord matrix-derived mesenchymal stem/stromal cells. J Tissue Eng Regen Med 2016; 11:1630-1640. [DOI: 10.1002/term.2200] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/29/2016] [Accepted: 03/14/2016] [Indexed: 12/16/2022]
Affiliation(s)
- António M. de Soure
- Department of Bioengineering and iBB-; Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa; Lisboa Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and iBB-; Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa; Lisboa Portugal
| | - Francisco Moreira
- Department of Bioengineering and iBB-; Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa; Lisboa Portugal
| | - Carla Lilaia
- Hospital São Francisco Xavier, Centro Hospitalar Lisboa Ocidental; Lisboa Portugal
| | - Shi-Hwei Liu
- R&D, AventaCell BioMedical Co., Ltd.; New Taipei City Taiwan
| | - Chen-Peng Ku
- R&D, AventaCell BioMedical Co., Ltd.; New Taipei City Taiwan
- R&D, AventaCell BioMedical Co., Ltd; Atlanta GA USA
| | - Yi-Feng Huang
- R&D, AventaCell BioMedical Co., Ltd.; New Taipei City Taiwan
- R&D, AventaCell BioMedical Co., Ltd; Atlanta GA USA
| | - William Milligan
- R&D, AventaCell BioMedical Co., Ltd.; New Taipei City Taiwan
- R&D, AventaCell BioMedical Co., Ltd; Atlanta GA USA
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB-; Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa; Lisboa Portugal
| | - Cláudia L. da Silva
- Department of Bioengineering and iBB-; Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa; Lisboa Portugal
| |
Collapse
|
45
|
Meng Y, Li X, Li Z, Liu C, Zhao J, Wang J, Liu Y, Yuan X, Cui Z, Yang X. Surface Functionalization of Titanium Alloy with miR-29b Nanocapsules To Enhance Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2016; 8:5783-5793. [PMID: 26887789 DOI: 10.1021/acsami.5b10650] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Titanium and its alloys have been widely used over the past 3 decades as implants for healing bone defects. Nevertheless, the bioinert property of titanium alloy limits its clinical application and surface modification method is frequently performed to improve the biological and chemical properties. Recently, the delivery of microRNA with osteogenesis capability has been recognized as a promising tool to enhance bone regeneration of implants. Here, we developed a biodegradable coating to modify the titanium surface in order to enhance osteogenic bioactivity. The previous developed nanocapsules were used as the building blocks, and then a bioactive titanium coating was designed to entrap the miR-29b nanocapsules. This coating was not only favorable for cell adhesion and growth but also provided sufficient microRNA transfection efficacy and osteoinductive potential, resulting in a significant enhancement of bone regeneration on the surface of bioinert titanium alloy.
Collapse
Affiliation(s)
- Yubin Meng
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| | - Xue Li
- Department of Clinical Microbiology, School of Laboratory Medicine, Tianjin Medical University , Tianjin 300203, China
| | - Zhaoyang Li
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| | - Chaoyong Liu
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| | - Jin Zhao
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| | - Jianwei Wang
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| | - Yunde Liu
- Department of Clinical Microbiology, School of Laboratory Medicine, Tianjin Medical University , Tianjin 300203, China
| | - Xubo Yuan
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| | - Zhenduo Cui
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| | - Xianjin Yang
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| |
Collapse
|
46
|
Evaluation of Tissue Homogenization to Support the Generation of GMP-Compliant Mesenchymal Stromal Cells from the Umbilical Cord. Stem Cells Int 2016; 2016:3274054. [PMID: 27034683 PMCID: PMC4806688 DOI: 10.1155/2016/3274054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/02/2015] [Accepted: 12/14/2015] [Indexed: 02/06/2023] Open
Abstract
Recent studies have demonstrated that the umbilical cord (UC) is an excellent source of mesenchymal stromal cells (MSCs). However, current protocols for extracting and culturing UC-MSCs do not meet current good manufacturing practice (cGMP) standards, in part due to the use of xenogeneic reagents. To support the development of a cGMP-compliant method, we have examined an enzyme-free isolation method utilizing tissue homogenization (t-H) followed by culture in human platelet lysate (PL) supplemented media. The yield and viability of cells after t-H were comparable to those obtained after collagenase digestion (Col-D). Importantly, kinetic analysis of cultured cells showed logarithmic growth over 10 tested passages, although the rate of cell division was lower for t-H as compared to Col-D. This slower growth of t-H-derived cells was also reflected in their longer population doubling time. Interestingly, there was no difference in the expression of mesenchymal markers and trilineage differentiation potential of cells generated using either method. Finally, t-H-derived cells had greater clonogenic potential compared to Col-D/FBS but not Col-D/PL and were able to maintain CFU-F capacity through P7. This bench scale study demonstrates the possibility of generating therapeutic doses of good quality UC-MSCs within a reasonable length of time using t-H and PL.
Collapse
|
47
|
Arutyunyan I, Elchaninov A, Makarov A, Fatkhudinov T. Umbilical Cord as Prospective Source for Mesenchymal Stem Cell-Based Therapy. Stem Cells Int 2016; 2016:6901286. [PMID: 27651799 PMCID: PMC5019943 DOI: 10.1155/2016/6901286] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/14/2016] [Indexed: 02/07/2023] Open
Abstract
The paper presents current evidence on the properties of human umbilical cord-derived mesenchymal stem cells, including origin, proliferative potential, plasticity, stability of karyotype and phenotype, transcriptome, secretome, and immunomodulatory activity. A review of preclinical studies and clinical trials using this cell type is performed. Prospects for the use of mesenchymal stem cells, derived from the umbilical cord, in cell transplantation are associated with the need for specialized biobanking and transplant standardization criteria.
