1
|
Xiao J, Zhang Q, Wu B, Wang M, Zhu Y, Zhao D, Zhao F, Xie Y. Effect of placental mesenchymal stem cells on promoting the healing of chronic burn wounds. Heliyon 2024; 10:e36584. [PMID: 39281490 PMCID: PMC11401119 DOI: 10.1016/j.heliyon.2024.e36584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
The treatment of chronic burn wounds is difficult in clinical practice. The ideal therapy is required to be continuously explored. Mesenchymal stem cells revolutionize the treatment of many diseases. The placental mesenchymal stem cells (PMSCs) have the characteristics of easy access, strong proliferation ability and multi-directional differentiation potential. The aim of this study was to investigate the potential of PMSCs in chronic burn wound healing. In this study, species of bacteria of 317 patients with chronic burn wounds have been analyzed. Samples of chronic burn wound fluid were collected from representative patients and then co-cultured with cells. In vitro studies showed that chronic burn wound fluid inhibited the proliferation of human keratinocytes and fibroblasts, while PMSCs can counteract the effects of burn wound fluid on inhibiting the proliferation and migration of human keratinocytes and fibroblasts. In addition, in vivo studies showed that a rat chronic burn wound model was successfully created. The expression of MMP-2, MMP-9, MDA, IL-6 and TNF-α in chronic burn wounds was significantly higher than that in acute burn wounds. Finally, the rat chronic burn wound model was used to verify that placental mesenchymal stem cell transplantation increased the wound healing rate, decreased the wound healing time, and promoted wound healing by increasing the thickness of epidermis and promoting the expression of P63 and CK10. The findings provide support for the hypothesis that PMSCs promote the repair of chronic burn wounds and key scientific data for the application of PMSCs as a new method for treating chronic burn wounds.
Collapse
Affiliation(s)
- Jinli Xiao
- Clinical Medical School, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Qing Zhang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Bowen Wu
- Clinical Medical School, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Maomao Wang
- Clinical Medical School, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yongzhao Zhu
- Surgery Lab, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Dan Zhao
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Fang Zhao
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yan Xie
- Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
2
|
Kang M, Yum HY, Kim HT, Park BJ, Cho DS, Choi Y, Kim HJ, Cho Y, Kim YJ, Lee DM, Lee DG, Song HC, Nam SH, Lee JH, Choi BO, Kim SW. Self-Powered Electrical Bandage Based on Body-Coupled Energy Harvesting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402491. [PMID: 38837481 DOI: 10.1002/adma.202402491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/06/2024] [Indexed: 06/07/2024]
Abstract
Self-powered electrical bandages (SEBs), integrated with wearable energy harvesters, can provide an effective and autonomous electrical stimulation (ES) solution for rapid and scarless wound healing. A continuously operating, wireless, and applicable-to-comprehensive-wound ES device is essential for the quick restoration of wounds and convenience. This work illustrates a SEB powered by body-coupled energy harvesting. The SEB continuously treats the wound with 60-Hz sinusoidal electrical potential gained from the coupling of the human body and ambient electrical waves. It is demonstrated that enough level of electrical potential can be applied to the wound, further enhanced by strong capacitive coupling arising from the use of high-permittivity poly(vinylidene fluoride-trifluoroethylene):CaCu3Ti4O12 (P(VDF-TrFE):CCTO) nanocomposite. The potential clinical efficacy of the SEB is illustrated by preclinical analysis of human fibroblasts and mouse wound model, thus confirming the successful expedition of wound recovery. This work suggests a new class of wearable devices to provide ES events and its potential for extension to other conventional wound care materials and device technology.
Collapse
Affiliation(s)
- Minki Kang
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Han-Yup Yum
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyoung Taek Kim
- Department of Research and Development, SEMS Co. Ltd, Suwon, 16229, Republic of Korea
| | - Byung-Joon Park
- Department of Materials Science and Engineering, Center for Human-oriented Triboelectric Energy Harvesting, Yonsei University, Seoul, 03722, Republic of Korea
| | - Daniel Sanghyun Cho
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - YoungHwan Choi
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06355, Republic of Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, 06355, Republic of Korea
| | - Hye Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06355, Republic of Korea
| | - Youngmin Cho
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Young-Jun Kim
- Advanced Functional Polymers Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Dong-Min Lee
- Department of Materials Science and Engineering, Center for Human-oriented Triboelectric Energy Harvesting, Yonsei University, Seoul, 03722, Republic of Korea
| | - Dong-Gyu Lee
- Electronic Materials Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyun-Cheol Song
- Electronic Materials Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KIST-SKKU Carbon-Neutral Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Soo Hyun Nam
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Jong Hee Lee
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06355, Republic of Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, 06355, Republic of Korea
| | - Byung-Ok Choi
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, 06355, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06355, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Sang-Woo Kim
- Department of Materials Science and Engineering, Center for Human-oriented Triboelectric Energy Harvesting, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
3
|
Sankar S, Kodiveri Muthukaliannan G. Deciphering the crosstalk between inflammation and biofilm in chronic wound healing: Phytocompounds loaded bionanomaterials as therapeutics. Saudi J Biol Sci 2024; 31:103963. [PMID: 38425782 PMCID: PMC10904202 DOI: 10.1016/j.sjbs.2024.103963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 03/02/2024] Open
Abstract
In terms of the economics and public health, chronic wounds exert a significant detrimental impact on the health care system. Bacterial infections, which cause the formation of highly resistant biofilms that elude standard antibiotics, are the main cause of chronic, non-healing wounds. Numerous studies have shown that phytochemicals are effective in treating a variety of diseases, and traditional medicinal plants often include important chemical groups such alkaloids, phenolics, tannins, terpenes, steroids, flavonoids, glycosides, and fatty acids. These substances are essential for scavenging free radicals which helps in reducing inflammation, fending off infections, and hastening the healing of wounds. Bacterial species can survive in chronic wound conditions because biofilms employ quorum sensing as a communication technique which regulates the expression of virulence components. Fortunately, several phytochemicals have anti-QS characteristics that efficiently block QS pathways, prevent drug-resistant strains, and reduce biofilm development in chronic wounds. This review emphasizes the potential of phytocompounds as crucial agents for alleviating bacterial infections and promoting wound healing by reducing the inflammation in chronic wounds, exhibiting potential avenues for future therapeutic approaches to mitigate the healthcare burden provided by these challenging conditions.
Collapse
Affiliation(s)
- Srivarshini Sankar
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632 014, Tamil Nadu, India
| | - Gothandam Kodiveri Muthukaliannan
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632 014, Tamil Nadu, India
| |
Collapse
|
4
|
Chakraborty A, Badhe RV, Abbas M, Chauhan A, Jaiswal A, Fareed R, Kumar V, Duan Y, Dutta N. Role of exosomal RNA in wound healing and tissue repair. EXOSOMAL RNA 2024:295-323. [DOI: 10.1016/b978-0-443-14008-2.00001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Humenik F, Danko J, Krešáková L, Vdoviaková K, Vrabec V, Vasilová E, Giretová M, Tóth Š, Fagová Z, Babík J, Medvecký Ľ. A Chitosan-Based Biomaterial Combined with Mesenchymal Stem Cell-Conditioned Medium for Wound Healing and Skin Regeneration. Int J Mol Sci 2023; 24:16080. [PMID: 38003269 PMCID: PMC10671656 DOI: 10.3390/ijms242216080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The aim of this study was to provide a beneficial treatment effect of novel chitosan bio-polymeric material enriched with mesenchymal stem cell products derived from the canine adipose tissue (AT-MSC) on the artificial skin defect in a rabbit model. For the objectivity of the regeneration evaluation, we used histological analysis and a scoring system created by us, taking into account all the attributes of regeneration, such as inflammatory reaction, necrosis, granulation, formation of individual skin layers and hair follicles. We observed an acceleration and improvement in the healing of an artificially created skin defect after eight and ten weeks in comparison with negative control (spontaneous healing without biomaterial). Moreover, we were able to described hair follicles and epidermis layer in histological skin samples treated with a chitosan-based biomaterial on the eighth week after grafting.
Collapse
Affiliation(s)
- Filip Humenik
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Ján Danko
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
- Educational, Scientific and Research Institute AGEL, 811 06 Bratislava, Slovakia
| | - Lenka Krešáková
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Katarína Vdoviaková
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Vladimír Vrabec
- Clinic of Birds, Exotic and Free Living Animals, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Emília Vasilová
- Clinic of Birds, Exotic and Free Living Animals, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Mária Giretová
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, 040 01 Košice, Slovakia
| | - Štefan Tóth
- Department of Histology and Embryology, University of Pavol Jozef Šafárik, 041 80 Košice, Slovakia
| | - Zuzana Fagová
- Department of Histology and Embryology, University of Pavol Jozef Šafárik, 041 80 Košice, Slovakia
| | - Ján Babík
- Clinic of Burns and Reconstructive Medicine, AGEL Hospital, 040 15 Košice-Šaca, Slovakia
| | - Ľubomír Medvecký
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, 040 01 Košice, Slovakia
| |
Collapse
|
6
|
Mukhopadhyay A, Gope A, Choudhury K, Chatterjee J, Mukherjee R. Modulation of Biophysical Cues in Nature Inspired Patterning of Porous Silk Fibroin Scaffold for Replenishable Controlled Drug Delivery. Macromol Biosci 2023; 23:e2300119. [PMID: 37269219 DOI: 10.1002/mabi.202300119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/24/2023] [Indexed: 06/04/2023]
Abstract
While a sticking plasteris enough for healing of most of the minor cuts they may get routinely, critical situations like surgical, gunshot, accidental or diabetic wounds;lacarations and other cutaneous deep cuts may require implants and simultaneous medications for healing. From the biophysical standpoint, an internal force-based physical surface stimulusis crucial for cellular sensing during wound repair. In this paper, the authors report the fabrication of a porous, biomimmetically patterned silk fibroin scaffold loaded with ampicillin, which exhibits controlled release of the drug along with possible replenishment of the same. In vitro swelling study reveals that the scaffolds with hierarchical surface patterns exhibit lower swelling and degradation than other types of scaffolds. The scaffolds, that show remarkable broad-spectrum antibacterial efficacy, exhibit Korsemeyer-Peppas model for the ampicillin release patterns due to the structural hydrophobicity imparted by the patterns. Four distinct cell-matrix adhesion regimes are investigated for the fibroblasts to eventually form cell sheets all over the hierarchical surface structures. 4',6-diamidino-2-phenylindole (DAPI) and Fluorescein Diacetate (FDA) fluorescent staining clearly demonstrate the superiority of patterned surface over its other variants. A comparative immunofluorescence study among collagen I, vinculin, and vimentin expressions substantiated the patterned surface to be superior to others.
Collapse
Affiliation(s)
- Anurup Mukhopadhyay
- Multimodal Imaging and Theranostics Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Ayan Gope
- Advanced Technology Development Centre, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Kabita Choudhury
- Department of Microbiology, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, 700014, India
| | - Jyotirmoy Chatterjee
- Dr.B.C.Roy Multi-Specialty Medical Research Centre, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Rabibrata Mukherjee
- Instability and Soft Patterning Laboratory, Department of Chemical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| |
Collapse
|
7
|
Rashid U, Saba E, Yousaf A, Tareen WA, Sarfraz A, Rhee MH, Sandhu MA. Autologous Platelet Lysate Is an Alternative to Fetal Bovine Serum for Canine Adipose-Derived Mesenchymal Stem Cell Culture and Differentiation. Animals (Basel) 2023; 13:2655. [PMID: 37627446 PMCID: PMC10451755 DOI: 10.3390/ani13162655] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The use of fetal bovine serum (FBS) in regenerative medicine raises serious ethical and scientific concerns. We have cultured and differentiated the canine mesenchymal stem cells (cMSCs) in five different media combinations of autologous platelet lysate (A-PL) and FBS; consisting of 0% A-PL and 10% FBS (M-1), 2.5% A-PL and 7.5% FBS (M-2), 5% A-PL and 5% FBS (M-3), 7.5% A-PL and 2.5% FBS (M-4), and 10% A-PL and 0% FBS (M-5). The cMSCs were evaluated for their doubling time, differentiation efficiency, and expression of CD73, CD90, CD105, and PDGFRα. The mRNA expression of NT5E, THY1, ENG, PPARγ, FABP4, FAS, SP7, BGLAP, and SPP1 was also assessed. The results indicated non-significant differences in cellular proliferation/viability; positive expression of surface markers, and PDGFRα with substantial adipo/osteogenic differentiation. The expression of adipogenic (PPARγ, FABP4, FAS), and osteogenic (SP7, BGLAP, SPP1) genes were higher (p < 0.05) in the M5 group. In conclusion, A-PL in cMSCs culture did not negatively affect cellular proliferation and viability but also enhanced their genetic potential for multilineage differentiation. Our results indicate that A-PL can be used as an alternative for FBS to develop potent cMSCs under good manufacturing practice protocol for regenerative medicine.
