1
|
Sieńko D, Szabłowska-Gadomska I, Nowak-Szwed A, Rudziński S, Gofron M, Zygmunciak P, Lewandowska-Szumieł M, Zgliczyński WS, Czupryniak L, Mrozikiewicz-Rakowska B. The Potential of Mesenchymal Stem/Stromal Cells in Diabetic Wounds and Future Directions for Research and Therapy-Is It Time for Use in Everyday Practice? Int J Mol Sci 2024; 25:12171. [PMID: 39596237 PMCID: PMC11594847 DOI: 10.3390/ijms252212171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The treatment of diabetic wounds is impaired by the intricate nature of diabetes and its associated complications, necessitating novel strategies. The utilization of mesenchymal stem/stromal cells (MSCs) as a therapeutic modality for chronic and recalcitrant wounds in diabetic patients is an active area of investigation aimed at enhancing its therapeutic potential covering tissue regeneration. The threat posed to the patient and their environment by the presence of a diabetic foot ulcer (DFU) is so significant that any additional therapeutic approach that opens new pathways to halt the progression of local changes, which subsequently lead to a generalized inflammatory process, offers a chance to reduce the risk of amputation or even death. This article explores the potential of MSCs in diabetic foot ulcer treatment, examining their mechanisms of action, clinical application challenges, and future directions for research and therapy.
Collapse
Affiliation(s)
- Damian Sieńko
- Department of Diabetology and Internal Diseases, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (A.N.-S.); (L.C.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Ilona Szabłowska-Gadomska
- Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097 Warsaw, Poland; (I.S.-G.); (S.R.); (M.L.-S.)
| | - Anna Nowak-Szwed
- Department of Diabetology and Internal Diseases, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (A.N.-S.); (L.C.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Stefan Rudziński
- Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097 Warsaw, Poland; (I.S.-G.); (S.R.); (M.L.-S.)
| | - Maksymilian Gofron
- Department of Urology, Municipal Complex Hospital, 42-200 Czestochowa, Poland;
| | - Przemysław Zygmunciak
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland; (P.Z.); (W.S.Z.)
| | - Małgorzata Lewandowska-Szumieł
- Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097 Warsaw, Poland; (I.S.-G.); (S.R.); (M.L.-S.)
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Wojciech Stanisław Zgliczyński
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland; (P.Z.); (W.S.Z.)
| | - Leszek Czupryniak
- Department of Diabetology and Internal Diseases, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (A.N.-S.); (L.C.)
| | - Beata Mrozikiewicz-Rakowska
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland; (P.Z.); (W.S.Z.)
| |
Collapse
|
2
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
3
|
Chocarro-Wrona C, López de Andrés J, Rioboó-Legaspi P, Pleguezuelos-Beltrán P, Antich C, De Vicente J, Gálvez-Martín P, López-Ruiz E, Marchal JA. Design and evaluation of a bilayered dermal/hypodermal 3D model using a biomimetic hydrogel formulation. Biomed Pharmacother 2024; 177:117051. [PMID: 38959608 DOI: 10.1016/j.biopha.2024.117051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024] Open
Abstract
Due to the limitations of the current skin wound treatments, it is highly valuable to have a wound healing formulation that mimics the extracellular matrix (ECM) and mechanical properties of natural skin tissue. Here, a novel biomimetic hydrogel formulation has been developed based on a mixture of Agarose-Collagen Type I (AC) combined with skin ECM-related components: Dermatan sulfate (DS), Hyaluronic acid (HA), and Elastin (EL) for its application in skin tissue engineering (TE). Different formulations were designed by combining AC hydrogels with DS, HA, and EL. Cell viability, hemocompatibility, physicochemical, mechanical, and wound healing properties were investigated. Finally, a bilayered hydrogel loaded with fibroblasts and mesenchymal stromal cells was developed using the Ag-Col I-DS-HA-EL (ACDHE) formulation. The ACDHE hydrogel displayed the best in vitro results and acceptable physicochemical properties. Also, it behaved mechanically close to human native skin and exhibited good cytocompatibility. Environmental scanning electron microscopy (ESEM) analysis revealed a porous microstructure that allows the maintenance of cell growth and ECM-like structure production. These findings demonstrate the potential of the ACDHE hydrogel formulation for applications such as an injectable hydrogel or a bioink to create cell-laden structures for skin TE.
Collapse
Affiliation(s)
- Carlos Chocarro-Wrona
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, Granada 18016, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada 18012, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada 18016, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada 18016, Spain; BioFab i3D, Biofabrication and 3D (bio)printing laboratory, Granada 18016, Spain
| | - Julia López de Andrés
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, Granada 18016, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada 18012, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada 18016, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada 18016, Spain; BioFab i3D, Biofabrication and 3D (bio)printing laboratory, Granada 18016, Spain
| | - Pablo Rioboó-Legaspi
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, Granada 18016, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada 18012, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada 18016, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada 18016, Spain; BioFab i3D, Biofabrication and 3D (bio)printing laboratory, Granada 18016, Spain
| | - Paula Pleguezuelos-Beltrán
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, Granada 18016, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada 18012, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada 18016, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada 18016, Spain; BioFab i3D, Biofabrication and 3D (bio)printing laboratory, Granada 18016, Spain
| | - Cristina Antich
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, Granada 18016, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada 18012, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada 18016, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada 18016, Spain; BioFab i3D, Biofabrication and 3D (bio)printing laboratory, Granada 18016, Spain; National Center for Advancing Translational Sciences, National Institute of Health, Rockville, MD 28050, United States
| | - Juan De Vicente
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada 18016, Spain; F2N2Lab, Magnetic Soft Matter Group, Department of Applied Physics, Faculty of Sciences, University of Granada, Granada 18071, Spain
| | | | - Elena López-Ruiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, Granada 18016, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada 18012, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada 18016, Spain; BioFab i3D, Biofabrication and 3D (bio)printing laboratory, Granada 18016, Spain; Department of Health Sciences, University of Jaén, Jaén 23071, Spain.
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, Granada 18016, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada 18012, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada 18016, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada 18016, Spain; BioFab i3D, Biofabrication and 3D (bio)printing laboratory, Granada 18016, Spain.
| |
Collapse
|
4
|
Li J, Liu Y, Zhang R, Yang Q, Xiong W, He Y, Ye Q. Insights into the role of mesenchymal stem cells in cutaneous medical aesthetics: from basics to clinics. Stem Cell Res Ther 2024; 15:169. [PMID: 38886773 PMCID: PMC11184751 DOI: 10.1186/s13287-024-03774-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
With the development of the economy and the increasing prevalence of skin problems, cutaneous medical aesthetics are gaining more and more attention. Skin disorders like poor wound healing, aging, and pigmentation have an impact not only on appearance but also on patients with physical and psychological issues, and even impose a significant financial burden on families and society. However, due to the complexities of its occurrence, present treatment options cannot produce optimal outcomes, indicating a dire need for new and effective treatments. Mesenchymal stem cells (MSCs) and their secretomics treatment is a new regenerative medicine therapy that promotes and regulates endogenous stem cell populations and/or replenishes cell pools to achieve tissue homeostasis and regeneration. It has demonstrated remarkable advantages in several skin-related in vivo and in vitro investigations, aiding in the improvement of skin conditions and the promotion of skin aesthetics. As a result, this review gives a complete description of recent scientific breakthroughs in MSCs for skin aesthetics and the limitations of their clinical applications, aiming to provide new ideas for future research and clinical transformation.
Collapse
Affiliation(s)
- Junyi Li
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Liu
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Zhang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianyu Yang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Xiong
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430030, China.
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
5
|
Bicer M. Revolutionizing dermatology: harnessing mesenchymal stem/stromal cells and exosomes in 3D platform for skin regeneration. Arch Dermatol Res 2024; 316:242. [PMID: 38795200 PMCID: PMC11127839 DOI: 10.1007/s00403-024-03055-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/09/2024] [Accepted: 04/26/2024] [Indexed: 05/27/2024]
Abstract
Contemporary trends reveal an escalating interest in regenerative medicine-based interventions for addressing refractory skin defects. Conventional wound healing treatments, characterized by high costs and limited efficacy, necessitate a more efficient therapeutic paradigm to alleviate the economic and psychological burdens associated with chronic wounds. Mesenchymal stem/stromal cells (MSCs) constitute cell-based therapies, whereas cell-free approaches predominantly involve the utilization of MSC-derived extracellular vesicles or exosomes, both purportedly safe and effective. Exploiting the impact of MSCs by paracrine signaling, exosomes have emerged as a novel avenue capable of positively impacting wound healing and skin regeneration. MSC-exosomes confer several advantages, including the facilitation of angiogenesis, augmentation of cell proliferation, elevation of collagen production, and enhancement of tissue regenerative capacity. Despite these merits, challenges persist in clinical applications due to issues such as poor targeting and facile removal of MSC-derived exosomes from skin wounds. Addressing these concerns, a three-dimensional (3D) platform has been implemented to emend exosomes, allowing for elevated levels, and constructing more stable granules possessing distinct therapeutic capabilities. Incorporating biomaterials to encapsulate MSC-exosomes emerges as a favorable approach, concentrating doses, achieving intended therapeutic effectiveness, and ensuring continual release. While the therapeutic potential of MSC-exosomes in skin repair is broadly recognized, their application with 3D biomaterial scenarios remains underexplored. This review synthesizes the therapeutic purposes of MSCs and exosomes in 3D for the skin restoration, underscoring their promising role in diverse dermatological conditions. Further research may establish MSCs and their exosomes in 3D as a viable therapeutic option for various skin conditions.