Collapse
Affiliation(s)
- Irina Arutyunyan
- 1Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, No. 4, Oparin Street, Moscow 117997, Russia
| | - Andrey Elchaninov
- 2Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, No. 1, Ostrovitianov Street, Moscow 117997, Russia
| | - Andrey Makarov
- 1Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, No. 4, Oparin Street, Moscow 117997, Russia
| | - Timur Fatkhudinov
- 1Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, No. 4, Oparin Street, Moscow 117997, Russia
- 2Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, No. 1, Ostrovitianov Street, Moscow 117997, Russia
- *Timur Fatkhudinov:
| |
Collapse
|
48
|
Kavanagh JN, Waring EJ, Prise KM. Radiation responses of stem cells: targeted and non-targeted effects. RADIATION PROTECTION DOSIMETRY 2015; 166:110-117. [PMID: 25877536 DOI: 10.1093/rpd/ncv161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Stem cells are fundamental to the development of any tissue or organism via their ability to self-renew, which is aided by their unlimited proliferative capacity and their ability to produce fully differentiated offspring, often from multiple lineages. Stems cells are long lived and have the potential to accumulate mutations, including in response to radiation exposure. It is thought that stem cells have the potential to be induced into a cancer stem cell phenotype and that these may play an important role in resistance to radiotherapy. For radiation-induced carcinogenesis, the role of targeted and non-targeted effects is unclear with tissue or origin being important. Studies of genomic instability and bystander responses have shown consistent effects in haematopoietic models. Several models of radiation have predicted that stem cells play an important role in tumour initiation and that bystander responses could play a role in proliferation and self-renewal.
Collapse
Affiliation(s)
- J N Kavanagh
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - E J Waring
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - K M Prise
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| |
Collapse
|
49
|
Meng YB, Li X, Li ZY, Zhao J, Yuan XB, Ren Y, Cui ZD, Liu YD, Yang XJ. microRNA-21 promotes osteogenic differentiation of mesenchymal stem cells by the PI3K/β-catenin pathway. J Orthop Res 2015; 33:957-64. [PMID: 25728838 DOI: 10.1002/jor.22884] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 02/24/2015] [Indexed: 02/04/2023]
Abstract
Osteogenesis of mesenchymal stem cells (MSCs) is essential for bone repair. Recently, microRNAs have been proven to play an important role in the regulation of MSC differentiation, including osteogenesis. Here, the function of microRNA-21 (miR-21) in the osteogenic differentiation of human umbilical cord mesenchymal stem cells (hUMSCs) was investigated. Briefly, the miR-21 mimics (m-miR-21) and the antisense miR-21 (as-miR-21) were transfected to hUMSCs, and the capacity of miR-21 for the osteogenic differentiation of hUMSCs was evaluated by the expression of osteogenic markers encoding alkaline phosphatase (ALP), runt-related gene-2 (RUNX-2) and osteocalcin (OCN), as well as by Alizarin red S staining. The results indicated that the overexpression of miR-21 elevated the expression level of the osteogenesis-related genes of hUMSCs. During this process, the PI3K-AKT signaling pathway activity had an increasing tendency responding to miR-21 up-regulation. This enhancement promoted the phosphorylation of GSK-3β, leading to the stabilization and high concentration accumulation of β-catenin in cytoplasm to activate the transcription of RUNX-2, and finally increased the osteogenesis of hUMSCs. This work demonstrated that miR-21 and its target PI3K-AKT-GSK3β pathway played an important role in the osteogenic differentiation of hUMSCs by stabilizing β-catenin.
Collapse
Affiliation(s)
- Yu-Bin Meng
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, P.R. China
| | - Xue Li
- Department of Clinical Microbiology, School of Laboratory Medicine, Tianjin Medical University, Tianjin, 300203, P.R. China
| | - Zhao-Yang Li
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, P.R. China
| | - Jin Zhao
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, P.R. China
| | - Xu-Bo Yuan
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, P.R. China
| | - Yu Ren
- Research Center of Basic Medical Sciences & Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin Medical University, Tianjin, 300070, P.R. China
| | - Zhen-Duo Cui
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, P.R. China
| | - Yun-De Liu
- Department of Clinical Microbiology, School of Laboratory Medicine, Tianjin Medical University, Tianjin, 300203, P.R. China
| | - Xian-Jin Yang
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, P.R. China
| |
Collapse
|
50
|
Olson AL, McNiece IK. Novel clinical uses for cord blood derived mesenchymal stromal cells. Cytotherapy 2015; 17:796-802. [PMID: 25819838 DOI: 10.1016/j.jcyt.2015.03.612] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/02/2015] [Indexed: 12/16/2022]
Abstract
Regenerative medicine offers new hope for many debilitating diseases that result in damage to tissues and organs. The concept is straightforward with replacement of damaged cells with new functional cells. However, most tissues and organs are complex structures involving multiple cell types, supportive structures, a microenvironment producing cytokines and growth factors and a vascular system to supply oxygen and other nutrients. Therefore repair, particularly in the setting of ischemic damage, may require delivery of multiple cell types providing new vessel formation, a new microenvironment and functional cells. The field of stem cell biology has identified a number of stem cell sources including embryonic stem cells and adult stem cells that offer the potential to replace virtually all functional cells of the body. The focus of this article is a discussion of the potential of mesenchymal stromal cells (MSCs) from cord blood (CB) for regenerative medicine approaches.
Collapse
Affiliation(s)
- Amanda L Olson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Ian K McNiece
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|