Collapse
Affiliation(s)
- Usman Rashid
- Department of Clinical Studies, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi 46300, Pakistan; (U.R.); (A.Y.)
| | - Evelyn Saba
- Department of Veterinary Biomedical Sciences, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi 46300, Pakistan; (E.S.); (W.A.T.)
| | - Arfan Yousaf
- Department of Clinical Studies, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi 46300, Pakistan; (U.R.); (A.Y.)
| | - Waleed Ahsan Tareen
- Department of Veterinary Biomedical Sciences, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi 46300, Pakistan; (E.S.); (W.A.T.)
| | - Adeel Sarfraz
- Department of Anatomy and Histology, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan;
| | - Man Hee Rhee
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Mansur Abdullah Sandhu
- Department of Veterinary Biomedical Sciences, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi 46300, Pakistan; (E.S.); (W.A.T.)
| |
Collapse
|
8
|
Fayed MAA, Al-Wahaibi LH, Bakr RO, Nour MS, Basudan OA, Parvez MK, Al-Dosari MS, Abdel-Mageed WM. Sterols from Centaurea pumilio L. with cell proliferative activity: In vitro and in silico studies. OPEN CHEM 2023; 21. [DOI: 10.1515/chem-2022-0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Abstract
Numerous studies highlighted the impact of natural products, particularly phytosterols, in wound healing while providing less expensive alternatives to chemically synthesized drugs, with less side effects. Centaurea pumilio L. (family Asteraceae) is a rare and endangered species of genus Centaurea with few reports concerning its chemistry. Our phytochemical investigation for the non-polar fraction of its aerial parts led to the isolation and identification of the new compound (6) identified as stigmast-1,5-dien-3-O-β-d-glucopyranoside along with five known sterols and triterpenes (1–5) identified as taraxasterol, β-sitosterol, stigmasterol, β-sitosterol glucoside, and stigmasterol-3-O-β-d-glucopyranoside. Structures of the isolated compounds have been characterized using 1D, 2D NMR, and mass spectral analyses. The cell viability and proliferative activity of the isolated compounds were evaluated using an MTT assay on cultured human primary umbilical vein endothelial cells (HUVEC). None of the compounds exhibited any sign of cytotoxicity. Nonetheless, compounds 5 and 6 moderately enhanced the HUVEC cell growth by 14 and 16%, respectively, at the maximal tested dose (50 µg/mL). As inhibition of glycogen synthase kinase 3-β (GSK3-β) enzyme is important to enhance the wound healing process; therefore, molecular docking was performed to understand the possible interactions between bioactive compounds 5 and 6 and GSK-3β binding pocket active amino acid residues. Both compounds were able to bind to the substrate‑binding site of GSK-3β and potentially interact with the key active site residues, forming strong π and hydrogen interactions with the catalytic site residues, revealing lower binding energy (−7.185 and −6.303 kcal/mol, respectively) than that of indirubin-3-monooxime (−5.303 kcal/mol); thereby representing strong natural replacements candidates for GSK-3β inhibitors.
Collapse
Affiliation(s)
- Marwa A. A. Fayed
- Department of Pharmacognosy, Faculty of Pharmacy, University of Sadat City , Sadat 32897 , Egypt
| | - Lamya H. Al-Wahaibi
- Department of Chemistry, Science College, Princess Nourah Bint Abdulrahman University , Riyadh , Saudi Arabia
| | - Riham O. Bakr
- Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA) , 11787 , Giza , Egypt
| | - Mai S. Nour
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA) , 11787 , Giza , Egypt
| | - Omer A. Basudan
- Department of Pharmacognosy, College of Pharmacy, King Saud University , P.O. Box 2457 , Riyadh 11451 , Saudi Arabia
| | - Mohammad K. Parvez
- Department of Pharmacognosy, College of Pharmacy, King Saud University , P.O. Box 2457 , Riyadh 11451 , Saudi Arabia
| | - Mohammed S. Al-Dosari
- Department of Pharmacognosy, College of Pharmacy, King Saud University , P.O. Box 2457 , Riyadh 11451 , Saudi Arabia
| | - Wael M. Abdel-Mageed
- Department of Pharmacognosy, College of Pharmacy, King Saud University , P.O. Box 2457 , Riyadh 11451 , Saudi Arabia
- Pharmacognosy Department, Faculty of Pharmacy, Assiut University 71526 , Assiut , Egypt
| |
Collapse
|
9
|
Feraru A, Tóth ZR, Mureșan-Pop M, Baia M, Gyulavári T, Páll E, Turcu RVF, Magyari K, Baia L. Anionic Polysaccharide Cryogels: Interaction and In Vitro Behavior of Alginate-Gum Arabic Composites. Polymers (Basel) 2023; 15:polym15081844. [PMID: 37111992 PMCID: PMC10146865 DOI: 10.3390/polym15081844] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/31/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
In the present study, polysaccharide-based cryogels demonstrate their potential to mimic a synthetic extracellular matrix. Alginate-based cryogel composites with different gum arabic ratios were synthesized by an external ionic cross-linking protocol, and the interaction between the anionic polysaccharides was investigated. The structural features provided by FT-IR, Raman, and MAS NMR spectra analysis indicated that a chelation mechanism is the main process linking the two biopolymers. In addition, SEM investigations revealed a porous, interconnected, and well-defined structure suitable as a scaffold in tissue engineering. The in vitro tests confirmed the bioactive character of the cryogels through the development of the apatite layer on the surface of the samples after immersion in simulated body fluid, identifying the formation of a stable phase of calcium phosphate and a small amount of calcium oxalate. Cytotoxicity tests performed on fibroblast cells demonstrated the non-toxic effect of alginate-gum arabic cryogel composites. In addition, an increase in flexibility was noted for samples with a high gum arabic content, which determines an appropriate environment to promote tissue regeneration. The newly obtained biomaterials that exhibit all these properties can be successfully involved in the regeneration of soft tissues, wound management, or controlled drug release systems.
Collapse
Affiliation(s)
- Alexandra Feraru
- Doctoral School of Physics, Babes-Bolyai University, M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania
- Nanostructured Materials and Bio-Nano-Interfaces Center, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, T. Laurian 42, 400271 Cluj-Napoca, Romania
| | - Zsejke-Réka Tóth
- Doctoral School of Physics, Babes-Bolyai University, M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania
- Nanostructured Materials and Bio-Nano-Interfaces Center, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, T. Laurian 42, 400271 Cluj-Napoca, Romania
| | - Marieta Mureșan-Pop
- Nanostructured Materials and Bio-Nano-Interfaces Center, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, T. Laurian 42, 400271 Cluj-Napoca, Romania
| | - Monica Baia
- Faculty of Physics, Babes-Bolyai University, M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania
- Institute for Research-Development-Innovation in Applied Natural Sciences, Babes-Bolyai University, Fântânele 30, 400294 Cluj-Napoca, Romania
| | - Tamás Gyulavári
- Department of Applied and Environmental Chemistry, University of Szeged, Rerrich B. Sqr. 1, 6720 Szeged, Hungary
| | - Emőke Páll
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, Manastur 3-5, 400372 Cluj-Napoca, Romania
| | - Romulus V F Turcu
- Faculty of Physics, Babes-Bolyai University, M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donath 67-103, 400293 Cluj-Napoca, Romania
| | - Klára Magyari
- Nanostructured Materials and Bio-Nano-Interfaces Center, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, T. Laurian 42, 400271 Cluj-Napoca, Romania
| | - Lucian Baia
- Nanostructured Materials and Bio-Nano-Interfaces Center, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, T. Laurian 42, 400271 Cluj-Napoca, Romania
- Faculty of Physics, Babes-Bolyai University, M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania
- Institute for Research-Development-Innovation in Applied Natural Sciences, Babes-Bolyai University, Fântânele 30, 400294 Cluj-Napoca, Romania
| |
Collapse
|
10
|
Ozel C, Apaydin E, Sariboyaci AE, Tamayol A, Avci H. A multifunctional sateen woven dressings for treatment of skin injuries. Colloids Surf B Biointerfaces 2023; 224:113197. [PMID: 36822118 DOI: 10.1016/j.colsurfb.2023.113197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Cutaneous wounds with impaired healing such as diabetic ulcers and burns constitute major and rapidly growing threat to healthcare systems worldwide. Accelerating wound healing requires the delivery of biological factors that induce angiogenesis, support cellular proliferation, and modulate inflammation while minimizing infection. In this study, we engineered a dressing made by weaving of composite fibers (CFs) carrying mesenchymal stem cells (MSCs) and a model antibiotic using a scalable sateen textile technique. In this regard, two different sets of CFs carrying MSCs or an antimicrobial agent were used to generate a multifunctional dressing. According to cell viability and metabolic activity as CCK-8 and live/dead with qRT-PCR results, more than %90 the encapsulated MSCs remain viable for 28 days and their expression levels of the wound repair factors including ECM remodeling, angiogenesis and immunomodulatory maintained in MSCs post dressing manufacturing for 14 days. Post 10 days culture of the dressing, MSCs within CFs had 10-fold higher collagen synthesis (p < 0.0001) determined by hydroxyproline assay which indicates the enhanced healing properties. According to in vitro antimicrobial activity results determined by disk diffusion and broth microdilution tests, the first day and the total amount of release gentamicin loaded dressing samples during the 28 days were higher than determined minimal inhibition concentration (MIC) values for S. aureus and K. pneumonia without negatively impacting the viability and functionality of encapsulated MSCs within the dressing. The dressing is also flexible and can conform to skin curvatures making the dressing suitable for the treatment of different skin injuries such as burns and diabetic ulcers.
Collapse
Affiliation(s)
- Ceren Ozel
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Elif Apaydin
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Department of Biochemistry, Institute of Health Sciences, Anadolu University, Eskişehir 26470, Turkey
| | - Ayla Eker Sariboyaci
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06269, USA.
| | - Huseyin Avci
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Department of Metallurgical and Materials Engineering, Eskişehir Osmangazi University, Eskişehir 26040, Turkey; Translational Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey.
| |
Collapse
|
11
|
Rodriguez-Menocal L, Davis SC, Guzman W, Gil J, Valdes J, Solis M, Higa A, Natesan S, Schulman CI, Christy RJ, Badiavas EV. Model to Inhibit Contraction in Third-Degree Burns Employing Split-Thickness Skin Graft and Administered Bone Marrow-Derived Stem Cells. J Burn Care Res 2023; 44:302-310. [PMID: 36048023 DOI: 10.1093/jbcr/irac119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Indexed: 11/14/2022]
Abstract
Third-degree burns typically result in pronounced scarring and contraction in superficial and deep tissues. Established techniques such as debridement and grafting provide benefit in the acute phase of burn therapy, nevertheless, scar and contraction remain a challenge in deep burns management. Our ambition is to evaluate the effectiveness of novel cell-based therapies, which can be implemented into the standard of care debridement and grafting procedures. Twenty-seven third-degree burn wounds were created on the dorsal area of Red Duroc pig. After 72 h, burns are surgically debrided using a Weck knife. Split-thickness skin grafts (STSGs) were then taken after debridement and placed on burn scars combined with bone marrow stem cells (BM-MSCs). Biopsy samples were taken on days 17, 21, and 45 posttreatment for evaluation. Histological analysis revealed that untreated control scars at 17 days are more raised than burns treated with STSGs alone and/or STSGs with BM-MSCs. Wounds treated with skin grafts plus BM-MSCs appeared thinner and longer, indicative of reduced contraction. qPCR revealed some elevation of α-SMA expression at day 21 and Collagen Iα2 in cells derived from wounds treated with skin grafts alone compared to wounds treated with STSGs + BM-MSCs. We observed a reduction level of TGFβ-1 expression at days 17, 21, and 45 in cells derived from wounds treated compared to controls. These results, where the combined use of stem cells and skin grafts stimulate healing and reduce contraction following third-degree burn injury, have a potential as a novel therapy in the clinic.