Collapse
Affiliation(s)
- Mesude Bicer
- Department of Bioengineering, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, 38080, Turkey.
| |
Collapse
|
6
|
Wang M, Zhao J, Li J, Meng M, Zhu M. Insights into the role of adipose-derived stem cells and secretome: potential biology and clinical applications in hypertrophic scarring. Stem Cell Res Ther 2024; 15:137. [PMID: 38735979 PMCID: PMC11089711 DOI: 10.1186/s13287-024-03749-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024] Open
Abstract
Scar tissue is the inevitable result of repairing human skin after it has been subjected to external destructive stimuli. It leads to localized damage to the appearance of the skin, accompanied by symptoms such as itching and pain, which reduces the quality of life of the patient and causes serious medical burdens. With the continuous development of economy and society, there is an increasing demand for beauty. People are looking forward to a safer and more effective method to eliminate pathological scarring. In recent years, adipose-derived stem cells (ADSCs) have received increasing attention from researchers. It can effectively improve pathological scarring by mediating inflammation, regulating fibroblast proliferation and activation, and vascular reconstruction. This review focuses on the pathophysiological mechanisms of hypertrophic scarring, summarizing the therapeutic effects of in vitro, in vivo, and clinical studies on the therapeutic effects of ADSCs in the field of hypertrophic scarring prevention and treatment, the latest application techniques, such as cell-free therapies utilizing ADSCs, and discussing the advantages and limitations of ADSCs. Through this review, we hope to further understand the characterization of ADSC and clarify the effectiveness of its application in hypertrophic scarring treatment, so as to provide clinical guidance.
Collapse
Affiliation(s)
- Menglin Wang
- Department of Plastic Surgery, The First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Xigang District, Dalian, 116011, China
| | - Jianyu Zhao
- Department of Orthopaedics, The First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Xigang District, Dalian, 116011, China
| | - Jiacheng Li
- Department of Plastic Surgery, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Meng Meng
- Department of Orthopaedics, The First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Xigang District, Dalian, 116011, China.
| | - Mengru Zhu
- Department of Plastic Surgery, The First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Xigang District, Dalian, 116011, China.
| |
Collapse
|
7
|
Lin Z, Shibuya Y, Imai Y, Oshima J, Sasaki M, Sasaki K, Aihara Y, Khanh VC, Sekido M. Therapeutic Potential of Adipose-Derived Stem Cell-Conditioned Medium and Extracellular Vesicles in an In Vitro Radiation-Induced Skin Injury Model. Int J Mol Sci 2023; 24:17214. [PMID: 38139042 PMCID: PMC10743562 DOI: 10.3390/ijms242417214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Radiotherapy (RT) is one of three major treatments for malignant tumors, and one of its most common side effects is skin and soft tissue injury. However, the treatment of these remains challenging. Several studies have shown that mesenchymal stem cell (MSC) treatment enhances skin wound healing. In this study, we extracted human dermal fibroblasts (HDFs) and adipose-derived stem cells (ADSCs) from patients and generated an in vitro radiation-induced skin injury model with HDFs to verify the effect of conditioned medium derived from adipose-derived stem cells (ADSC-CM) and extracellular vesicles derived from adipose-derived stem cells (ADSC-EVs) on the healing of radiation-induced skin injury. The results showed that collagen synthesis was significantly increased in wounds treated with ADSC-CM or ADSC-EVs compared with the control group, which promoted the expression of collagen-related genes and suppressed the expression of inflammation-related genes. These findings indicated that treatment with ADSC-CM or ADSC-EVs suppressed inflammation and promoted extracellular matrix deposition; treatment with ADSC-EVs also promoted fibroblast proliferation. In conclusion, these results demonstrate the effectiveness of ADSC-CM and ADSC-EVs in the healing of radiation-induced skin injury.
Collapse
Affiliation(s)
- Zhixiang Lin
- Department of Plastic and Reconstructive Surgery, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan (Y.I.); (M.S.)
| | - Yoichiro Shibuya
- Department of Plastic and Reconstructive Surgery, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan (Y.I.); (M.S.)
| | - Yukiko Imai
- Department of Plastic and Reconstructive Surgery, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan (Y.I.); (M.S.)
| | - Junya Oshima
- Department of Plastic and Reconstructive Surgery, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan (Y.I.); (M.S.)
| | - Masahiro Sasaki
- Department of Plastic and Reconstructive Surgery, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan (Y.I.); (M.S.)
- Department of Plastic and Reconstructive Surgery, Mito Saiseikai General Hospital, Mito 311-4145, Ibaraki, Japan
| | - Kaoru Sasaki
- Department of Plastic and Reconstructive Surgery, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan (Y.I.); (M.S.)
| | - Yukiko Aihara
- Department of Plastic and Reconstructive Surgery, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan (Y.I.); (M.S.)
| | - Vuong Cat Khanh
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Mitsuru Sekido
- Department of Plastic and Reconstructive Surgery, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan (Y.I.); (M.S.)
| |
Collapse
|
8
|
Wu D, Liu X, Jin Z. Placental mesenchymal stem cells-secreted proenkephalin suppresses the p38 MAPK signaling to block hyperproliferation of keloid fibroblasts. Tissue Cell 2023; 85:102218. [PMID: 37913601 DOI: 10.1016/j.tice.2023.102218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/25/2023] [Accepted: 09/09/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Thanks to their multi-potency and secretory functions, mesenchymal stem cells (MSCs) have long been established as an ideal cell type for skin wound healing and a candidate therapeutic strategy for excessive pathological scarring in the meantime. This study focuses on the effect of placental MSCs (PMSCs) on the activity of keloid fibroblasts (KFs) and the potential involvement of proenkephalin (PENK). METHODS Secretory protein of PMSC that are lowly expressed in KFs were predicted by bioinformatics analyses. The expression of PENK in KFs was detected by RT-qPCR and western blot analysis. PMSCs were co-cultured with KFs and dermal fibroblasts (DFs) to examine their effect on proliferation, migration, invasion, and apoptosis of the distinct cell types. PENK secretion by PMSCs and its uptake by KFs were examined by ELISA, WB, and immunofluorescence staining. Loss-of-functions of PENK and p38-MAPK were induced to examine the activity of KFs in vitro and in mice. RESULTS PENK, a secretory protein of PMSCs, was conspicuously downregulated in KFs compared to normal DFs. PMSC stimulation suppressed proliferation, migration, invasion, and resistance to apoptosis of the co-cultured KFs but not DFs, which was ascribed to the upregulation of PENK protein in KFs. PMSCs-secreted PENK suppressed p38 phosphorylation in KFs. The proliferative and aggressive properties of KFs in vitro and the nodule-forming capacity of KFs in vivo were promoted upon PENK downregulation but suppressed by the p38 MAPK inhibitor SB202190. CONCLUSION This work unravels that PMSCs-secreted PENK suppresses the p38 MAPK signaling to block hyperproliferation of KFs.