Collapse
Affiliation(s)
- Luis Rodriguez-Menocal
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery/Interdisciplinary/Stem Cell Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Stephen C Davis
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, Florida, USA
| | - Wellington Guzman
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery/Interdisciplinary/Stem Cell Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Joel Gil
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, Florida, USA
| | - Jose Valdes
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, Florida, USA
| | - Michael Solis
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, Florida, USA
| | - Alexander Higa
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, Florida, USA
| | - Shanmugasundaram Natesan
- Extremity Trauma and Regenerative Medicine Program, US Army Institute of Surgical Research, Texas, USA
| | - Carl I Schulman
- Department of Surgery, Ryder Trauma Center, University of Miami School of Medicine, Miami, Florida, USA
| | - Robert J Christy
- Extremity Trauma and Regenerative Medicine Program, US Army Institute of Surgical Research, Texas, USA
| | - Evangelos V Badiavas
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery/Interdisciplinary/Stem Cell Institute, University of Miami School of Medicine, Miami, Florida, USA
| |
Collapse
|
12
|
Zhang Q, Kong L, Wang Q, Wang H, Yang Y, Fu J, Zhang Y, Dong J, Zeng C, Liu H. A biotin-stabilized HKUST-1/ADM scaffold for facilitating MSC endothelial differentiation and vascularization in diabetic wound healing. Biomater Sci 2023; 11:854-872. [PMID: 36515094 DOI: 10.1039/d2bm01443b] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inadequate angiogenesis in diabetic wound healing has been identified as one of the most difficult issues to treat. Copper ions (Cu2+) have been confirmed to stimulate angiogenesis; nevertheless, the rapid rise in non-physiological Cu2+ concentrations increases the danger of ion poisoning. For the first time, biotin was used to stabilize a copper-based metal-organic framework (HKUST-1) to change its hydrophobicity and achieve sustained release of Cu2+. The inability to offer a suitable area for the dynamic interaction between cells and growth factors still restricts the use of nanomaterials for the regeneration of injured skin in diabetes. Acellular dermal matrix (ADM) scaffolds are collagen fibers with natural spatial tissue that can create a biological "niche" for cell attachment and growth. In this study, biotin-stabilized HKUST-1 (B-HKUST-1) nanoparticles were modified with an ADM to form a novel scaffold (ADM-B-HKUST-1). Notably, Cu2+ and mesenchymal stem cells (MSCs) released by the composite scaffold may synergistically promote MSC adhesion, proliferation and endothelial differentiation by upregulating the expression of transforming growth factor-β (TGF-β), vascular endothelial growth factor (VEGF) and alpha-smooth muscle actin (α-SMA). Overall, the ADM-B-HKUST1 scaffold combines the dual advantages of the sustained release of Cu2+ and creating a biological "niche" can provide a potential strategy for enhancing angiogenesis and promoting diabetic wound healing.
Collapse
Affiliation(s)
- Qiong Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Linghong Kong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Qi Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Hui Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Yongzhen Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Jinping Fu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Yue Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Jianyue Dong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, 518110, China
- Department of General Medicine, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China.
| | - Hanping Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
13
|
Jenssen AB, Mohamed-Ahmed S, Kankuri E, Brekke RL, Guttormsen AB, Gjertsen BT, Mustafa K, Almeland SK. Administration Methods of Mesenchymal Stem Cells in the Treatment of Burn Wounds. EUROPEAN BURN JOURNAL 2022; 3:493-516. [PMID: 39600017 PMCID: PMC11571831 DOI: 10.3390/ebj3040043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 11/29/2024]
Abstract
Cellular therapies for burn wound healing, including the administration of mesenchymal stem or stromal cells (MSCs), have shown promising results. This review aims to provide an overview of the current administration methods in preclinical and clinical studies of bone-marrow-, adipose-tissue-, and umbilical-cord-derived MSCs for treating burn wounds. Relevant studies were identified through a literature search in PubMed and Embase and subjected to inclusion and exclusion criteria for eligibility. Additional relevant studies were identified through a manual search of reference lists. A total of sixty-nine studies were included in this review. Of the included studies, only five had clinical data from patients, one was a prospective case-control, three were case reports, and one was a case series. Administration methods used were local injection (41% in preclinical and 40% in clinical studies), cell-seeded scaffolds (35% and 20%), topical application (17% and 60%), and systemic injection (1% and 0%). There was great heterogeneity between the studies regarding experimental models, administration methods, and cell dosages. Local injection was the most common administration method in animal studies, while topical application was used in most clinical reports. The best delivery method of MSCs in burn wounds is yet to be identified. Although the potential of MSC treatment for burn wounds is promising, future research should focus on examining the effect and scalability of such therapy in clinical trials.
Collapse
Affiliation(s)
- Astrid Bjørke Jenssen
- Norwegian National Burn Center, Department of Plastic, Hand, and Reconstructive Surgery, Haukeland University Hospital, 5021 Bergen, Norway
| | - Samih Mohamed-Ahmed
- Center for Translational Oral Research (TOR), Tissue Engineering Group, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5020 Bergen, Norway
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Ragnvald Ljones Brekke
- Norwegian National Burn Center, Department of Plastic, Hand, and Reconstructive Surgery, Haukeland University Hospital, 5021 Bergen, Norway
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5020 Bergen, Norway
| | - Anne Berit Guttormsen
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5020 Bergen, Norway
- Department of Anesthesia and Intensive Care, Haukeland University Hospital, 5021 Bergen, Norway
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
- Department of Medicine, Hematology Section, Haukeland University Hospital, 5021 Bergen, Norway
| | - Kamal Mustafa
- Center for Translational Oral Research (TOR), Tissue Engineering Group, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5020 Bergen, Norway
| | - Stian Kreken Almeland
- Norwegian National Burn Center, Department of Plastic, Hand, and Reconstructive Surgery, Haukeland University Hospital, 5021 Bergen, Norway
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5020 Bergen, Norway
| |
Collapse
|
14
|
Selenium-Stimulated Exosomes Enhance Wound Healing by Modulating Inflammation and Angiogenesis. Int J Mol Sci 2022; 23:ijms231911543. [PMID: 36232844 PMCID: PMC9570007 DOI: 10.3390/ijms231911543] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/29/2022] Open
Abstract
Mesenchymal stem cell (MSC)-derived exosomes have emerged as an attractive cell-free tool in tissue engineering and regenerative medicine. The current study aimed to examine the anti-inflammatory, pro-angiogenic, and wound-repair effects of both exosomes and selenium-stimulated exosomes, and check whether the latter had superior wound healing capacity over others. The cellular and molecular network of exosomes, as a paracrine signal, was extensively studied by performing miRNA arrays to explore the key mediators of exosomes in wound healing. Selenium is known to play a critical role in enhancing the proliferation, multi-potency, and anti-inflammatory effects of MSCs. Selenium-stimulated exosomes showed significant effects in inhibiting inflammation and improving pro-angiogenesis in human umbilical vein endothelial cells. Cell growth and the migration of human dermal fibroblasts and wound regeneration were more enhanced in the selenium-stimulated exosome group than in the selenium and exosome groups, thereby further promoting the wound healing in vivo. Taken together, selenium was found to augment the therapeutic effects of adipose MSC-derived exosomes in tissue regeneration. We concluded that selenium may be considered a vital agent for wound healing in stem cell-based cell-free therapies.
Collapse
|
15
|
Wei X, Li M, Zheng Z, Ma J, Gao Y, Chen L, Peng Y, Yu S, Yang L. Senescence in chronic wounds and potential targeted therapies. BURNS & TRAUMA 2022; 10:tkab045. [PMID: 35187179 PMCID: PMC8853744 DOI: 10.1093/burnst/tkab045] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/15/2021] [Accepted: 11/19/2021] [Indexed: 01/10/2023]
Abstract
Chronic wounds (e.g. diabetic wounds, pressure wounds, vascular ulcers, etc.) do not usually heal in a timely and orderly manner but rather last for years and may lead to irreversible adverse events, resulting in a substantial financial burden for patients and society. Recently, a large amount of evidence has proven that cellular senescence has a crucial influence on chronic nonhealing wounds. As a defensive mechanism, cell senescence is a manner of cell-cycle arrest with increased secretory phenotype to resist death, preventing cells from stress-induced damage in cancer and noncancer diseases. A growing amount of research has advanced the perception of cell senescence in various chronic wounds and focuses on pathological and physiological processes and therapies targeting senescent cells. However, previous reviews have failed to sum up novel understandings of senescence in chronic wounds and emerging strategies targeting senescence. Herein, we discuss the characteristics and mechanisms of cellular senescence and the link between senescence and chronic wounds as well as some novel antisenescence strategies targeting other diseases that may be applied for chronic wounds.
Collapse
Affiliation(s)
- Xuerong Wei
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Minxiong Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, 200011, Shanghai, China
| | - Zijun Zheng
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Jun Ma
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Yujie Peng
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Shengxiang Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| |
Collapse
|
16
|
Schulman CI, Namias N, Pizano L, Rodriguez-Menocal L, Aickara D, Guzman W, Candanedo A, Maranda E, Beirn A, Badiavas EV. The effect of mesenchymal stem cells improves the healing of burn wounds: a phase 1 dose-escalation clinical trial. Scars Burn Heal 2022; 8:20595131211070783. [PMID: 35781931 PMCID: PMC9247372 DOI: 10.1177/20595131211070783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background Stem cell therapy holds promise to improve healing and stimulate tissue
regeneration after burn injury. Preclinical evidence has supported this;
however, clinical studies are lacking. We examined the application of bone
marrow-derived mesenchymal stem cells (BM-MSC) to deep second-degree burn
injuries using a two-dose escalation protocol. Methods Ten individuals aged 18 years or older with deep second-degree burn wounds
were enrolled. The first five patients were administered 2.5 × 10³
BM-MSC/cm2 to their wounds. After safety of the initial dose
level was assessed, a second group of five patients was treated with a
higher concentration of 5 × 10³ allogeneic BM-MSC/cm2. Safety was
assessed clinically and by evaluating cytokine levels in mixed recipient
lymphocyte/donor BM-MSC reactions (INFγ, IL-10 and TNFα). At each visit, we
performed wound measurements and assessed wounds using a Patient and
Observer Scar Assessment Scale (POSAS). Results All patients responded well to treatment, with 100% closure of wounds and
minimal clinical evidence of fibrosis. No adverse reactions or evidence of
rejection were observed for both dose levels. Patients receiving the first
dose concentration had a wound closure rate of 3.64 cm2/day.
Patients receiving the second dose concentration demonstrated a wound
closure rate of 10.47 cm2/day. The difference in healing rates
between the two groups was not found to be statistically significant
(P = 0.17). Conclusion BM-MSC appear beneficial in optimising wound healing in patients with deep
second-degree burn wounds. Adverse outcomes were not observed when
administering multiple doses of allogeneic BM-MSC. Lay Summary Thermal injuries are a significant source of morbidity and mortality,
constituting 5%–20% of all injuries and 4% of all deaths. Despite overall
improvements in the management of acutely burned patients, morbidities
associated with deeper burn injuries remain commonplace. Burn patients are
too often left with significant tissue loss, scarring and contractions
leading to physical loss of function and long-lasting psychological and
emotional impacts. In previous studies, we have demonstrated the safety and efficacy of
administering bone marrow-derived mesenchymal stem cells (BM-MSC) to chronic
wounds with substantial improvement in healing and evidence of tissue
regeneration. In this report, we have examined the application of BM-MSC to
deep second-degree burn injuries in patients. The aim of the present phase I/II clinical trial was to examine the safety
and efficacy of administering allogeneic BM-MSC to deep second-degree burns.
We utilised two different dose levels at concentrations 2.5 × 103
and 5 × 103 cells/cm2. Patients with deep
second-degree burn wounds up to 20% of the total body surface area were
eligible for treatment. Allogeneic BM-MSC were applied to burn wounds
topically or by injection under transparent film dressing <7 days after
injury. Patients were followed for at least six months after treatment. Using two dose levels allowed us to gain preliminary information as to
whether different amounts of BM-MSC administered to burn wounds will result
in significant differences in safety/ clinical response. Once the safety and
dose-response analysis were completed, we evaluated the efficacy of
allogeneic stem cell therapy in the treatment of deep second-degree burn
wounds. In this study, we examined the role of allogeneic BM-MSC treatment in
patients with deep second-degree burn injuries, in a dose-dependent manner.
No significant related adverse events were reported. Safety was evaluated
both clinically and by laboratory-based methods. Efficacy was assessed
clinically through evidence of re-pigmentation, hair follicle restoration
and regenerative change. While these findings are encouraging, more studies
will be needed to better establish the benefit of BM-MSC in the treatment of
burn injuries.
Collapse
Affiliation(s)
- Carl I Schulman
- Department of Surgery, Ryder Trauma Center, University of Miami School of Medicine, Miami, FL, USA
| | - Nicholas Namias
- Department of Surgery, Ryder Trauma Center, University of Miami School of Medicine, Miami, FL, USA
| | - Louis Pizano
- Department of Surgery, Ryder Trauma Center, University of Miami School of Medicine, Miami, FL, USA
| | - Luis Rodriguez-Menocal
- Department of Dermatology and Cutaneous Surgery. Leonard M Miller School of Medicine, University of Miami, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Divya Aickara
- Department of Dermatology and Cutaneous Surgery. Leonard M Miller School of Medicine, University of Miami, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Wellington Guzman
- Department of Surgery, Ryder Trauma Center, University of Miami School of Medicine, Miami, FL, USA
| | - Ambar Candanedo
- Department of Surgery, Ryder Trauma Center, University of Miami School of Medicine, Miami, FL, USA
| | - Eric Maranda
- Department of Dermatology and Cutaneous Surgery. Leonard M Miller School of Medicine, University of Miami, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Audrey Beirn
- Department of Dermatology and Cutaneous Surgery. Leonard M Miller School of Medicine, University of Miami, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Evangelos V Badiavas
- Department of Dermatology and Cutaneous Surgery. Leonard M Miller School of Medicine, University of Miami, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| |
Collapse
|
17
|
Marofi F, Alexandrovna KI, Margiana R, Bahramali M, Suksatan W, Abdelbasset WK, Chupradit S, Nasimi M, Maashi MS. MSCs and their exosomes: a rapidly evolving approach in the context of cutaneous wounds therapy. Stem Cell Res Ther 2021; 12:597. [PMID: 34863308 PMCID: PMC8642895 DOI: 10.1186/s13287-021-02662-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/14/2021] [Indexed: 12/18/2022] Open
Abstract
Currently, mesenchymal stem/stromal stem cell (MSC) therapy has become a promising option for accelerating cutaneous wound healing. In vivo reports have outlined the robust competences of MSCs to offer a solid milieu by inhibition of inflammatory reactions, which in turn, enables skin regeneration. Further, due to their great potential to stimulate angiogenesis and also facilitate matrix remodeling, MSCs hold substantial potential as future therapeutic strategies in this context. The MSCs-induced wound healing is thought to mainly rely on the secretion of a myriad of paracrine factors in addition to their direct differentiation to skin-resident cells. Besides, MSCs-derived exosomes as nanoscale and closed membrane vesicles have recently been suggested as an effective and cell-free approach to support skin regeneration, circumventing the concerns respecting direct application of MSCs. The MSCs-derived exosomes comprise molecular components including lipid, proteins, DNA, microRNA, and also mRNA, which target molecular pathways and also biological activities in recipient cells (e.g., endothelial cell, keratinocyte, and fibroblast). The secreted exosome modifies macrophage activation, stimulates angiogenesis, and instigates keratinocytes and dermal fibroblast proliferations as well as migrations concurrently regulate inherent potential of myofibroblast for adjustment of turnover of the ECM. In the present review, we will focus on the recent findings concerning the application of MSCs and their derivative exosome to support wound healing and skin regeneration, with special focus on last decade in vivo reports.
Collapse
Affiliation(s)
- Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master’s Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Mahta Bahramali
- Biotechnology Department, University of Tehran, Tehran, Iran
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210 Thailand
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
| | | | - Marwah Suliman Maashi
- Stem Cells and Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah, Saudi Arabia
| |
Collapse
|
18
|
Bray ER, Oropallo AR, Grande DA, Kirsner RS, Badiavas EV. Extracellular Vesicles as Therapeutic Tools for the Treatment of Chronic Wounds. Pharmaceutics 2021; 13:1543. [PMID: 34683836 PMCID: PMC8541217 DOI: 10.3390/pharmaceutics13101543] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/17/2022] Open
Abstract
Chronic wounds develop when the orderly process of cutaneous wound healing is delayed or disrupted. Development of a chronic wound is associated with significant morbidity and financial burden to the individual and health-care system. Therefore, new therapeutic modalities are needed to address this serious condition. Mesenchymal stem cells (MSCs) promote skin repair, but their clinical use has been limited due to technical challenges. Extracellular vesicles (EVs) are particles released by cells that carry bioactive molecules (lipids, proteins, and nucleic acids) and regulate intercellular communication. EVs (exosomes, microvesicles, and apoptotic bodies) mediate key therapeutic effects of MSCs. In this review we examine the experimental data establishing a role for EVs in wound healing. Then, we explore techniques for designing EVs to function as a targeted drug delivery system and how EVs can be incorporated into biomaterials to produce a personalized wound dressing. Finally, we discuss the status of clinically deploying EVs as a therapeutic agent in wound care.
Collapse
Affiliation(s)
- Eric R. Bray
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alisha R. Oropallo
- Comprehensive Wound Healing Center and Hyperbarics, Department of Vascular Surgery, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell Health, Hempstead, NY 11549, USA; (A.R.O.); (D.A.G.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Daniel A. Grande
- Comprehensive Wound Healing Center and Hyperbarics, Department of Vascular Surgery, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell Health, Hempstead, NY 11549, USA; (A.R.O.); (D.A.G.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Department of Orthopedic Surgery, Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY 11040, USA
| | - Robert S. Kirsner
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
| | - Evangelos V. Badiavas
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
19
|
Update on the Basic Science Concepts and Applications of Adipose-Derived Stem Cells in Hand and Craniofacial Surgery. Plast Reconstr Surg 2021; 148:475e-486e. [PMID: 34432707 DOI: 10.1097/prs.0000000000008279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
SUMMARY Adipose-derived stem cell therapy offers plastic surgeons a novel treatment alternative for conditions with few therapeutic options. Adipose-derived stem cells are a promising treatment because of their broad differentiation potential, capacity for self-renewal, and ease of isolation. Over the past decade, plastic surgeons have attempted to harness adipose-derived stem cells' unique cellular characteristics to improve the survival of traditional fat grafting procedures, a process known as cell-assisted lipotransfer. However, the full implications of cell-assisted lipotransfer in clinical practice remain incompletely understood, stressing the urgent need to assess the scientific evidence supporting adipose-derived stem cell-based interventions. Furthermore, with the strict regulatory climate surrounding tissue explantation therapies, reviewing the safety and efficacy of these treatments will clarify their regulatory viability moving forward. In this report, the authors provide a comprehensive, up-to-date appraisal of best evidence-based practices supporting adipose-derived stem cell-derived therapies, highlighting the known mechanisms behind current clinical applications in tissue engineering and regenerative medicine specific to plastic and reconstructive surgery. The authors outline best practices for the harvest and isolation of adipose-derived stem cells and discuss why procedure standardization will elucidate the scientific bases for their broad use. Finally, the authors discuss challenges posed by U.S. Food and Drug Administration oversight of these cell-based therapies and examine the role of adipose-derived stem cell-based applications in the future of plastic surgery.
Collapse
|
20
|
Proteomic analysis of bone marrow-derived mesenchymal stem cell extracellular vesicles from healthy donors: implications for proliferation, angiogenesis, Wnt signaling, and the basement membrane. Stem Cell Res Ther 2021; 12:328. [PMID: 34090527 PMCID: PMC8180068 DOI: 10.1186/s13287-021-02405-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Background Bone marrow-derived mesenchymal stem cells (BM-MSCs) have shown therapeutic potential in various in vitro and in vivo studies in cutaneous wound healing. Furthermore, there are ubiquitous studies highlighting the pro-regenerative effects of BM-MSC extracellular vesicles (BM-MSC EVs). The similarities and differences in BM-MSC EV cargo among potential healthy donors are not well understood. Variation in EV protein cargo is important to understand, as it may be useful in identifying potential therapeutic applications in clinical trials. We hypothesized that the donors would share both important similarities and differences in cargo relating to cell proliferation, angiogenesis, Wnt signaling, and basement membrane formation—processes shown to be critical for effective cutaneous wound healing. Methods We harvested BM-MSC EVs from four healthy human donors who underwent strict screening for whole bone marrow donation and further Good Manufacturing Practices-grade cell culture expansion for candidate usage in clinical trials. BM-MSC EV protein cargo was determined via mass spectrometry and Proteome Discoverer software. Corresponding proteomic networks were analyzed via the UniProt Consortium and STRING consortium databases. Results More than 3000 proteins were identified in each of the donors, sharing > 600 proteins among all donors. Despite inter-donor variation in protein identities, there were striking similarities in numbers of proteins per biological functional category. In terms of biologic function, the proteins were most associated with transport of ions and proteins, transcription, and the cell cycle, relating to cell proliferation. The donors shared essential cargo relating to angiogenesis, Wnt signaling, and basement membrane formation—essential processes in modulating cutaneous wound repair. Conclusions Healthy donors of BM-MSC EVs contain important similarities and differences among protein cargo that may play important roles in their pro-regenerative functions. Further studies are needed to correlate proteomic signatures to functional outcomes in cutaneous repair.
Collapse
|
21
|
Human Adipose Mesenchymal Stem Cell-Derived Exosomes: A Key Player in Wound Healing. Tissue Eng Regen Med 2021; 18:537-548. [PMID: 33547566 DOI: 10.1007/s13770-020-00316-x] [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] [Received: 08/31/2020] [Revised: 10/05/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Human adipose-derived mesenchymal stem cells (AMSCs) are an attractive resource for wound healing because their regenerative capacity improves injury repair. Recently, stem cell-derived exosomes have been shown to play a positive role in stem cell-based therapies. However, the effects of exosomes derived from AMSCs (AEXOs) on wound healing are unclear. In this study, we aimed to examine the role of AEXOs in attenuating inflammation and explore their effects in normal wound healing. METHODS We isolated exosomes from AMSCs and established a cellular model of inflammation by treatment with the inflammatory cytokines, interferon gamma and tumor necrosis factor alpha, to determine whether AEXOs can inhibit inflammation. We examined the wound healing effects of AEXOs in in vitro wound healing models and performed a miRNA array to understand the role of AEXOs in inflammation and wound healing. RESULTS A significant difference was observed in wound closure and the expression of anti-inflammatory and wound-healing-related factors between control and AEXO-treated cells. CONCLUSION Our results showed that besides alleviating the inflammation response, AEXOs also promote wound healing. Thus, AEXOs represent a novel, stem-cell-based, therapeutic strategy for wound healing.
Collapse
|
22
|
Rothmiller S, Jäger N, Meier N, Meyer T, Neu A, Steinritz D, Thiermann H, Scherer M, Rummel C, Mangerich A, Bürkle A, Schmidt A. Chronic senescent human mesenchymal stem cells as possible contributor to the wound healing disorder after exposure to the alkylating agent sulfur mustard. Arch Toxicol 2021; 95:727-747. [PMID: 33491125 PMCID: PMC7870771 DOI: 10.1007/s00204-020-02946-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022]
Abstract
Wound healing is a complex process, and disturbance of even a single mechanism can result in chronic ulcers developing after exposure to the alkylating agent sulfur mustard (SM). A possible contributor may be SM-induced chronic senescent mesenchymal stem cells (MSCs), unable to fulfil their regenerative role, by persisting over long time periods and creating a proinflammatory microenvironment. Here we show that senescence induction in human bone marrow derived MSCs was time- and concentration-dependent, and chronic senescence could be verified 3 weeks after exposure to between 10 and 40 µM SM. Morphological changes, reduced clonogenic and migration potential, longer scratch closure times, differences in senescence, motility and DNA damage response associated genes as well as increased levels of proinflammatory cytokines were revealed. Selective removal of these cells by senolytic drugs, in which ABT-263 showed initial potential in vitro, opens the possibility for an innovative treatment strategy for chronic wounds, but also tumors and age-related diseases.
Collapse
Affiliation(s)
- Simone Rothmiller
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
- Molecular Toxicology Group, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Niklas Jäger
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Nicole Meier
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Thimo Meyer
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Adrian Neu
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
- Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Goethestr. 33, 80336, Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Michael Scherer
- Department of Traumatology and Orthopedics, HELIOS Amper Clinics, Krankenhausstraße 15, 85221, Dachau, Germany
| | - Christoph Rummel
- Department of Orthopedics and Sports Medicine, Wolfart Clinic, Waldstraße 7, 82166, Gräfelfing, Germany
| | - Aswin Mangerich
- Molecular Toxicology Group, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany.
- Faculty of Human Sciences, Institute for Sports Sciences, Universität Der Bundeswehr München, Werner-Heisenberg-Weg 39, 85577, Neubiberg, Germany.
| |
Collapse
|
23
|
Catanzano O, Quaglia F, Boateng JS. Wound dressings as growth factor delivery platforms for chronic wound healing. Expert Opin Drug Deliv 2021; 18:737-759. [PMID: 33338386 DOI: 10.1080/17425247.2021.1867096] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Years of tissue engineering research have clearly demonstrated the potential of integrating growth factors (GFs) into scaffolds for tissue regeneration, a concept that has recently been applied to wound dressings. The old concept of wound dressings that only take a passive role in wound healing has now been overtaken, and advanced dressings which can take an active part in wound healing, are of current research interest.Areas covered: In this review we will focus on the recent strategies for the delivery of GFs to wound sites with an emphasis on the different approaches used to achieve fine tuning of spatial and temporal concentrations to achieve therapeutic efficacy.Expert opinion: The use of GFs to accelerate wound healing and reduce scar formation is now considered a feasible therapeutic approach in patients with a high risk of infections and complications. The integration of micro - and nanotechnologies into wound dressings could be the key to overcome the inherent instability of GFs and offer adequate control over the release rate. Many investigations have led to encouraging outcomes in various in vitro and in vivo wound models, and it is expected that some of these technologies will satisfy clinical needs and will enter commercialization.