Collapse
Affiliation(s)
- Di Wu
- Department of Dermtology, Jilin Central Hospital, Jilin 132001, Jilin, PR China; Department of Dermtology, Affiliated Hospital of Yanbian University, Yanji 133000, Jilin, PR China
| | - Xiao Liu
- Department of Dermtology, Jilin Central Hospital, Jilin 132001, Jilin, PR China
| | - Zhehu Jin
- Department of Dermtology, Affiliated Hospital of Yanbian University, Yanji 133000, Jilin, PR China.
| |
Collapse
|
9
|
Li W, Hu J, Chen C, Li X, Zhang H, Xin Y, Tian Q, Wang S. Emerging advances in hydrogel-based therapeutic strategies for tissue regeneration. Regen Ther 2023; 24:459-471. [PMID: 37772128 PMCID: PMC10523184 DOI: 10.1016/j.reth.2023.09.007] [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: 04/17/2023] [Revised: 08/14/2023] [Accepted: 09/07/2023] [Indexed: 09/30/2023] Open
Abstract
Significant developments in cell therapy and biomaterial science have broadened the therapeutic landscape of tissue regeneration. Tissue damage is a complex biological process in which different types of cells play a specific role in repairing damaged tissues and growth factors strictly regulate the activity of these cells. Hydrogels have become promising biomaterials for tissue regeneration if appropriate materials are selected and the hydrogel properties are well-regulated. Importantly, they can be used as carriers for living cells and growth factors due to the high water-holding capacity, high permeability, and good biocompatibility of hydrogels. Cell-loaded hydrogels can play an essential role in treating damaged tissues and open new avenues for cell therapy. There is ample evidence substantiating the ability of hydrogels to facilitate the delivery of cells (stem cell, macrophage, chondrocyte, and osteoblast) and growth factors (bone morphogenetic protein, transforming growth factor, vascular endothelial growth factor and fibroblast growth factor). This paper reviewed the latest advances in hydrogels loaded with cells or growth factors to promote the reconstruction of tissues. Furthermore, we discussed the shortcomings of the application of hydrogels in tissue engineering to promote their further development.
Collapse
Affiliation(s)
- Wenqi Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jing Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Cheng Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xinyue Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Honghua Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yanru Xin
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qingchang Tian
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Shuling Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
10
|
Correa-Araujo L, Prieto-Abello L, Lara-Bertrand A, Medina-Solano M, Guerrero L, Camacho B, Silva-Cote I. Bioengineered skin constructs based on mesenchymal stromal cells and acellular dermal matrix exposed to inflammatory microenvironment releasing growth factors involved in skin repair. Stem Cell Res Ther 2023; 14:306. [PMID: 37880776 PMCID: PMC10601120 DOI: 10.1186/s13287-023-03535-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Skin tissue engineering is a rapidly evolving field of research that effectively combines stem cells and biological scaffolds to replace damaged tissues. Human Wharton's jelly mesenchymal stromal cells (hWJ-MSCs) are essential to generate tissue constructs, due to their potent immunomodulatory effects and release of paracrine factors for tissue repair. Here, we investigated whether hWJ-MSC grown on human acellular dermal matrix (hADM) scaffolds and exposed to a proinflammatory environment maintain their ability to produce in vitro growth factors involved in skin injury repair and promote in vivo wound healing. METHODS We developed a novel method involving physicochemical and enzymatic treatment of cadaveric human skin to obtain hADM scaffold. Subsequently, skin bioengineered constructs were generated by seeding hWJ-MSCs on the hADM scaffold (construct 1) and coating it with human platelet lysate clot (hPL) (construct 2). Either construct 1 or 2 were then incubated with proinflammatory cytokines (IL-1α, IL-1β, IL-6, TNF-α) for 12, 24, 48, 72 and 96 h. Supernatants from treated and untreated constructs and hWJ-MSCs on tissue culture plate (TCP) were collected, and concentration of the following growth factors, bFGF, EGF, HGF, PDGF, VEGF and Angiopoietin-I, was determined by immunoassay. We also asked whether hWJ-MSCs in the construct 1 have potential toward epithelial differentiation after being cultured in an epithelial induction stimulus using an air-liquid system. Immunostaining was used to analyze the synthesis of epithelial markers such as filaggrin, involucrin, plakoglobin and the mesenchymal marker vimentin. Finally, we evaluated the in vivo potential of hADM and construct 1 in a porcine full-thickness excisional wound model. RESULTS We obtained and characterized the hADM and confirmed the viability of hWJ-MSCs on the scaffold. In both constructs without proinflammatory treatment, we reported high bFGF production. In contrast, the levels of other growth factors were similar to the control (hWJ-MSC/TCP) with or without proinflammatory treatment. Except for PDGF in the stimulated group. These results indicated that the hADM scaffold maintained or enhanced the production of these bioactive molecules by hWJ-MSCs. On the other hand, increased expression of filaggrin, involucrin, and plakoglobin and decreased expression of vimentin were observed in constructs cultured in an air-liquid system. In vivo experiments demonstrated the potential of both hADM and hADM/hWJ-MSCs constructs to repair skin wounds with the formation of stratified epithelium, basement membrane and dermal papillae, improving the appearance of the repaired tissue. CONCLUSIONS hADM is viable to fabricate a tissue construct with hWJ-MSCs able to promote the in vitro synthesis of growth factors and differentiation of these cells toward epithelial lineage, as well as, promote in a full-thickness skin injury the new tissue formation. These results indicate that hADM 3D architecture and its natural composition improved or maintained the cell function supporting the potential therapeutic use of this matrix or the construct for wound repair and providing an effective tissue engineering strategy for skin repair.
Collapse
Affiliation(s)
- Luz Correa-Araujo
- Tissue Engineering Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud - IDCBIS, Carrera 32 # 12-81, Secretaria Distrital de Salud, Bogotá, Colombia
| | - Leonardo Prieto-Abello
- Tissue Engineering Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud - IDCBIS, Carrera 32 # 12-81, Secretaria Distrital de Salud, Bogotá, Colombia
| | - Adriana Lara-Bertrand
- Tissue Engineering Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud - IDCBIS, Carrera 32 # 12-81, Secretaria Distrital de Salud, Bogotá, Colombia
| | - Martha Medina-Solano
- Tissue Engineering Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud - IDCBIS, Carrera 32 # 12-81, Secretaria Distrital de Salud, Bogotá, Colombia
| | - Linda Guerrero
- Tissue Bank, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud - IDCBIS, Bogotá, Colombia
| | - Bernardo Camacho
- Tissue Engineering Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud - IDCBIS, Carrera 32 # 12-81, Secretaria Distrital de Salud, Bogotá, Colombia
- Tissue Bank, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud - IDCBIS, Bogotá, Colombia
| | - Ingrid Silva-Cote
- Tissue Engineering Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud - IDCBIS, Carrera 32 # 12-81, Secretaria Distrital de Salud, Bogotá, Colombia.
| |
Collapse
|
11
|
Andalib E, Kashfi M, Mahmoudvand G, Rezaei E, Mahjoor M, Torki A, Afkhami H. Application of hypoxia-mesenchymal stem cells in treatment of anaerobic bacterial wound infection: wound healing and infection recovery. Front Microbiol 2023; 14:1251956. [PMID: 37869672 PMCID: PMC10586055 DOI: 10.3389/fmicb.2023.1251956] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Mesenchymal stromal cells, commonly referred to as MSCs, are a type of multipotent stem cells that are typically extracted from adipose tissue and bone marrow. In the field of tissue engineering and regenerative medicine, MSCs and their exosomes have emerged as revolutionary tools. Researchers are now devoting greater attention to MSCs because of their ability to generate skin cells like fibroblasts and keratinocytes, as well as their distinctive potential to decrease inflammation and emit pro-angiogenic molecules at the site of wounds. More recent investigations revealed that MSCs can exert numerous direct and indirect antimicrobial effects that are immunologically mediated. Collectively, these antimicrobial properties can remove bacterial infections when the MSCs are delivered in a therapeutic setting. Regardless of the positive therapeutic potential of MSCs for a multitude of conditions, transplanted MSC cell retention continues to be a major challenge. Since MSCs are typically administered into naturally hypoxic tissues, understanding the impact of hypoxia on the functioning of MSCs is crucial. Hypoxia has been postulated to be among the factors determining the differentiation of MSCs, resulting in the production of inflammatory cytokines throughout the process of tissue regeneration and wound repair. This has opened new horizons in developing MSC-based systems as a potent therapeutic tool in oxygen-deprived regions, including anaerobic wound infection sites. This review sheds light on the role of hypoxia-MSCs in the treatment of anaerobic bacterial wound infection in terms of both their regenerative and antimicrobial activities.