Collapse
Affiliation(s)
- Ovidio Catanzano
- Institute for Polymers Composites and Biomaterials (IPCB) - CNR, Pozzuoli, Italy
| | - Fabiana Quaglia
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, Naples, Italy
| | - Joshua S Boateng
- School of Science, Faculty of Engineering and Science, University of Greenwich, Medway, Central Avenue, Chatham Maritime, Kent, UK
| |
Collapse
|
24
|
Hou Y, Li J, Guan S, Witte F. The therapeutic potential of MSC-EVs as a bioactive material for wound healing. ENGINEERED REGENERATION 2021. [DOI: 10.1016/j.engreg.2021.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
25
|
Wang X, Deng M, Yu Z, Cai Y, Liu W, Zhou G, Wang X, Cao Y, Li W, Zhang W. Cell-free fat extract accelerates diabetic wound healing in db/db mice. Am J Transl Res 2020; 12:4216-4227. [PMID: 32913499 PMCID: PMC7476113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/16/2020] [Indexed: 06/11/2023]
Abstract
Cell-free fat extract (CEFFE), the liquid fraction derived from fat tissues, is enriched with a variety of growth factors and possesses pro-angiogenic, anti-apoptotic, and anti-oxidative properties. The aim of this study was to determine if CEFFE could accelerate chronic wound healing in mice with diabetes and investigate its underlying mechanisms. A model of circular full-thickness wound (6 mm diameter) was produced in the central dorsal region of spontaneous type 2 diabetes mellitus db/db mice. The mice were divided to three groups depending on dosage of CEFFE administered for the study; high dose CEFFE group (CEFFEhigh; administered 2.5 ml/kg/day via subcutaneous injection for six days), low dose CEFFE group (CEFFElow; administered 2.5 ml/kg/day via subcutaneous injection for three days), and a control group receiving phosphate buffer solution. Wound closure was evaluated on day 3, 7, 10, and 14 post-operation. Histological analyses, including hematoxylin-eosin staining and Masson's trichrome staining and immunohistological staining of anti-CD31 and anti-CD68, were also performed. Moreover, the effects of CEFFE on proliferation, migration, and tube formation of human immortal keratinocyte cells (HaCaT) and human vascular endothelial cells (HUVEC) were tested in vitro. The results showed that the local injection of CEFFE significantly accelerated wound healing in mice with diabetes. CEFFE improved re-epithelization and collagen secretion, promoted angiogenesis, and inhibited inflammatory macrophage infiltration in vivo. CEFFE also promoted HaCaT proliferation and migration and enhanced tubular formation in cultured HUVEC. It was concluded that CEFFE accelerates wound healing through pro-angiogenic and anti-inflammatory activities.
Collapse
Affiliation(s)
- Xiangsheng Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Mingwu Deng
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Ziyou Yu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Yizuo Cai
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
- National Tissue Engineering Center of ChinaShanghai 200041, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
- National Tissue Engineering Center of ChinaShanghai 200041, China
| | - Xiansong Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
- National Tissue Engineering Center of ChinaShanghai 200041, China
| | - Wei Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
- National Tissue Engineering Center of ChinaShanghai 200041, China
| |
Collapse
|
26
|
Katzengold R, Orlov A, Gefen A. A novel system for dynamic stretching of cell cultures reveals the mechanobiology for delivering better negative pressure wound therapy. Biomech Model Mechanobiol 2020; 20:193-204. [PMID: 32803464 DOI: 10.1007/s10237-020-01377-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
Serious wounds, both chronic and acute (e.g., surgical), are among the most common, expensive and difficult-to-treat health problems. Negative pressure wound therapy (NPWT) is considered a mainstream procedure for treating both wound types. Soft tissue deformation stimuli are the crux of NPWT, enhancing cell proliferation and migration from peri-wound tissues which contributes to healing. We developed a dynamic stretching device (DSD) contained in a miniature incubator for applying controlled deformations to fibroblast wound assays. Prior to the stretching experiments, fibroblasts were seeded in 6-well culture plates with elastic substrata and let to reach confluency. Squashing damage was then induced at the culture centers, and the DSD was activated to deliver stretching regimes that represented common clinical NPWT protocols at two peak strain levels, 0.5% and 3%. Analyses of the normalized maximal migration rate (MMR) data for the collective cell movement revealed that for the 3% strain level, the normalized MMR of cultures subjected to a 0.1 Hz stretch frequency regime was ~ 1.4 times and statistically significantly greater (p < 0.05) than that of the cultures subjected to no-stretch (control) or to static stretch (2nd control). Correspondingly, analysis of the time to gap closure data indicated that the closure time of the wound assays subjected to the 0.1 Hz regime was ~ 30% shorter than that of the cultures subjected to the control regimes (p < 0.05). Other simulated NPWT protocols did not emerge as superior to the controls. The present method and system are a powerful platform for further revealing the mechanobiology of NPWT and for improving this technology.
Collapse
Affiliation(s)
- Rona Katzengold
- The Herbert J. Berman Chair in Vascular Bioengineering, Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Alexey Orlov
- The Herbert J. Berman Chair in Vascular Bioengineering, Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Amit Gefen
- The Herbert J. Berman Chair in Vascular Bioengineering, Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
27
|
Wang Z, Shi C. Cellular senescence is a promising target for chronic wounds: a comprehensive review. BURNS & TRAUMA 2020; 8:tkaa021. [PMID: 32607375 PMCID: PMC7309580 DOI: 10.1093/burnst/tkaa021] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/07/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022]
Abstract
Chronic wounds include, but are not limited to, radiation ulcers, pressure ulcers, vascular ulcers and diabetic foot ulcers. These chronic wounds can persist for years without healing and severe ulcers may lead to amputation. Unfortunately, the underlying pathologies of refractory chronic wounds are not fully characterized, and new treatments are urgently needed. Recently, increasing evidence has indicated that cell senescence plays an important role in the development of chronic wounds, and preventing cell senescence or removing senescent cells holds promise as a new therapeutic strategy. In this review, we aim to probe these latest findings to promote the understanding of cellular senescence in the pathological process and potential management of chronic wounds.
Collapse
Affiliation(s)
- Ziwen Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Chongqing, 400038, China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Chongqing, 400038, China
| |
Collapse
|
28
|
He L, Zhu C, Jia J, Hao XY, Yu XY, Liu XY, Shu MG. ADSC-Exos containing MALAT1 promotes wound healing by targeting miR-124 through activating Wnt/β-catenin pathway. Biosci Rep 2020; 40:BSR20192549. [PMID: 32342982 PMCID: PMC7214401 DOI: 10.1042/bsr20192549] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 12/18/2022] Open
Abstract
Cutaneous wound is a soft tissue injury that is difficult to heal during aging. It has been demonstrated that adipose-derived stem cells (ADSCs) and its secreted exosomes exert crucial functions in cutaneous wound healing. The present study aimed to elucidate the mechanism of exosomes derived from ADSCs (ADSC-Exos) containing MALAT1 in wound healing. ADSCs were isolated from human normal subcutaneous adipose tissues and identified by flow cytometry analysis. Exosomes were extracted from ADSC supernatants and MALAT1 expression was determined using qRT-PCR analysis. HaCaT and HDF cells were exposed to hydrogen peroxide (H2O2) for simulating the skin lesion model. Subsequently, CCK-8, flow cytometry, wound healing and transwell assays were employed to validate the role of ADSC-Exos containing MALAT1 in the skin lesion model. Besides, cells were transfected with sh-MALAT1 to verify the protective role of MALAT1 in wound healing. The binding relationship between MALAT1 and miR-124 were measured by dual-luciferase reporter assay. ADSC-Exos promoted cell proliferation, migration, and inhibited cell apoptosis of HaCaT and HDF cells impaired by H2O2. However, the depletion of MALAT1 in ADSC-Exos lose these protective effects on HaCaT and HDF cells. Moreover, miR-124 was identified to be a target of MALAT1. Furthermore, ADSC-Exos containing MALAT1 could mediate H2O2-induced wound healing by targeting miR-124 and activating Wnt/β-catenin pathway. ADSC-Exos containing MALAT1 play a positive role in cutaneous wound healing possibly via targeting miR-124 through activating the Wnt/β-catenin pathway, which may provide novel insights into the therapeutic target for cutaneous wound healing.
Collapse
Affiliation(s)
- Lin He
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, P.R. China
| | - Chan Zhu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Jing Jia
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, P.R. China
| | - Xiao-Yan Hao
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, P.R. China
| | - Xue-Yuan Yu
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, P.R. China
| | - Xiang-Yu Liu
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, P.R. China
| | - Mao-Guo Shu
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, P.R. China
| |
Collapse
|
29
|
Zhao G, Liu F, Liu Z, Zuo K, Wang B, Zhang Y, Han X, Lian A, Wang Y, Liu M, Zou F, Li P, Liu X, Jin M, Liu JY. MSC-derived exosomes attenuate cell death through suppressing AIF nucleus translocation and enhance cutaneous wound healing. Stem Cell Res Ther 2020; 11:174. [PMID: 32393338 PMCID: PMC7212595 DOI: 10.1186/s13287-020-01616-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/02/2020] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Background Skin wounding is very common and may be slow to heal. Increasing evidence shows that exosomes derived from mesenchymal stem cells (MSCs) dramatically enhance skin wound healing in a paracrine manner. However, the mechanism underlying this phenomenon has not yet been elucidated. Thus, the objective of the present study was to identify the signaling pathways and paracrine factors by which MSC-derived exosomes promote de novo skin tissue regeneration in response to wound healing. Methods In vitro and in vivo skin wound healing models were created by treating immortalized human keratinocytes (HaCaT) with hydrogen peroxide (H2O2) and excising full-thickness mouse skin, respectively. Exosomes were extracted from human umbilical cord Wharton’s jelly MSCs (hucMSC-Ex) by ultracentrifugation of cell culture supernatant. Results The hucMSC-Ex treatment significantly increased HaCaT cell proliferation and migration in a time- and dose-dependent manner, suppressed HaCaT apoptosis induced with H2O2 by inhibiting nuclear translocation of apoptosis-inducing factor (AIF) and upregulating poly ADP ribose polymerase 1 (PARP-1) and poly (ADP-ribose) (PAR). The animal experiments showed that relative to hucMSCs, hucMSC-Ex attenuated full-thickness skin wounding by enhancing epidermal re-epithelialization and dermal angiogenesis. Conclusions These findings indicated that direct administration of hucMSC-Ex may effectively treat cutaneous wounding and could be of great value in clinical settings.
Collapse
Affiliation(s)
- Guifang Zhao
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China.,Department of Pathology, Jilin Medical University, Jilin, China
| | - Feilin Liu
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, China
| | - Zinan Liu
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Kuiyang Zuo
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Bo Wang
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Yuying Zhang
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Xing Han
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Aobo Lian
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Yuan Wang
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Mingsheng Liu
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Fei Zou
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Pengdong Li
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Xiaomei Liu
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Minghua Jin
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Jin Yu Liu
- Department of Toxicology, School of Public Health, Jilin University, No. 1163 Xinmin Street, Changchun, Jilin, 130021, China.
| |
Collapse
|
30
|
Vivcharenko V, Wojcik M, Przekora A. Cellular Response to Vitamin C-Enriched Chitosan/Agarose Film with Potential Application as Artificial Skin Substitute for Chronic Wound Treatment. Cells 2020; 9:cells9051185. [PMID: 32397594 PMCID: PMC7290375 DOI: 10.3390/cells9051185] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/30/2020] [Accepted: 05/08/2020] [Indexed: 12/13/2022] Open
Abstract
The treatment of chronic wounds is still a meaningful challenge to physicians. The aim of this work was to produce vitamin C-enriched chitosan/agarose (CHN/A) film that could serve as potential artificial skin substitute for chronic wound treatment. The biomaterial was fabricated by a newly developed and simplified method via mixing acidic chitosan solution with alkaline agarose solution that allowed to obtain slightly acidic pH (5.97) of the resultant material, which is known to support skin regeneration. Vitamin C was immobilized within the matrix of the film by entrapment method during production process. Produced films (CHN/A and CHN/A + vit C) were subjected to comprehensive evaluation of cellular response with the use of human skin fibroblasts, epidermal keratinocytes, and macrophages. It was demonstrated that novel biomaterials support adhesion and growth of human skin fibroblasts and keratinocytes, have ability to slightly reduce transforming growth factor-beta 1 (TGF-β1) (known to be present at augmented levels in the epidermis of chronic wounds), and increase platelet-derived growth factor-BB (PDGF-BB) secretion by the cells. Nevertheless, addition of vitamin C to the biomaterial formulation does not significantly improve its biological properties due to burst vitamin release profile. Obtained results clearly demonstrated that produced CHN/A film has great potential to be used as cellular dermal, epidermal, or dermo-epidermal graft pre-seeded with human skin cells for chronic wound treatment.
Collapse
|
31
|
Baldan-Martin M, Martin-Rojas T, Corbacho-Alonso N, Lopez JA, Sastre-Oliva T, Gil-Dones F, Vazquez J, Arevalo JM, Mourino-Alvarez L, Barderas MG. Comprehensive Proteomic Profiling of Pressure Ulcers in Patients with Spinal Cord Injury Identifies a Specific Protein Pattern of Pathology. Adv Wound Care (New Rochelle) 2020; 9:277-294. [PMID: 32226651 PMCID: PMC7099418 DOI: 10.1089/wound.2019.0968] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/10/2019] [Indexed: 12/26/2022] Open
Abstract
Objective: Severe pressure ulcers (PUs) do not respond to conservative wound therapy and need surgical repair. To better understand the pathogenesis and to advance on new therapeutic options, we focused on the proteomic analysis of PU, which offers substantial opportunities to identify significant changes in protein abundance during the course of PU formation in an unbiased manner. Approach: To better define the protein pattern of this pathology, we performed a proteomic approach in which we compare severe PU tissue from spinal cord injury (SCI) patients with control tissue from the same patients. Results: We found 76 proteins with difference in abundance. Of these, 10 proteins were verified as proteins that define the pathology: antithrombin-III, alpha-1-antitrypsin, kininogen-1, alpha-2-macroglobulin, fibronectin, apolipoprotein A-I, collagen alpha-1 (XII) chain, haptoglobin, apolipoprotein B-100, and complement factor B. Innovation: This is the first study to analyze differential abundance protein of PU tissue from SCI patients using high-throughput protein identification and quantification by tandem mass tags followed by liquid chromatography tandem mass spectrometry. Conclusion: Differential abundance proteins are mainly involved in tissue regeneration. These proteins might be considered as future therapeutic options to enhance the physiological response and permit cellular repair of damaged tissue.
Collapse
Affiliation(s)
- Montserrat Baldan-Martin
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Tatiana Martin-Rojas
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Nerea Corbacho-Alonso
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Juan Antonio Lopez
- Department of Plastic Surgery, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Tamara Sastre-Oliva
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Felix Gil-Dones
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Jesus Vazquez
- Department of Plastic Surgery, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | | | - Laura Mourino-Alvarez
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Maria G. Barderas
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| |
Collapse
|
32
|
Shukla L, Yuan Y, Shayan R, Greening DW, Karnezis T. Fat Therapeutics: The Clinical Capacity of Adipose-Derived Stem Cells and Exosomes for Human Disease and Tissue Regeneration. Front Pharmacol 2020; 11:158. [PMID: 32194404 PMCID: PMC7062679 DOI: 10.3389/fphar.2020.00158] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
Fat grafting is a well-established surgical technique used in plastic surgery to restore deficient tissue, and more recently, for its putative regenerative properties. Despite more frequent use of fat grafting, however, a scientific understanding of the mechanisms underlying either survival or remedial benefits of grafted fat remain lacking. Clinical use of fat grafts for breast reconstruction in tissues damaged by radiotherapy first provided clues regarding the clinical potential of stem cells to drive tissue regeneration. Healthy fat introduced into irradiated tissues appeared to reverse radiation injury (fibrosis, scarring, contracture and pain) clinically; a phenomenon since validated in several animal studies. In the quest to explain and enhance these therapeutic effects, adipose-derived stem cells (ADSCs) were suggested as playing a key role and techniques to enrich ADSCs in fat, in turn, followed. Stem cells - the body's rapid response 'road repair crew' - are on standby to combat tissue insults. ADSCs may exert influences either by releasing paracrine-signalling factors alone or as cell-free extracellular vesicles (EVs, exosomes). Alternatively, ADSCs may augment vital immune/inflammatory processes; or themselves differentiate into mature adipose cells to provide the 'building-blocks' for engineered tissue. Regardless, adipose tissue constitutes an ideal source for mesenchymal stem cells for therapeutic application, due to ease of harvest and processing; and a relative abundance of adipose tissue in most patients. Here, we review the clinical applications of fat grafting, ADSC-enhanced fat graft, fat stem cell therapy; and the latest evolution of EVs and nanoparticles in healing, cancer and neurodegenerative and multiorgan disease.
Collapse
Affiliation(s)
- Lipi Shukla
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia.,Department of Plastic Surgery, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Yinan Yuan
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia
| | - Ramin Shayan
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia.,Department of Plastic Surgery, St Vincent's Hospital, Fitzroy, VIC, Australia.,Plastic, Hand and Faciomaxillary Surgery Unit, Alfred Hospital, Prahran, VIC, Australia.,Department of Plastic and Reconstructive Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Tara Karnezis
- O'Brien Institute Department, St Vincent's Institute for Medical Research, Fitzroy, VIC, Australia
| |
Collapse
|
33
|
Can supernatant from immortalized adipose tissue MSC replace cell therapy? An in vitro study in chronic wounds model. Stem Cell Res Ther 2020; 11:29. [PMID: 31964417 PMCID: PMC6975034 DOI: 10.1186/s13287-020-1558-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/13/2019] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) secrete a cocktail of growth factors and cytokines, which could promote tissue regeneration and wound healing. Therefore, in clinical practice, post-culture MSC supernatant treatment could be a more attractive alternative to autologous stem cell transplantation. In this study, we compared the regenerative properties of supernatants harvested from four newly established human adipose tissue mesenchymal stem cell lines (HATMSCs) derived from chronic wound patients or healthy donors. Methods HATMSC supernatants were produced in a serum-free medium under hypoxia and their content was analyzed by a human angiogenesis antibody array. The regenerative effect of HATMSCs supernatants was investigated in an in vitro model of chronic wound, where cells originating from human skin, such as microvascular endothelial cells (HSkMEC.2), keratinocytes (HaCaT), and fibroblasts (MSU-1.1), were cultured in serum-free and oxygen-reduced conditions. The effect of supernatant treatment was evaluated using an MTT assay and light microscopy. In addition, fibroblasts and HATMSCs were labeled with PKH67 and PKH26 dye, respectively, and the effect of supernatant treatment was compared to that obtained when fibroblasts and HATMSCs were co-cultured, using flow cytometry and fluorescent microscopy. Results A wide panel of angiogenesis-associated cytokines such as angiogenin, growth-regulated oncogene (GRO), interleukin-6 and 8 (IL-6, IL-8), vascular endothelial growth factor (VEGF), insulin growth factor 1 (IGF-1), and matrix metalloproteinase (MMP) were found in all tested HATMSCs supernatants. Moreover, supernatant treatment significantly enhanced the survival of fibroblasts, endothelial cells, and keratinocytes in our chronic wound model in vitro. Importantly, we have shown that in in vitro settings, HATMSC supernatant treatment results in superior fibroblast proliferation than in the case of co-culture with HATMSCs. Conclusions Our results suggest that therapy based on bioactive factors released by the immortalized atMSC into supernatant has important effect on skin-derived cell proliferation and might preclude the need for a more expensive and difficult cell therapy approach to improve chronic wound healing.
Collapse
|
34
|
Luo X, Gao ZX, Lin SW, Tong ML, Liu LL, Lin LR, Ke WJ, Yang TC. Recombinant Treponema pallidum protein Tp0136 promotes fibroblast migration by modulating MCP-1/CCR2 through TLR4. J Eur Acad Dermatol Venereol 2020; 34:862-872. [PMID: 31856347 DOI: 10.1111/jdv.16162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/12/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Chancre self-healing is an important clinical feature in the early stages of syphilis infection. Wound healing may involve an important mechanism by the migration of fibroblasts filling the injured lesion. However, the specific mechanism underlying this process is still unknown. OBJECTIVES We aimed to analyse the role of Tp0136 in the migration of fibroblasts and the related mechanism. METHODS The migration ability of fibroblasts was detected by a wound-healing assay. RT-PCR and ELISA detected the expression of MCP-1, IL-6 and MMP-9. TLR4 expression was detected by RT-PCR. The protein levels of CCR2 and relevant signalling pathway molecules were measured by Western blotting. RESULTS Tp0136 significantly promoted fibroblast migration. Subsequently, the levels of MCP-1 and its receptor CCR2 were increased in this process. The migration of fibroblasts was significantly inhibited by an anti-MCP-1 neutralizing antibody or CCR2 inhibitors. Furthermore, studies demonstrated that Tp0136 could activate the ERK/JNK/PI3K/NF-κB signalling pathways through TLR4 activity and that signalling pathways inhibitors could weaken MCP-1 secretion and fibroblast migration. CONCLUSIONS These findings demonstrate that Tp0136 promotes the migration of fibroblasts by inducing MCP-1/CCR2 expression through signalling involving the TLR4, ERK, JNK, PI3K and NF-κB signalling pathways, which could contribute to the mechanism of chancre self-healing in syphilis.
Collapse
Affiliation(s)
- X Luo
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Z-X Gao
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - S-W Lin
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - M-L Tong
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China.,Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| | - L-L Liu
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China.,Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| | - L-R Lin
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China.,Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| | - W-J Ke
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - T-C Yang
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China.,Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
35
|
Li X, Kim J, Wu J, Ahamed AI, Wang Y, Martins-Green M. N-Acetyl-cysteine and Mechanisms Involved in Resolution of Chronic Wound Biofilm. J Diabetes Res 2020; 2020:9589507. [PMID: 32083136 PMCID: PMC7007959 DOI: 10.1155/2020/9589507] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/18/2019] [Accepted: 12/07/2019] [Indexed: 12/29/2022] Open
Abstract
Chronic wounds are a major global health problem with the presence of biofilm significantly contributing to wound chronicity. Current treatments are ineffective in resolving biofilm and simultaneously killing the bacteria; therefore, effective biofilm-resolving drugs are needed. We have previously shown that, together with α-tocopherol, N-acetyl-cysteine (NAC) significantly improves the healing of biofilm-containing chronic wounds, in a diabetic mouse model we developed, by causing disappearance of the bacteria and breakdown of the extracellular polymeric substance (EPS). We hypothesize that NAC creates a microenvironment that affects bacterial survival and EPS integrity. To test this hypothesis, we developed an in vitro biofilm system using microbiome taken directly from diabetic mouse chronic wounds. For these studies, we chose mice in which chronic wound microbiome was rich in Pseudomonas aeruginosa (97%). We show that NAC at concentrations with pH < pKa causes bacterial cell death and breakdown of EPS. When used before biofilm is formed, NAC leads to bacterial cell death whereas treatment after the biofilm is established NAC causes biofilm dismantling accompanied by bacterial cell death. Mechanistically, we show that NAC can penetrate the bacterial membrane, increase oxidative stress, and halt protein synthesis. We also show that low pH is important for the actions of NAC and that bacterial death occurs independently of the presence of biofilm. In addition, we show that both the acetyl and carboxylic groups play key roles in NAC functions. The results presented here provide insight into the mechanisms by which NAC dismantles biofilm and how it could be used to treat chronic wounds after debridement (NAC applied at the start of culture) or without debridement (NAC applied when biofilm is already formed). This approach can be taken to develop biofilm from microbiome taken directly from human chronic wounds to test molecules that could be effective for the treatment of specific biofilm compositions.
Collapse
Affiliation(s)
- Xin Li
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, USA
| | - Jane Kim
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, USA
| | - Jiabin Wu
- Department of Chemistry, University of California, Riverside, CA, USA
| | - Alaa' I Ahamed
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, USA
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, CA, USA
| | - Manuela Martins-Green
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, USA
| |
Collapse
|
36
|
Ekasaputra VM, Putra A, Muhar AM, Varessa J, Cikita RB, Wijaya SAP, Nazar MA. Mesenchymal Stem Cell-injected Omental Patch More Effective Promoting Wound Healing in Bowel Perforation Animal Model. Med Arch 2020; 74:332-336. [PMID: 33424084 PMCID: PMC7780763 DOI: 10.5455/medarh.2020.74.332-336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction: Bowel perforation (BP) occurs as the complication of many gastrointestinal problems. Omental patch (OP) is one of the methods to place omentum flaps in the perforated area. Mesenchymal stem cells (MSCs) may increase regeneration process in all tissues. Aim: to demonstrate the role of MSC in accelerating of wound healing process by analyzing fibroblast and collagen appearance in perforated bowel conditions. Methods: Using a BP rabbit model, 18 rabbit were randomly assigned into three groups: combination of umbilical cord (UC)-MSCs injection and OP (T1), OP only (T2) and vehicle control (Veh). Hematoxylin-eosin staining and Masson’s trichrome staining were performed to analyze the level of fibroblast and collagen. Wound length were measured using standardized caliper. Results: The study showed a significant (P<0.05) increase of fibroblast and collagen amount on T1 and T2, in which T1 was higher than T2. This result was also followed by the decrease of wound length. Conclusion: The combination of MSCs and OP-sutured in perforated bowel are better to accelerate wound healing than OP only in BP cases.