Collapse
Affiliation(s)
- Elahe Andalib
- Department of Microbiology, School of Basic Sciences, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Mojtaba Kashfi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Elaheh Rezaei
- Department of Microbiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Torki
- Department of Medical Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
12
|
Shu F, Lu J, Zhang W, Huang H, Lin J, Jiang L, Liu W, Liu T, Xiao S, Zheng Y, Xia Z. JAM-A Overexpression in Human Umbilical Cord-Derived Mesenchymal Stem Cells Accelerated the Angiogenesis of Diabetic Wound By Enhancing Both Paracrine Function and Survival of Mesenchymal Stem Cells. Stem Cell Rev Rep 2023; 19:1554-1575. [PMID: 37060532 DOI: 10.1007/s12015-023-10518-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 04/16/2023]
Abstract
Mesenchymal stem cells (MSCs) is promising in promoting wound healing mainly due to their paracrine function. Nonetheless, the transplanted MSCs presented poor survival with cell dysfunction and paracrine problem in diabetic environment, thus limiting their therapeutic efficacy and clinical application. JAM-A, an adhesion molecule, has been reported to play multi-functional roles in diverse cells. We therefore investigated the potential effect of JAM-A on MSCs under diabetic environment and explored the underlying mechanism. Indeed, high-glucose condition inhibited MSCs viability and JAM-A expression. However, JAM-A abnormality was rescued by lentivirus transfection and JAM-A overexpression promoted MSCs proliferation, migration and adhesion under hyperglycemia. Moreover, JAM-A overexpression attenuated high-glucose-induced ROS production and MSCs apoptosis. The bio-effects of JAM-A on MSCs under hyperglycemia were confirmed by RNA-seq with enrichment analyses. Moreover, Luminex chip results showed JAM-A overexpression dramatically upregulated PDGF-BB and VEGF in the supernatant of MSCs, which was verified by RT-qPCR and western blotting. The supernatant was further found to facilitate HUVECs proliferation, migration and angiogenesis under hyperglycemia. In vivo experiments revealed JAM-A overexpression significantly enhanced MSCs survival, promoted wound angiogenesis, and thus accelerated diabetic wound closure, partially by enhancing PDGF-BB and VEGF expression. This study firstly demonstrated that JAM-A expression of MSCs was inhibited upon high-glucose stimulation. JAM-A overexpression alleviated high-glucose-induced MSCs dysfunction, enhanced their anti-oxidative capability, protected MSCs from hyperglycemia-induced apoptosis and improved their survival, thus strengthening MSCs paracrine function to promote angiogenesis and significantly accelerating diabetic wound healing, which offers a promising strategy to maximize MSCs-based therapy in diabetic wound.
Collapse
Affiliation(s)
- Futing Shu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jianyu Lu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Wei Zhang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Hongchao Huang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jiezhi Lin
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Luofeng Jiang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Wenzhang Liu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Tianyi Liu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China.
| | - Yongjun Zheng
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China.
| | - Zhaofan Xia
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China.
- Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
13
|
Pham TLB, Nguyen DPH, Luu TTT, Nguyen LS, Binh NT, Nguyen QD, Tran PA. Encapsulation of Human Umbilical Cord Mesenchymal Stem Cells in LunaGel Photocrosslinkable Extracellular Matrix and Subcutaneous Transplantation in Mice. Biomedicines 2023; 11:biomedicines11041158. [PMID: 37189776 DOI: 10.3390/biomedicines11041158] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Stem cells have significant potential in regenerative medicines. However, a major issue with implanting stem cells in the regeneration of new tissue is the methods to implant them and cell viability and functions before and after implantation. Here we developed a simple yet effective method that used photo-crosslinkable gelatin-based hydrogel (LunaGelTM) as a scaffold for the encapsulation, expansion, and eventually, transplantation of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) into mice subcutaneously. We demonstrated the proliferation and maintenance of the original expression of mesenchymal stem cell markers as well as the ability to differentiate into mesoderm-derived cells. The hydrogel was highly stable with no signs of degradation after 20 days in PBS. The hUC-MSCs remained viable after transplantation into mice's subcutaneous pockets and migrated to integrate with the surrounding tissues. We showed a collagen-rich layer surrounding the transplanted cell-laden scaffold indicating the effects of growth factors secreted by the hUC-MSCs. A connective tissue layer was found between the implanted cell-laden scaffold and the collagen layer, and immunohistochemical staining results suggested that this tissue was derived from the MSCs which migrated from within the scaffold. The results, thus, also suggested a protective effect the scaffold has on the encapsulated cells from the antibodies and cytotoxic cells of the host immune system.
Collapse
Affiliation(s)
- Truc Le-Buu Pham
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | | | - Thao Thi-Thu Luu
- Histology-Embryology-Pathology Department, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Luong Si Nguyen
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Nguyen Trong Binh
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Quan Dang Nguyen
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Phong Anh Tran
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
| |
Collapse
|
14
|
Abstract
Pathological hair loss (also known as alopecia) and shortage of hair follicle (HF) donors have posed an urgent requirement for HF regeneration. With the revelation of mechanisms in tissue engineering, the proliferation of HFs in vitro has achieved more promising trust for the treatments of alopecia and other skin impairments. Theoretically, HF organoids have great potential to develop into native HFs and attachments such as sweat glands after transplantation. However, since the rich extracellular matrix (ECM) deficiency, the induction characteristics of skin-derived cells gradually fade away along with their trichogenic capacity after continuous cell passaging in vitro. Therefore, ECM-mimicking support is an essential prelude before HF transplantation is implemented. This review summarizes the status of providing various epidermal and dermal cells with a three-dimensional (3D) scaffold to support the cell homeostasis and better mimic in vivo environments for the sake of HF regeneration. HF-relevant cells including dermal papilla cells (DPCs), hair follicle stem cells (HFSCs), and mesenchymal stem cells (MSCs) are able to be induced to form HF organoids in the vitro culture system. The niche microenvironment simulated by different forms of biomaterial scaffold can offer the cells a network of ordered growth environment to alleviate inductivity loss and promote the expression of functional proteins. The scaffolds often play the role of ECM substrates and bring about epithelial-mesenchymal interaction (EMI) through coculture to ensure the functional preservation of HF cells during in vitro passage. Functional HF organoids can be formed either before or after transplantation into the dermis layer. Here, we review and emphasize the importance of 3D culture in HF regeneration in vitro. Finally, the latest progress in treatment trials and critical analysis of the properties and benefits of different emerging biomaterials for HF regeneration along with the main challenges and prospects of HF regenerative approaches are discussed.
Collapse
Affiliation(s)
- Wei Zheng
- College of Food Science & Technology, Shanghai Ocean University, Shanghai 201306, P.R. China
| | - Chang-Hua Xu
- College of Food Science & Technology, Shanghai Ocean University, Shanghai 201306, P.R. China
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai 201306, China
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai 201306, China
- National R&D Branch Center for Freshwater Aquatic Products Processing Technology (Shanghai), Shanghai 201306, China
| |
Collapse
|
15
|
Brembilla NC, Modarressi A, André-Lévigne D, Brioudes E, Lanza F, Vuagnat H, Durual S, Marger L, Boehncke WH, Krause KH, Preynat-Seauve O. Adipose-Derived Stromal Cells within a Gelatin Matrix Acquire Enhanced Regenerative and Angiogenic Properties: A Pre-Clinical Study for Application to Chronic Wounds. Biomedicines 2023; 11:biomedicines11030987. [PMID: 36979966 PMCID: PMC10046849 DOI: 10.3390/biomedicines11030987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
This study evaluates the influence of a gelatin sponge on adipose-derived stromal cells (ASC). Transcriptomic data revealed that, compared to ASC in a monolayer, a cross-linked porcine gelatin sponge strongly influences the transcriptome of ASC. Wound healing genes were massively regulated, notably with the inflammatory and angiogenic factors. Proteomics on conditioned media showed that gelatin also acted as a concentrator and reservoir of the regenerative ASC secretome. This secretome promoted fibroblast survival and epithelialization, and significantly increased the migration and tubular assembly of endothelial cells within fibronectin. ASC in gelatin on a chick chorioallantoic membrane were more connected to vessels than an empty sponge, confirming an increased angiogenesis in vivo. No tumor formation was observed in immunodeficient nude mice to which an ASC gelatin sponge was transplanted subcutaneously. Finally, ASC in a gelatin sponge prepared from outbred rats accelerated closure and re-vascularization of ischemic wounds in the footpads of rats. In conclusion, we provide here preclinical evidence that a cross-linked porcine gelatin sponge is an optimal carrier to concentrate and increase the regenerative activity of ASC, notably angiogenic. This formulation of ASC represents an optimal, convenient and clinically compliant option for the delivery of ASC on ischemic wounds.
Collapse
Affiliation(s)
- Nicolo Costantino Brembilla
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Division of Dermatology and Venereology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Ali Modarressi
- Division of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Dominik André-Lévigne
- Division of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Estelle Brioudes
- Laboratory of Therapy and Stem Cells, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Florian Lanza
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Hubert Vuagnat
- Program for Wounds and Wound Healing, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Stéphane Durual
- Laboratory of Biomaterials, Faculty of Dental Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Laurine Marger
- Laboratory of Biomaterials, Faculty of Dental Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Division of Dermatology and Venereology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Laboratory of Therapy and Stem Cells, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Olivier Preynat-Seauve
- Laboratory of Therapy and Stem Cells, Geneva University Hospitals, 1205 Geneva, Switzerland
- Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
16
|
Jiménez-Gastélum G, Ramos-Payán R, López-Gutierrez J, Ayala-Ham A, Silva-Benítez E, Bermúdez M, Romero-Quintana JG, Sanchez-Schmitz G, Aguilar-Medina M. An extracellular matrix hydrogel from porcine urinary bladder for tissue engineering: In vitro and in vivo analyses. Biomed Mater Eng 2022:BME221450. [PMID: 37125540 DOI: 10.3233/bme-221450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND The necessity to manufacture scaffolds with superior capabilities of biocompatibility and biodegradability has led to the production of extracellular matrix (ECM) scaffolds. Among their advantages, they allow better cell colonization, which enables its successful integration into the hosted tissue, surrounding the area to be repaired and their formulations facilitate placing it into irregular shapes. The ECM from porcine urinary bladder (pUBM) comprises proteins, proteoglycans and glycosaminoglycans which provide support and enable signals to the cells. These properties make it an excellent option to produce hydrogels that can be used in regenerative medicine. OBJECTIVE The goal of this study was to assess the biocompatibility of an ECM hydrogel derived from the porcine urinary bladder (pUBMh) in vitro using fibroblasts, macrophages, and adipose-derived mesenchymal stem cells (AD-MCSs), as well as biocompatibility in vivo using Wistar rats. METHODS Effects upon cells proliferation/viability was measured using MTT assay, cytotoxic effects were analyzed by quantifying lactate dehydrogenase release and the Live/Dead Cell Imaging assay. Macrophage activation was assessed by quantification of IL-6, IL-10, IL-12p70, MCP-1, and TNF-α using a microsphere-based cytometric bead array. For in vivo analysis, Wistar rats were inoculated into the dorsal sub-dermis with pUBMh. The specimens were sacrificed at 24 h after inoculation for histological study. RESULTS The pUBMh obtained showed good consistency and absence of cell debris. The biocompatibility tests in vitro revealed that the pUBMh promoted cell proliferation and it is not cytotoxic on the three tested cell lines and induces the production of pro-inflammatory cytokines on macrophages, mainly TNF-α and MCP-1. In vivo, pUBMh exhibited fibroblast-like cell recruitment, without tissue damage or inflammation. CONCLUSION The results show that pUBMh allows cell proliferation without cytotoxic effects and can be considered an excellent biomaterial for tissue engineering.
Collapse
Affiliation(s)
| | - Rosalío Ramos-Payán
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Culiacan, Mexico
| | | | - Alfredo Ayala-Ham
- Faculty of Biology, Autonomous University of Sinaloa, Culiacan, Mexico
- Faculty of Odontology, Autonomous University of Sinaloa, Culiacan, Mexico
| | | | - Mercedes Bermúdez
- Faculty of Odontology, Autonomous University of Chihuahua, Chihuahua, Mexico
| | | | - Guzman Sanchez-Schmitz
- Boston Children's Hospital and Harvard Medical School, Harvard University, Boston, MA, USA
| | - Maribel Aguilar-Medina
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Culiacan, Mexico
| |
Collapse
|
17
|
Wang Y, Wang G, Liu H. Tenascin-C: A Key Regulator in Angiogenesis during Wound Healing. Biomolecules 2022; 12:1689. [PMID: 36421704 PMCID: PMC9687801 DOI: 10.3390/biom12111689] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Injury repair is a complex physiological process in which multiple cells and molecules are involved. Tenascin-C (TNC), an extracellular matrix (ECM) glycoprotein, is essential for angiogenesis during wound healing. This study aims to provide a comprehensive review of the dynamic changes and functions of TNC throughout tissue regeneration and to present an up-to-date synthesis of the body of knowledge pointing to multiple mechanisms of TNC at different restoration stages. (2) Methods: A review of the PubMed database was performed to include all studies describing the pathological processes of damage restoration and the role, structure, expression, and function of TNC in post-injury treatment; (3) Results: In this review, we first introduced the construction and expression signature of TNC. Then, the role of TNC during the process of damage restoration was introduced. We highlight the temporal heterogeneity of TNC levels at different restoration stages. Furthermore, we are surprised to find that post-injury angiogenesis is dynamically consistent with changes in TNC. Finally, we discuss the strategies for TNC in post-injury treatment. (4) Conclusions: The dynamic expression of TNC has a significant impact on angiogenesis and healing wounds and counters many negative aspects of poorly healing wounds, such as excessive inflammation, ischemia, scarring, and wound infection.
Collapse
Affiliation(s)
- Yucai Wang
- Department of Orthopaedic Surgery, Tangdu Hospital, AirForce Medical University, Xi’an 710000, China
| | - Guangfu Wang
- Vasculocardiology Department, The Fourth People’s Hospital of Jinan, Jinan 250000, China
| | - Hao Liu
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| |
Collapse
|
18
|
Luo Y, Xu X, Ye Z, Xu Q, Li J, Liu N, Du Y. 3D bioprinted mesenchymal stromal cells in skin wound repair. Front Surg 2022; 9:988843. [PMID: 36311952 PMCID: PMC9614372 DOI: 10.3389/fsurg.2022.988843] [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: 07/07/2022] [Accepted: 09/20/2022] [Indexed: 11/07/2022] Open
Abstract
Skin tissue regeneration and repair is a complex process involving multiple cell types, and current therapies are limited to promoting skin wound healing. Mesenchymal stromal cells (MSCs) have been proven to enhance skin tissue repair through their multidifferentiation and paracrine effects. However, there are still difficulties, such as the limited proliferative potential and the biological processes that need to be strengthened for MSCs in wound healing. Recently, three-dimensional (3D) bioprinting has been applied as a promising technology for tissue regeneration. 3D-bioprinted MSCs could maintain a better cell ability for proliferation and expression of biological factors to promote skin wound healing. It has been reported that 3D-bioprinted MSCs could enhance skin tissue repair through anti-inflammatory, cell proliferation and migration, angiogenesis, and extracellular matrix remodeling. In this review, we will discuss the progress on the effect of MSCs and 3D bioprinting on the treatment of skin tissue regeneration, as well as the perspective and limitations of current research.
Collapse
|
19
|
Sylakowski K, Hwang MP, Justin A, Whaley D, Wang Y, Wells A. The matricellular protein decorin delivered intradermally with coacervate improves wound resolution in the CXCR3-deficient mouse model of hypertrophic scarring. Wound Repair Regen 2022; 30:436-447. [PMID: 35470921 DOI: 10.1111/wrr.13017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/17/2022] [Accepted: 04/14/2022] [Indexed: 01/26/2023]
Abstract
Cutaneous wound healing is an intricate orchestration of three overlapping phases of repair that encompass numerous cell types, signalling cascades, and microenvironment modifications to reach a successful resolution. Disruption of any of these steps will create an abnormal healing response resulting in either ulceration or excessive scarring. It has become evident that the extracellular matrix and its associated components are key orchestrators during this process. One of these essential matrix proteins is decorin, a small leucine-rich proteoglycan (SLRP) that acts as a regulator of collagen fibrillogenesis and a non-competitive inhibitor of multiple growth factors signalling cascades. Decorin is a necessary shut-off switch for the pro-reparative mechanism of the tissue replacement phase and limits the occurrence of hypertrophic scarring by preventing excessive repair. We investigated the use of decorin as a therapeutic by administering the matrix protein anchored in a slow-release coacervate in a hypertrophic scarring mouse model. The results show that early wound healing phase measurements exhibit little difference in performance compared to our coacervate-only baseline or HB-EGF-treated control mice. However, during the resolution phase of wound healing, the decorin-treatment significantly reduces cutaneous thickness, enhances collagen alignment, and improves overall wound scoring in the mice. Thus, mice treated with decorin display better healing outcomes and could limit the hypertrophic scarring phenotype in the coacervate only, and HB-EGF controls. These results suggest that decorin may be a promising tool and alternative therapy for patients who suffer from over-exuberant matrix deposition during wound healing.