Collapse
Affiliation(s)
| | - Agung Putra
- Stem Cell and Cancer Research, Sultan Agung Islamic University, Semarang, Indonesia.,Department of Pathological Anatomy, Sultan Agung Islamic University, Semarang, Indonesia.,Department of Postgraduate Biomedical Science, Sultan Agung Islamic University, Semarang, Indonesia
| | - Adi Muradi Muhar
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | | | | | | | - Mohammad Ariq Nazar
- Stem Cell and Cancer Research, Sultan Agung Islamic University, Semarang, Indonesia
| |
Collapse
|
37
|
Mesdom P, Colle R, Lebigot E, Trabado S, Deflesselle E, Fève B, Becquemont L, Corruble E, Verstuyft C. Human Dermal Fibroblast: A Promising Cellular Model to Study Biological Mechanisms of Major Depression and Antidepressant Drug Response. Curr Neuropharmacol 2020; 18:301-318. [PMID: 31631822 PMCID: PMC7327943 DOI: 10.2174/1570159x17666191021141057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/15/2019] [Accepted: 10/19/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Human dermal fibroblasts (HDF) can be used as a cellular model relatively easily and without genetic engineering. Therefore, HDF represent an interesting tool to study several human diseases including psychiatric disorders. Despite major depressive disorder (MDD) being the second cause of disability in the world, the efficacy of antidepressant drug (AD) treatment is not sufficient and the underlying mechanisms of MDD and the mechanisms of action of AD are poorly understood. OBJECTIVE The aim of this review is to highlight the potential of HDF in the study of cellular mechanisms involved in MDD pathophysiology and in the action of AD response. METHODS The first part is a systematic review following PRISMA guidelines on the use of HDF in MDD research. The second part reports the mechanisms and molecules both present in HDF and relevant regarding MDD pathophysiology and AD mechanisms of action. RESULTS HDFs from MDD patients have been investigated in a relatively small number of works and most of them focused on the adrenergic pathway and metabolism-related gene expression as compared to HDF from healthy controls. The second part listed an important number of papers demonstrating the presence of many molecular processes in HDF, involved in MDD and AD mechanisms of action. CONCLUSION The imbalance in the number of papers between the two parts highlights the great and still underused potential of HDF, which stands out as a very promising tool in our understanding of MDD and AD mechanisms of action.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Céline Verstuyft
- Address correspondence to this author at the Laboratoire de Pharmacologie, Salle 416, Bâtiment Université, Hôpital du Kremlin Bicêtre, 78 rue du Général Leclerc, 94275 Le Kremlin-Bicêtre, France; Tel: +33145213588; E-mail:
| |
Collapse
|
38
|
Rahmanzade R. Redefinition of tumor capsule: Rho-dependent clustering of cancer-associated fibroblasts in favor of tensional homeostasis. Med Hypotheses 2019; 135:109425. [PMID: 31760246 DOI: 10.1016/j.mehy.2019.109425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/29/2019] [Accepted: 10/09/2019] [Indexed: 11/16/2022]
Abstract
Fibroblasts are the most frequent cells of the connective tissues. Having the ability to sense and respond to mechanical stimuli in addition to the biochemical ones makes them crucial for such a composite-like and tension-preserving tissue. Over the last decade, the investigation of the role of these cells in tumor progression was a hot topic of research in tumor biology. Literatures almost unanimously describe the re-education of stromal fibroblasts by tumor cells in favor of tumor progression, which resulted in the birth of a new nomenclature, the cancer-associated fibroblasts. On the other hand, some studies reported anti-tumor roles for these cells. Herein, author suggests that the previously described pro-migratory and pro-contractile contexts, which respectively results in divergent and convergent distribution of fibroblasts by changing Rho-Rac1 balance, could be applied for cancer-associated fibroblasts as well. Based on this proposed concept, stromal fibroblasts could represent different roles, either pro-tumor or anti-tumor, during the course of tumor progression. In the earlier phases, they tend to assemble along tumor-stroma interface in the form of tumor capsules in order to resist tumor growth and to maintain tensional homeostasis in stroma. But in later phases, after being chronically subjected to tumor-induced chemical and mechanical stimuli, they will gradually lose their substantial abilities to oppose tumor expansion and, in contrary, will promote tumorigenesis. In summary, this paper redefines tumor capsule from chemical and mechanical standpoints as Rho-dependent clustering of cancer-associated fibroblasts in favor of tensional homeostasis. Furthermore, it proposes that stromal fibroblasts will undergo some irreversible epigenetic changes in Rac1- and Rho-related proteins through tumor-stroma crosstalk, which irreversibly diminish their ability of capsule formation. Finally, the author discusses the possible researches helping us to assess the proposed concept and its clinical implications.
Collapse
Affiliation(s)
- Ramin Rahmanzade
- Biomedical Research & Training, University Hospital Basel, Mittlere Strasse 91, 4031 Basel, Switzerland.
| |
Collapse
|
39
|
Critical role of the endogenous renin-angiotensin system in maintaining self-renewal and regeneration potential of epidermal stem cells. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2647-2656. [DOI: 10.1016/j.bbadis.2019.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 12/11/2022]
|
40
|
Advanced drug delivery systems and artificial skin grafts for skin wound healing. Adv Drug Deliv Rev 2019; 146:209-239. [PMID: 30605737 DOI: 10.1016/j.addr.2018.12.014] [Citation(s) in RCA: 340] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/27/2018] [Accepted: 12/27/2018] [Indexed: 12/14/2022]
Abstract
Cutaneous injuries, especially chronic wounds, burns, and skin wound infection, require painstakingly long-term treatment with an immense financial burden to healthcare systems worldwide. However, clinical management of chronic wounds remains unsatisfactory in many cases. Various strategies including growth factor and gene delivery as well as cell therapy have been used to enhance the healing of non-healing wounds. Drug delivery systems across the nano, micro, and macroscales can extend half-life, improve bioavailability, optimize pharmacokinetics, and decrease dosing frequency of drugs and genes. Replacement of the damaged skin tissue with substitutes comprising cell-laden scaffold can also restore the barrier and regulatory functions of skin at the wound site. This review covers comprehensively the advanced treatment strategies to improve the quality of wound healing.
Collapse
|
41
|
Lasocka I, Jastrzębska E, Szulc-Dąbrowska L, Skibniewski M, Pasternak I, Kalbacova MH, Skibniewska EM. The effects of graphene and mesenchymal stem cells in cutaneous wound healing and their putative action mechanism. Int J Nanomedicine 2019; 14:2281-2299. [PMID: 31015759 PMCID: PMC6448540 DOI: 10.2147/ijn.s190928] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This study provides a review of the therapeutic potential of graphene dressing scaffolds and mesenchymal stem cells (MSCs) and their synergistic effects with respect to cutaneous wound healing. This study also considers their putative action mechanism based on the antibacterial, immunomodulating, angiogenic, matrix remodeling effects of materials belonging to the graphene family and MSCs during the wound healing process. In addition, this study discusses the cytocompatibility of graphene, its uses as a platform for skin substitutes, the properties it possesses with respect to providing protection against microbial invasion as well as strategies aimed at minimizing the chance of the occurrence of sepsis. MSCs are capable of secreting several factors that exert a therapeutic impact on reparative processes and tissue regeneration. In light of experiments conducted to date, graphene combined with MSCs appears to have the potential to enhance both the wound healing process and infection control at the injury site.
Collapse
Affiliation(s)
- Iwona Lasocka
- Department of Animal Environment Biology, Faculty of Animal Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Elżbieta Jastrzębska
- Department of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Lidia Szulc-Dąbrowska
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Michał Skibniewski
- Department of Morphological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland,
| | - Iwona Pasternak
- Faculty of Physics, Warsaw University of Technology, Warsaw, Poland
| | - Marie Hubalek Kalbacova
- Institute of Pathological Physiology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic,
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic,
| | - Ewa M Skibniewska
- Department of Animal Environment Biology, Faculty of Animal Science, Warsaw University of Life Sciences, Warsaw, Poland
| |
Collapse
|
42
|
Autologous bone marrow-derived cells for venous leg ulcers treatment: a pilot study. Cytotherapy 2019; 21:189-199. [PMID: 30700393 DOI: 10.1016/j.jcyt.2019.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/30/2018] [Accepted: 01/08/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Chronic venous leg ulcers (VLUs) are a common problem in clinical practice and available treatments are not satisfactory. The use of adjuvant therapies in combination with lower limb compression may lead to improved healing rates. Chronic wounds are candidates for new strategies in the emergent field of regenerative medicine. Bone marrow-derived cells (BMDCs) contain cells and secrete cytokines known to participate in wound healing. Thus, BMDC therapy seems a logical strategy for the treatment of chronic wounds. Our objective was to evaluate feasibility, safety and initial clinical outcome of autologous BMDC therapy associated with standard treatment in patients with VLUs. METHODS We conducted an open-label, single-arm, prospective pilot clinical trial in four patients with six chronic VLUs. The study protocol was approved by the institutional and national review boards and ethics committees. Bone marrow was harvest, processed and then administered by multiple injections into the ulcers. All patients received standard treatment and non-healing characteristics of the VLUs were confirmed at study entry. RESULTS Ulcer size and wound pain evaluated 12 months after BMDC treatment were significantly reduced (P < 0.05). BMDC treatment was safe and well tolerated in long-term follow-up. DISCUSSION Despite the low number of patients studied, our results showed that autologous BMDC treatment could be a useful, feasible and safe procedure to enhance ulcer healing. However, randomized controlled trials with more patients are needed to address this question and translate this approach into clinical practice.
Collapse
|
43
|
Ma T, Fu B, Yang X, Xiao Y, Pan M. Adipose mesenchymal stem cell-derived exosomes promote cell proliferation, migration, and inhibit cell apoptosis via Wnt/β-catenin signaling in cutaneous wound healing. J Cell Biochem 2019; 120:10847-10854. [PMID: 30681184 DOI: 10.1002/jcb.28376] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/10/2019] [Indexed: 12/15/2022]
Abstract
Cutaneous wounds, a type of soft tissue injury, are difficult to heal in aging. Differentiation, migration, proliferation, and apoptosis of skin cells are identified as key factors during wound healing processes. Mesenchymal stem cells have been documented as possible candidates for wound healing treatment because their use could augment the regenerative capacity of many tissues. However, the effects of exosomes derived from adipose-derived stem cell (ADSC-exos) on cutaneous wound healing remain to be carefully elucidated. In this present study, HaCaT cells were exposed to hydrogen peroxide (H2 O 2 ) for the establishment of the skin lesion model. Cell Counting Kit-8 assay, migration assay, and flow cytometry assay were conducted to detect the biological function of ADSC-exos in skin lesion model. Finally, the possible mechanism was further investigated using Western blot assay. The successful construction of the skin lesion model was confirmed by results of the enhanced cell apoptosis of HaCaT cells induced by H 2 O 2 , the increased Bax expression and decreased Bcl-2 expression. CD9 and CD63 expression evidenced the existence of ADSC-exos. The results of functional experiments demonstrated that ADSC-exos could prompt cell proliferation and migration of HaCaT cells, and repress cell apoptosis of HaCaT cells. In addition, the activation of Wnt/β-catenin signaling was confirmed by the enhanced expression of β-catenin at the protein level. Collectively, our findings suggest that ADSC-exos play a positive role in cutaneous wound healing possibly via Wnt/β-catenin signaling. Our study may provide new insights into the therapeutic target for cutaneous wound healing.
Collapse
Affiliation(s)
- Tao Ma
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Bingchuan Fu
- Department of Medical Cosmetology, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yang
- Department of Otolaryngology, The 161th Hospital of the Chinese People's Liberation Army, Wuhan, Hubei, China
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Mengxiong Pan
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
44
|
Hassanshahi A, Hassanshahi M, Khabbazi S, Hosseini‐Khah Z, Peymanfar Y, Ghalamkari S, Su Y, Xian CJ. Adipose‐derived stem cells for wound healing. J Cell Physiol 2018; 234:7903-7914. [DOI: 10.1002/jcp.27922] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/24/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Alireza Hassanshahi
- Department of Genetics Faculty of Basic Sciences, Islamic Azad University Shahrekord Iran
| | - Mohammadhossein Hassanshahi
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia Adelaide South Australia Australia
| | - Samira Khabbazi
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia Adelaide South Australia Australia
| | - Zahra Hosseini‐Khah
- Department of Immunology School of Medicine, Mazandaran University of Medical Sciences Sari Iran
| | - Yaser Peymanfar
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia Adelaide South Australia Australia
| | | | - Yu‐Wen Su
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia Adelaide South Australia Australia
| | - Cory J. Xian
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia Adelaide South Australia Australia
| |
Collapse
|
45
|
Cooper DR, Wang C, Patel R, Trujillo A, Patel NA, Prather J, Gould LJ, Wu MH. Human Adipose-Derived Stem Cell Conditioned Media and Exosomes Containing MALAT1 Promote Human Dermal Fibroblast Migration and Ischemic Wound Healing. Adv Wound Care (New Rochelle) 2018; 7:299-308. [PMID: 30263873 PMCID: PMC6158770 DOI: 10.1089/wound.2017.0775] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/08/2018] [Indexed: 12/15/2022] Open
Abstract
Objective: Chronically ill patients heal recalcitrant ulcerative wounds more slowly. Human adipose-derived stem cells (hADSCs) play an important role in tissue regeneration and exosomes secreted by hADSC contribute to their paracrine signaling. In addition to cytokines, lipids and growth factors, hADSC secrete mRNA, miRNA, and long noncoding (lnc) RNA into exosomes. In this study we examined the role of lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1), an abundant lncRNA in exosomes from conditioned media (CM), on cell migration and ischemic wound healing. Approach: CM and isolated exosomes from hADSC were applied to human dermal fibroblast (HDF) in scratch assays and electric cell-substrate impedance sensing (ECIS) assays. CM was also applied to a rat model of ischemic wound healing and wound closure was followed. Results: CM stimulated cell migration of HDFs in vitro by 48%. CM stimulated the closure of ischemic wounds in a rat model 50% faster than unconditioned media. The depletion of MALAT1 in adipose-derived stem cell (ADSC) CM significantly reduced cell migration. Since MALAT1 is secreted into exosomes, a purified population of exosomes was applied to HDF where they enhanced cell migration in a similar manner to FGF-2 or basic fibroblast growth factor (bFGF) in ECIS wound healing assays. The uptake of exosomes by HDF was shown using dynasore, an inhibitor that blocks clathrin- and caveolin-dependent endocytosis. Depletion of MALAT1 in hADSC with antisense oligonucleotides resulted in exosomes without MALAT1. These exosomes had an effect similar to the unconditioned, control media in ECIS assays. Innovation: Exosomes contain lncRNA MALAT1 and other factors that have the potential to stimulate HDF cell migration and angiogenesis involved in wound healing without applying stem cells to wounds. Conclusion: Our results show the potential of using topically applied ADSC-derived exosomes containing MALAT1 for treating ischemic wounds. This allows for harnessing the power of stem cell paracrine signaling capabilities without applying the cells.