Collapse
Affiliation(s)
- Kyle Sylakowski
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,R&D Service, Pittsburgh, VA Health System, Pittsburgh, Pennsylvania, USA
| | - Mintai Peter Hwang
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Amritha Justin
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Diana Whaley
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,R&D Service, Pittsburgh, VA Health System, Pittsburgh, Pennsylvania, USA
| | - Yadong Wang
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,R&D Service, Pittsburgh, VA Health System, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Ayala-Ham A, Aguilar-Medina M, León-Félix J, Romero-Quintana JG, Bermúdez M, López-Gutierrez J, Jiménez-Gastélum G, Avendaño-Félix M, Lizárraga-Verdugo E, Castillo-Ureta H, López-Camarillo C, Ramos-Payan R. Extracellular matrix hydrogel derived from bovine bone is biocompatible in vitro and in vivo. Biomed Mater Eng 2022; 33:491-504. [DOI: 10.3233/bme-211387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Nowadays, biomaterials used as a scaffold must be easy to deliver in the bone defect area. Extracellular matrix (ECM) hydrogels are highly hydrated polymers that can fill irregular shapes and act as bioactive materials. OBJECTIVE: This work aims to show the effects of ECM hydrogels derived from bovine bone (bECMh) on proliferation, cytotoxicity and expression of pro-inflammatory cytokines in three cells types involved in tissue regeneration, as well as biocompatibility in vivo. METHODS: In vitro, we used an extract of bECMh to test it on macrophages, fibroblasts, and adipose-derived mesenchymal stem cells (AD-MCSs). Cell proliferation was measured using the MTT assay, cytotoxicity was measured by quantifying lactate dehydrogenase release and the Live/Dead Cell Imaging assays. Concentrations of IL-6, IL-10, IL-12p70, MCP-1 and TNF-α were quantified in the supernatants using a microsphere-based cytometric bead array. For in vivo analysis, Wistar rats were inoculated into the dorsal sub-dermis with bECMh, taking as reference the midline of the back. The specimens were sacrificed at 24 h for histological study. RESULTS: In vitro, this hydrogel behaves as a dynamic biomaterial that increases fibroblast proliferation, induces the production of pro-inflammatory cytokines in macrophages, among which MCP-1 and TNF-α stand out. In vivo, bECMh allows the colonization of host fibroblast-like and polymorphonuclear cells, without tissue damage or inflammation. CONCLUSIONS: The results indicate that bECMh is a biocompatible material that could be used as a scaffold, alone or in conjunction with cells or functional biomolecules, enhancing proliferation and allowing the filling of bone defects to its further regeneration.
Collapse
Affiliation(s)
- Alfredo Ayala-Ham
- , Autonomous University of Sinaloa, , Mexico
- , Autonomous University of Sinaloa, , Mexico
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bosanquet DC, Harding KG. Wound healing: potential therapeutic options. Br J Dermatol 2021; 187:149-158. [PMID: 34726774 DOI: 10.1111/bjd.20772] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2021] [Indexed: 12/22/2022]
Abstract
This review highlights the range of therapeutic options available to clinicians treating difficult-to-heal wounds. While certain treatments are established in daily clinical practice, most therapeutic interventions lack robust and rigorous data regarding their efficacy, which would help to determine when, and for whom, they should be used. The purpose of this review is to give a broad overview of the available interventions, with a brief summary of the evidence base for each intervention.
Collapse
Affiliation(s)
- D C Bosanquet
- South East Wales Vascular Network, Aneurin Bevan University Health Board, Royal Gwent Hospital, Cardiff Road, Newport, NP16 2UB, UK
| | - K G Harding
- Clinical Innovation Hub, Cardiff University, Cardiff, CF14 4XN, UK.,Skin Research Institute Singapore (SRIS), Singapore
| |
Collapse
|
22
|
The Role of MSC in Wound Healing, Scarring and Regeneration. Cells 2021; 10:cells10071729. [PMID: 34359898 PMCID: PMC8305394 DOI: 10.3390/cells10071729] [Citation(s) in RCA: 171] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue repair and regeneration after damage is not completely understood, and current therapies to support this process are limited. The wound healing process is associated with cell migration and proliferation, extracellular matrix remodeling, angiogenesis and re-epithelialization. In normal conditions, a wound will lead to healing, resulting in reparation of the tissue. Several risk factors, chronic inflammation, and some diseases lead to a deficient wound closure, producing a scar that can finish with a pathological fibrosis. Mesenchymal stem/stromal cells (MSCs) are widely used for their regenerative capacity and their possible therapeutically potential. Derived products of MSCs, such as exosomes or extravesicles, have shown a therapeutic potential similar to MSCs, and these cell-free products may be interesting in clinics. MSCs or their derivative products have shown paracrine beneficial effects, regulating inflammation, modifying the fibroblast activation and production of collagen and promoting neovascularization and re-epithelialization. This review describes the effects of MSCs and their derived products in each step of the wound repair process. As well, it reviews the pre-clinical and clinical use of MSCs to benefit in skin wound healing in diabetic associated wounds and in pathophysiological fibrosis.
Collapse
|
23
|
Spencer H, Moshkbouymatin N, Webb WR, Joshi A, D'Souza A. Update on the role of emerging stem cell technology in head and neck medicine. Head Neck 2021; 43:1928-1938. [PMID: 33751726 DOI: 10.1002/hed.26674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
Head and neck surgery is a broad discipline that involves the management of complex conditions such as burns, skin cancer, head and neck cancer, congenital abnormalities, and facial rejuvenation. For patients with cancer, surgery, radiotherapy, and chemotherapy are often the main modes of treatment. Many patients require follow-up reconstructive surgery, and the use of stem cells offers novel treatments that could aid recovery. Laryngeal, tracheal, and neuronal tissues are frequently damaged by surgery in the head and neck and these tissues have little intrinsic regenerative ability. Pluripotent embryonic stem cells retain the ability to differentiate into a wide variety of cells meaning that large tissue defects can be reduced by stimulating new cell growth. Research has demonstrated potential benefits of using stem cells in facial rejuvenation procedures and the management of burns sequelae. The advancements made in the use of adult progenitor stem cells as a possible source for pluripotent stem cells (induced pluripotent stem cells) mean that ethical considerations around the use of embryological tissue can be minimized, allowing for more research to take place. Currently, the evidence base for the use of stem cells in head and neck surgery is limited, but it has now been proven that stem cells can act as a source for lost or damaged tissue in the head and neck. With continuous advancements being made in the fields of tissue engineering, it is likely that stem cells will play a major role in head and neck surgery in the future.
Collapse
Affiliation(s)
- Harry Spencer
- Department of Vascular Surgery, St Georges Hospital, London, UK
| | | | - William Richard Webb
- Faculty of Medicine, Health and Social Sciences, Canterbury Christ Church University, Chatham, UK
| | - Anil Joshi
- Department of Ear, Nose and Throat Surgery, University Hospital Lewisham, London, UK
| | - Alwyn D'Souza
- Department of Ear, Nose and Throat Surgery, University Hospital Lewisham, London, UK
| |
Collapse
|
24
|
Sierra-Sánchez Á, Montero-Vilchez T, Quiñones-Vico MI, Sanchez-Diaz M, Arias-Santiago S. Current Advanced Therapies Based on Human Mesenchymal Stem Cells for Skin Diseases. Front Cell Dev Biol 2021; 9:643125. [PMID: 33768095 PMCID: PMC7985058 DOI: 10.3389/fcell.2021.643125] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/18/2021] [Indexed: 12/17/2022] Open
Abstract
Skin disease may be related with immunological disorders, external aggressions, or genetic conditions. Injuries or cutaneous diseases such as wounds, burns, psoriasis, and scleroderma among others are common pathologies in dermatology, and in some cases, conventional treatments are ineffective. In recent years, advanced therapies using human mesenchymal stem cells (hMSCs) from different sources has emerged as a promising strategy for the treatment of many pathologies. Due to their properties; regenerative, immunomodulatory and differentiation capacities, they could be applied for the treatment of cutaneous diseases. In this review, a total of thirteen types of hMSCs used as advanced therapy have been analyzed, considering the last 5 years (2015-2020). The most investigated types were those isolated from umbilical cord blood (hUCB-MSCs), adipose tissue (hAT-MSCs) and bone marrow (hBM-MSCs). The most studied diseases were wounds and ulcers, burns and psoriasis. At preclinical level, in vivo studies with mice and rats were the main animal models used, and a wide range of types of hMSCs were used. Clinical studies analyzed revealed that cell therapy by intravenous administration was the advanced therapy preferred except in the case of wounds and burns where tissue engineering was also reported. Although in most of the clinical trials reviewed results have not been posted yet, safety was high and only local slight adverse events (mild nausea or abdominal pain) were reported. In terms of effectiveness, it was difficult to compare the results due to the different doses administered and variables measured, but in general, percentage of wound's size reduction was higher than 80% in wounds, Psoriasis Area and Severity Index and Severity Scoring for Atopic Dermatitis were significantly reduced, for scleroderma, parameters such as Modified Rodnan skin score (MRSC) or European Scleroderma Study Group activity index reported an improvement of the disease and for hypertrophic scars, Vancouver Scar Scale (VSS) score was decreased after applying these therapies. On balance, hMSCs used for the treatment of cutaneous diseases is a promising strategy, however, the different experimental designs and endpoints stablished in each study, makes necessary more research to find the best way to treat each patient and disease.
Collapse
Affiliation(s)
- Álvaro Sierra-Sánchez
- Cell Production and Tissue Engineering Unit, Andalusian Network of Design and Translation of Advanced Therapies, Virgen de las Nieves University Hospital, Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain
| | - Trinidad Montero-Vilchez
- Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.,Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain
| | - María I Quiñones-Vico
- Cell Production and Tissue Engineering Unit, Andalusian Network of Design and Translation of Advanced Therapies, Virgen de las Nieves University Hospital, Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.,Department of Dermatology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Manuel Sanchez-Diaz
- Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.,Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain
| | - Salvador Arias-Santiago
- Cell Production and Tissue Engineering Unit, Andalusian Network of Design and Translation of Advanced Therapies, Virgen de las Nieves University Hospital, Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.,Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain.,Department of Dermatology, Faculty of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
25
|
Bojanic C, To K, Hatoum A, Shea J, Seah KTM, Khan W, Malata CM. Mesenchymal stem cell therapy in hypertrophic and keloid scars. Cell Tissue Res 2021; 383:915-930. [PMID: 33386995 PMCID: PMC7960584 DOI: 10.1007/s00441-020-03361-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022]
Abstract
Scars are the normal outcome of wound repair and involve a co-ordinated inflammatory and fibrotic process. When a scar does not resolve, uncontrolled chronic inflammation can persist and elicits excessive scarring that leads to a range of abnormal phenotypes such as hypertrophic and keloid scars. These pathologies result in significant impairment of quality of life over a long period of time. Existing treatment options are generally unsatisfactory, and there is mounting interest in innovative cell-based therapies. Despite the interest in mesenchymal stem cells (MSCs), there is yet to be a human clinical trial that investigates the potential of MSCs in treating abnormal scarring. A synthesis of existing evidence of animal studies may therefore provide insight into the barriers to human application. The aim of this PRISMA systematic review was to evaluate the effectiveness of MSC transplantation in the treatment of hypertrophic and keloid scars in in vivo models. A total of 11 case-control studies were identified that treated a total of 156 subjects with MSCs or MSC-conditioned media. Ten studies assessed hypertrophic scars, and one looked at keloid scars. All studies evaluated scars in terms of macroscopic and histological appearances and most incorporated immunohistochemistry. The included studies all found improvements in the above outcomes with MSC or MSC-conditioned media without complications. The studies reviewed support a role for MSC therapy in treating scars that needs further exploration. The transferability of these findings to humans is limited by factors such as the reliability and validity of the disease model, the need to identify the optimal MSC cell source, and the outcome measures employed.
Collapse
Affiliation(s)
- Christine Bojanic
- Plastic & Reconstructive Surgery Department, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kendrick To
- Division of Trauma and Orthopaedics, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Adam Hatoum
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Jessie Shea
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - K T Matthew Seah
- Division of Trauma and Orthopaedics, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Wasim Khan
- Division of Trauma and Orthopaedics, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Charles M Malata
- Plastic & Reconstructive Surgery Department, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Cambridge Breast Unit, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- School of Medicine, Anglia Ruskin University, Cambridge & Chelmsford, UK
| |
Collapse
|
26
|
Could cold plasma act synergistically with allogeneic mesenchymal stem cells to improve wound skin regeneration in a large size animal model? Res Vet Sci 2021; 136:97-110. [PMID: 33596495 DOI: 10.1016/j.rvsc.2021.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/05/2021] [Accepted: 01/24/2021] [Indexed: 02/06/2023]
Abstract
Skin wound healing may sometimes lead to open sores that persist for long periods and expensive hospitalization is needed. Among different kinds of therapeutic innovative approaches, mesenchymal stem cells (MSCs) and low-temperature atmospheric pressure cold plasma (ionized gas) have been recently tested to improve this regenerative process. To optimize wound healing the present study intended to combine, for the first time, these two novel approaches in a large size animal wound healing model with the aim of assessing the putative dual beneficial effects. Based on clinical, histopathological, and molecular results a synergistic action in a second intention healing wound in sheep has been observed. Experimental wounds treated with cold plasma and MSCs showed a slower but more effective healing compared to the single treatment, as observed in previous studies. The combined treatment improved the correct development of skin appendages and structural proteins of the dermis showing the potential of the dual combination as a safe and effective tool for skin regeneration in the veterinary clinical field.
Collapse
|
27
|
An Update on the Potential of Mesenchymal Stem Cell Therapy for Cutaneous Diseases. Stem Cells Int 2021; 2021:8834590. [PMID: 33505474 PMCID: PMC7806381 DOI: 10.1155/2021/8834590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/21/2020] [Accepted: 12/25/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem or stromal cells (MSCs) are nonhematopoietic postnatal stem cells with self-renewal, multipotent differentiation, and potent immunomodulatory and anti-inflammatory capabilities, thus playing an important role in tissue repair and regeneration. Numerous clinical and preclinical studies have demonstrated the potential application of MSCs in the treatment of tissue inflammation and immune diseases, including inflammatory skin diseases. Therefore, understanding the biological and immunological characteristics of MSCs is important to standardize and optimize MSC-based regenerative therapy. In this review, we highlight the mechanisms underlying MSC-mediated immunomodulation and tissue repair/regeneration and present the latest development of MSC-based clinical trials on cutaneous diseases.
Collapse
|
28
|
Beeken LJ, Ting DS, Sidney LE. Potential of mesenchymal stem cells as topical immunomodulatory cell therapies for ocular surface inflammatory disorders. Stem Cells Transl Med 2021; 10:39-49. [PMID: 32896982 PMCID: PMC7780815 DOI: 10.1002/sctm.20-0118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/10/2020] [Accepted: 07/30/2020] [Indexed: 12/17/2022] Open
Abstract
Ocular surface inflammatory disorders (OSIDs) are a group of highly prevalent, heterogeneous diseases that display a variety of aetiologies and symptoms and are risk factors for serious complications, including ocular and cornea impairment. Corneal inflammation is a common factor of all OSIDs, regardless of their cause or symptoms. Current medications include over-the-counter lubricating eye drops, corticosteroids, and ciclosporin, which either do not treat the corneal inflammation or have been associated with multiple side effects leading to alternative treatments being sought. Regenerative medicine cell therapies, particularly mesenchymal stem cells (MSCs), have shown great promise for immunosuppression and disease amelioration across multiple tissues, including the cornea. However, for successful development and clinical translation of MSC therapy for OSIDs, significant problems must be addressed. This review aims to highlight considerations, including whether the source of MSC isolation impacts the efficacy and safety of the therapy, in addition to assessing the feasibility of MSC topical application to the cornea and ocular surface through analysis of potential scaffolds and cell carriers for application to the eye. The literature contains limited data assessing MSCs incorporated into scaffolds for corneal administration, thus here we highlight the necessity of further investigations to truly exploit the potential of an MSC-based cell therapy for the treatment of OSIDs.
Collapse
Affiliation(s)
- Lydia J. Beeken
- Academic Ophthalmology, Division of Clinical NeurosciencesUniversity of Nottingham, Queens Medical Centre CampusNottinghamUK
| | - Darren S.J. Ting
- Academic Ophthalmology, Division of Clinical NeurosciencesUniversity of Nottingham, Queens Medical Centre CampusNottinghamUK
| | - Laura E. Sidney
- Academic Ophthalmology, Division of Clinical NeurosciencesUniversity of Nottingham, Queens Medical Centre CampusNottinghamUK
| |
Collapse
|
29
|
Liu J, Qiu X, Lv Y, Zheng C, Dong Y, Dou G, Zhu B, Liu A, Wang W, Zhou J, Liu S, Liu S, Gao B, Jin Y. Apoptotic bodies derived from mesenchymal stem cells promote cutaneous wound healing via regulating the functions of macrophages. Stem Cell Res Ther 2020; 11:507. [PMID: 33246491 PMCID: PMC7694913 DOI: 10.1186/s13287-020-02014-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 11/03/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND As the major interface between the body and the external environment, the skin is liable to various injuries. Skin injuries often lead to severe disability, and the exploration of promising therapeutic strategies is of great importance. Exogenous mesenchymal stem cell (MSC)-based therapy is a potential strategy due to the apparent therapeutic effects, while the underlying mechanism is still elusive. Interestingly, we observed the extensive apoptosis of exogenous bone marrow mesenchymal stem cells (BMMSCs) in a short time after transplantation in mouse skin wound healing models. Considering the roles of extracellular vesicles (EVs) in intercellular communication, we hypothesized that the numerous apoptotic bodies (ABs) released during apoptosis may partially contribute to the therapeutic effects. METHODS ABs derived from MSCs were extracted, characterized, and applied in mouse skin wound healing models, and the therapeutic effects were evaluated. Then, the target cells of ABs were explored, and the effects of ABs on macrophages were investigated in vitro. RESULTS We found ABs derived from MSCs promoted cutaneous wound healing via triggering the polarization of macrophages towards M2 phenotype. In addition, the functional converted macrophages further enhanced the migration and proliferation abilities of fibroblasts, which together facilitated the wound healing process. CONCLUSIONS Collectively, our study demonstrated that transplanted MSCs promoted cutaneous wound healing partially through releasing apoptotic bodies which could convert the macrophages towards an anti-inflammatory phenotype that plays a crucial role in the tissue repair process.