Collapse
Affiliation(s)
- Denise R. Cooper
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Chunyan Wang
- Department of Physiology and Pharmacology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Rehka Patel
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Andrea Trujillo
- Department of Physiology and Pharmacology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Niketa A. Patel
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Jamie Prather
- Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Lisa J. Gould
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Physiology and Pharmacology, University of South Florida Morsani College of Medicine, Tampa, Florida
- Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Mack H. Wu
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
- Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| |
Collapse
|
46
|
Development and Characterisation of a Human Chronic Skin Wound Cell Line-Towards an Alternative for Animal Experimentation. Int J Mol Sci 2018; 19:ijms19041001. [PMID: 29584680 PMCID: PMC5979489 DOI: 10.3390/ijms19041001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 12/30/2022] Open
Abstract
Background: Chronic skin wounds are a growing financial burden for healthcare providers, causing discomfort/immobility to patients. Whilst animal chronic wound models have been developed to allow for mechanistic studies and to develop/test potential therapies, such systems are not good representations of the human chronic wound state. As an alternative, human chronic wound fibroblasts (CWFs) have permitted an insight into the dysfunctional cellular mechanisms that are associated with these wounds. However, such cells strains have a limited replicative lifespan and therefore a limited reproducibility/usefulness. Objectives: To develop/characterise immortalised cell lines of CWF and patient-matched normal fibroblasts (NFs). Methods and Results: Immortalisation with human telomerase resulted in both CWF and NF proliferating well beyond their replicative senescence end-point (respective cell strains senesced as normal). Gene expression analysis demonstrated that, whilst proliferation-associated genes were up-regulated in the cell lines (as would be expected), the immortalisation process did not significantly affect the disease-specific genotype. Immortalised CWF (as compared to NF) also retained a distinct impairment in their wound repopulation potential (in line with CWF cell strains). Conclusions: These novel CWF cell lines are a credible animal alternative and could be a valuable research tool for understanding both the aetiology of chronic skin wounds and for therapeutic pre-screening.
Collapse
|
47
|
Schreier C, Rothmiller S, Scherer MA, Rummel C, Steinritz D, Thiermann H, Schmidt A. Mobilization of human mesenchymal stem cells through different cytokines and growth factors after their immobilization by sulfur mustard. Toxicol Lett 2018; 293:105-111. [PMID: 29426001 DOI: 10.1016/j.toxlet.2018.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/19/2018] [Accepted: 02/05/2018] [Indexed: 01/28/2023]
Abstract
INTRODUCTION The chemical warfare agent sulfur mustard (SM), also known as mustard gas, was first used in World War I. Although prohibited by the chemical warfare convention, significant amounts of SM still exist and have still to be regarded as a threat for military personnel and civilians. After SM exposure, the most prominent clinical symptom is the development of extensive non-healing skin wounds. This chronic wound healing dysfunction is persisting over long time. Mesenchymal stem cells (MSC) are known to play an important role in wound healing. Moreover, it is also known that patients with chronic wound healing diseases have compromised mesenchymal stem cell functionality. Based on these observations and the known relationship between wound healing dysfunction and MSC function we investigated the impact of sulfur mustard on human MSC. MATERIAL & METHODS Mesenchymal stem cells (MSC) were isolated from femoral heads of healthy donors. They were cultured for less than four passages. MSC were exposed towards different sulfur mustard concentrations. After exposure we analyzed the secretome and the migration capacity. The migration capacity under influence of SM was analyzed after treatment with various cytokines. RESULTS SM exposure (even at very low concentrations) showed negative effects on the migration capability. Many cytokines that are necessary for MSC migration were secreted in a reduced manner. The reduced migratory capacity can be compensated in part by the addition of cytokines. Here especially IL-8 (e and m) and IL-6 significantly compensated the SM induced migration reduction. DISCUSSION The effect of sulfur mustard on MSC might play an important role in the persistence of long-term adverse effects; here the reduced migration could particularly be important. The compensation of the SM-induced migration reduction by addition of cytokines could possibly solve this problem. Moreover, our current results will help to understand the relationship between alkylating agents and MSC and thus will also give guidance in the future perspective for the therapeutic use of MSC in patients suffering from sulfur mustard induced chronic skin wounds.
Collapse
Affiliation(s)
- Cassandra Schreier
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 Munich, Germany
| | - Simone Rothmiller
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 Munich, Germany
| | - Michael A Scherer
- Department of Traumatology and Orthopedics, HELIOS Amper Clinics, Krankenhausstrasse 15, 85221 Dachau, Germany
| | - Christoph Rummel
- Wolfart Clinic, Department of Orthopedics and Sports Medicine, Waldstrasse 7, 82166 Gräfelfing, Germany
| | - Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 Munich, Germany; Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Goethestr. 33, 80336 Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 Munich, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937 Munich, Germany; Universität der Bundeswehr, Fakultät für Humanwissenschaften, Department für Sportwissenschaft, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany.
| |
Collapse
|
48
|
Peix L, Evans IC, Pearce DR, Simpson JK, Maher TM, McAnulty RJ. Diverse functions of clusterin promote and protect against the development of pulmonary fibrosis. Sci Rep 2018; 8:1906. [PMID: 29382921 PMCID: PMC5789849 DOI: 10.1038/s41598-018-20316-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/16/2018] [Indexed: 01/06/2023] Open
Abstract
Pulmonary fibrosis is a progressive scarring disorder of the lung with dismal prognosis and no curative therapy. Clusterin, an extracellular chaperone and regulator of cell functions, is reduced in bronchoalveolar lavage fluid of patients with pulmonary fibrosis. However, its distribution and role in normal and fibrotic human lung are incompletely characterized. Immunohistochemical localization of clusterin revealed strong staining associated with fibroblasts in control lung and morphologically normal areas of fibrotic lung but weak or undetectable staining in fibrotic regions and particularly fibroblastic foci. Clusterin also co-localized with elastin in vessel walls and additionally with amorphous elastin deposits in fibrotic lung. Analysis of primary lung fibroblast isolates in vitro confirmed the down-regulation of clusterin expression in fibrotic compared with control lung fibroblasts and further demonstrated that TGF-β1 is capable of down-regulating fibroblast clusterin expression. shRNA-mediated down-regulation of clusterin did not affect TGF-β1-induced fibroblast-myofibroblast differentiation but inhibited fibroblast proliferative responses and sensitized to apoptosis. Down-regulation of clusterin in fibrotic lung fibroblasts at least partly due to increased TGF-β1 may therefore represent an appropriate but insufficient response to limit fibroproliferation. Reduced expression of clusterin in the lung may also limit its extracellular chaperoning activity contributing to dysregulated deposition of extracellular matrix proteins.
Collapse
Affiliation(s)
- Lizzy Peix
- UCL Respiratory Centre for Inflammation and Tissue Repair, Rayne Building, University College London, London, WC1E 6JF, UK
- GlaxoSmithKline, Stevenage, UK
| | - Iona C Evans
- UCL Respiratory Centre for Inflammation and Tissue Repair, Rayne Building, University College London, London, WC1E 6JF, UK
- UCL Institute for Woman's Health, University College London, London, UK
| | - David R Pearce
- UCL Respiratory Centre for Inflammation and Tissue Repair, Rayne Building, University College London, London, WC1E 6JF, UK
| | | | - Toby M Maher
- NIHR Respiratory Biomedical Research Unit, Royal Brompton Hospital, London, UK
- Fibrosis Research Group, National Heart and Lung Institute, Imperial College, London, UK
| | - Robin J McAnulty
- UCL Respiratory Centre for Inflammation and Tissue Repair, Rayne Building, University College London, London, WC1E 6JF, UK.
| |
Collapse
|
49
|
Geraili A, Jafari P, Hassani MS, Araghi BH, Mohammadi MH, Ghafari AM, Tamrin SH, Modarres HP, Kolahchi AR, Ahadian S, Sanati-Nezhad A. Controlling Differentiation of Stem Cells for Developing Personalized Organ-on-Chip Platforms. Adv Healthc Mater 2018; 7. [PMID: 28910516 DOI: 10.1002/adhm.201700426] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/01/2017] [Indexed: 01/09/2023]
Abstract
Organ-on-chip (OOC) platforms have attracted attentions of pharmaceutical companies as powerful tools for screening of existing drugs and development of new drug candidates. OOCs have primarily used human cell lines or primary cells to develop biomimetic tissue models. However, the ability of human stem cells in unlimited self-renewal and differentiation into multiple lineages has made them attractive for OOCs. The microfluidic technology has enabled precise control of stem cell differentiation using soluble factors, biophysical cues, and electromagnetic signals. This study discusses different tissue- and organ-on-chip platforms (i.e., skin, brain, blood-brain barrier, bone marrow, heart, liver, lung, tumor, and vascular), with an emphasis on the critical role of stem cells in the synthesis of complex tissues. This study further recaps the design, fabrication, high-throughput performance, and improved functionality of stem-cell-based OOCs, technical challenges, obstacles against implementing their potential applications, and future perspectives related to different experimental platforms.
Collapse
Affiliation(s)
- Armin Geraili
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
| | - Parya Jafari
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
- Department of Electrical Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohsen Sheikh Hassani
- Department of Systems and Computer Engineering; Carleton University; 1125 Colonel By Drive Ottawa K1S 5B6 ON Canada
| | - Behnaz Heidary Araghi
- Department of Materials Science and Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Mohammad Ghafari
- Department of Stem Cells and Developmental Biology; Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology; Tehran 16635-148 Iran
| | - Sara Hasanpour Tamrin
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Ahmad Rezaei Kolahchi
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
- Center for Bioengineering Research and Education; Biomedical Engineering Program; University of Calgary; Calgary T2N 1N4 AB Canada
| |
Collapse
|
50
|
Ebnerasuly F, Hajebrahimi Z, Tabaie SM, Darbouy M. Effect of Simulated Microgravity Conditions on Differentiation of Adipose Derived Stem Cells towards Fibroblasts Using Connective Tissue Growth Factor. IRANIAN JOURNAL OF BIOTECHNOLOGY 2017; 15:241-251. [PMID: 29845076 DOI: 10.15171/ijb.1747] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 06/14/2017] [Accepted: 07/08/2017] [Indexed: 12/12/2022]
Abstract
Background: Mesenchymal stem cells (MSCs) are multipotent cells able to differentiating into a variety of mesenchymal tissues including osteoblasts, adipocytes and several other tissues. Objectives: Differentiation of MSCs into fibroblast cells in vitro is an attractive strategy to achieve fibroblast cell and use them for purposes such as regeneration medicine. The goal of this study was investigate the simulated microgravity effect on differentiation of Adipose Derived Stem Cells (ADSCs) to fibroblasts. Materials and Methods: To fibroblast differentiation 100 ng.mL-1 of connective tissue growth factor (CTGF), and for simulation microgravity, 2D clinostat was used. After isolation the human ADSCs from adipose, cells were passaged, and at passages 3 they were used for characterization and subsequent steps. After 7 days of CTGF and simulated microgravity treatment, proliferation, and differentiation were analyzed collectively by MTT assay, quantitative PCR analyses, and Immunocytochemistry staining. Results: MTT assay revealed that CTGF stimulate the proliferation but simulated microgravity didn't have statistically significant effect on cell proliferation. In RNA level the expression of these genes are investigated: collagen type I (COLI), elastin (ELA), collagen type III (ColIII), Matrix Metalloproteinases I(MMP1), Fibronectin 1 (FN1), CD44, Fibroblast Specific protein (FSP-1), Integrin Subunit Beta 1 (ITGB1), Vimentin (VIM) and Fibrillin (FBN). We found that expression of ELN, FN1, FSP1, COL1A1, ITGB1, MMP1 and COL3A1 in both condition, and VIM and FBN1 just in differentiation medium in normal gravity increased. In protein level the expression of COL III and ELN in simulated microgravity increased. Conclusions: These findings collectively demonstrate that the simulated microgravity condition alters the marker fibroblast gene expression in fibroblast differentiation process.
Collapse
Affiliation(s)
- Farid Ebnerasuly
- Department of Biology, Fars Science and Research Branch , Islamic Azad University, Marvdasht, Iran.,Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Zahra Hajebrahimi
- Aerospace Research Institute, Ministry of Science Research and Technology, Tehran, Iran
| | - Seyed Mehdi Tabaie
- Medical Laser Research Center, Iranian Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Mojtaba Darbouy
- Department of Biology, Fars Science and Research Branch , Islamic Azad University, Marvdasht, Iran.,Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| |
Collapse
|