Collapse
Affiliation(s)
- Jin Liu
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.,State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xinyu Qiu
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yajie Lv
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.,Department of Dermatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Chenxi Zheng
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yan Dong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Geng Dou
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Bin Zhu
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.,Department of Stomatology, General Hospital of Tibet Military Region, Lhasa, 850007, Tibet, China
| | - Anqi Liu
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Wei Wang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, Fourth Military Medical University, Xi'an, China
| | - Jun Zhou
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Siying Liu
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Bo Gao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
30
|
Advanced Medical Therapies in the Management of Non-Scarring Alopecia: Areata and Androgenic Alopecia. Int J Mol Sci 2020; 21:ijms21218390. [PMID: 33182308 PMCID: PMC7664905 DOI: 10.3390/ijms21218390] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/28/2022] Open
Abstract
Alopecia is a challenging condition for both physicians and patients. Several topical, intralesional, oral, and surgical treatments have been developed in recent decades, but some of those therapies only provide partial improvement. Advanced medical therapies are medical products based on genes, cells, and/or tissue engineering products that have properties in regenerating, repairing, or replacing human tissue. In recent years, numerous applications have been described for advanced medical therapies. With this background, those therapies may have a role in the treatment of various types of alopecia such as alopecia areata and androgenic alopecia. The aim of this review is to provide dermatologists an overview of the different advanced medical therapies that have been applied in the treatment of alopecia, by reviewing clinical and basic research studies as well as ongoing clinical trials.
Collapse
|
31
|
Burja B, Barlič A, Erman A, Mrak-Poljšak K, Tomšič M, Sodin-Semrl S, Lakota K. Human mesenchymal stromal cells from different tissues exhibit unique responses to different inflammatory stimuli. Curr Res Transl Med 2020; 68:217-224. [PMID: 32843323 DOI: 10.1016/j.retram.2020.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 04/30/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Mesenchymal stromal cell (MSC) - based therapies are emerging as promising treatment of various autoimmune diseases, however the utility of different MSC tissue sources remains elusive. We aimed to characterize MSC from different origins, namely bone marrow (BM), adipose tissue (AT) and umbilical cord (UC) and determine their functional effects on normal human lung fibroblasts (NHLF). METHODS BM-, AT- or UC-MSC were isolated each from 3 different healthy donors. The gene expression and protein secretion were analyzed at basal level, along with TNFα-, IL-1β- and SAA- stimulated cells using real-time PCR and Luminex technology. Effect of conditioned medium (CM) from different MSC sources on migration was determined with wound scratch assay, while mitotic and apoptotic rates were studied using immunofluorescence microscopy. RESULTS BM-MSC expressed highest basal mRNA levels of SDF1 and VCAM-1, while other genes were similarly expressed between MSC origins. TNFα priming of AT-MSC gained a prominent increase in IDO1 and CCL5 gene expression, with 928-fold and 4396-fold changes, respectively. Among all tissue sources, basal UC-MSC released highest protein levels of most measured analytes, including IL-6, IL-8, MCP-1, ICAM1, HGF, MMP1 and CH3L1. BM- and AT-MSC derived CM enhanced wound closure in NHLF, while an opposite effect was observed with UC-MSC derived CM. Our data also suggests that MSC-CM could contribute to decreased mitotic potential and increased apoptotic rate in lung fibroblasts. CONCLUSIONS Our study highlights origin-specific MSC profile differences and emphasizes a heterogenic response of different MSC to inflammatory stimuli.
Collapse
Affiliation(s)
- Blaž Burja
- University Medical Centre Ljubljana, Department of Rheumatology, Ljubljana, Slovenia; University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia.
| | | | - Andreja Erman
- University of Ljubljana, Faculty of Medicine, Institute of Cell Biology, Ljubljana, Slovenia.
| | - Katjuša Mrak-Poljšak
- University Medical Centre Ljubljana, Department of Rheumatology, Ljubljana, Slovenia.
| | - Matija Tomšič
- University Medical Centre Ljubljana, Department of Rheumatology, Ljubljana, Slovenia; University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia.
| | - Snezna Sodin-Semrl
- University Medical Centre Ljubljana, Department of Rheumatology, Ljubljana, Slovenia; University of Primorska, FAMNIT, Koper, Slovenia.
| | - Katja Lakota
- University Medical Centre Ljubljana, Department of Rheumatology, Ljubljana, Slovenia; University of Primorska, FAMNIT, Koper, Slovenia.
| |
Collapse
|
32
|
Shojaei-Ghahrizjani F, Rahmati S, Mirzaei SA, Banitalebi-Dehkordi M. Does survivin overexpression enhance the efficiency of fibroblast cell-based wound therapy? Mol Biol Rep 2020; 47:5851-5864. [PMID: 32691274 DOI: 10.1007/s11033-020-05656-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/08/2020] [Indexed: 11/28/2022]
Abstract
Cell-based wound therapy is faced with some limiting factors that decrease the therapeutic efficacy of transplanted cells. In this study, we aimed to genetically modify fibroblast cells with anti-apoptotic Survivin gene (Birc5) before cell transplantation. In vitro, pIRES2-eGFP-Survivin plasmid was transfected into the fibroblast cells and the growth curve was evaluated for transfected and normal cells performing MTT assay. In vivo, two 6-diameter cutaneous wounds were created at mice dorsal skin. Fibrin clot was used as a delivery vehicle to transfer cells into the wound bed. The effects of four treatment groups including (a) Cell-SVV-Clot (b) Cell-GFP-Clot, (c) Normal cell-Clot and, (d) Clot alone were evaluated. After 1,2,3,7 and 14 days post-transplantation, the wounds were photographed for evaluating the wound closure rate and wound samples were obtained. Angiogenesis and formation of granulated tissue were assessed via H&E staining for wound samples. The expression levels of Survivin, VEGF, and bFGF genes were also determined using qRT-PCR. The MTT assay showed similar proliferation potential of transfected cells with normal cells verifying that Survivin had no detrimental effect. Compared to the Normal cell-Clot group, the Survivin overexpression was seen for 3 days in the Cell-SVV-Clot group verifying the cell survival during the early stage of wound healing. The Survivin further upregulated VEGF and bFGF expressions resulting in more angiogenesis and formation of granulated tissue by day 3 and 14. The treated wounds with Cell-SVV-Clot were regenerated with a higher wound closure rate by day 7 compared to Normal cell-Clot and Clot groups. Survivin enhanced wound healing through induction of VEGF and bFGF at particular times post-wounding that led to a more structured-epidermis with higher angiogenesis and granulation tissue formation rate.
Collapse
Affiliation(s)
- Fereshteh Shojaei-Ghahrizjani
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shima Rahmati
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mehdi Banitalebi-Dehkordi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
33
|
Piejko M, Radziun K, Bobis-Wozowicz S, Waligórska A, Zimoląg E, Nessler M, Chrapusta A, Madeja Z, Drukała J. Adipose-Derived Stromal Cells Seeded on Integra ® Dermal Regeneration Template Improve Post-Burn Wound Reconstruction. Bioengineering (Basel) 2020; 7:bioengineering7030067. [PMID: 32630660 PMCID: PMC7552717 DOI: 10.3390/bioengineering7030067] [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: 06/10/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/29/2022] Open
Abstract
Fibrosis of burn-related wounds remains an unresolved clinical issue that leads to patient disability. The aim of this study was to assess the efficacy of the transplantation of adipose-derived stromal cells seeded onto a collagen-based matrix in the reconstruction of burn-related scars. Here, we characterized an in vitro interaction between adipose-derived stromal cells and a collagen-based matrix, Integra®DRT. Our results show that transcription of pro-angiogenic, remodeling, and immunomodulatory factors was more significant in adipose-derived stromal cells than in fibroblasts. Transcription of metalloproteinases 2 and 9 is positively correlated with the collagenolytic activity of the adipose-derived stromal cells seeded onto Integra®DRT. The increase in the enzymatic activity corresponds to the decrease in the elasticity of the whole construct. Finally, we validated the treatment of a post-excision wound using adipose-derived stromal cells and an Integra®DRT construct in a 25-year-old woman suffering from burn-related scars. Scarless healing was observed in the area treated by adipose-derived stromal cells and the Integra®DRT construct but not in the reference area where Integra®DRT was applied without cells. This clinical observation may be explained by in vitro findings: Enhanced transcription of the vascular endothelial growth factor as well as remodeling of the collagen-based matrix decreased mechanical stress. Our experimental treatment demonstrated that the adipose-derived stromal cells seeded onto Integra®DRT exhibit valuable properties that may improve post-excision wound healing and facilitate skin regeneration without scars.
Collapse
Affiliation(s)
- Marcin Piejko
- Cell Bank, Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.P.); (K.R.); (S.B.-W.); (E.Z.); (Z.M.)
- Department of General Surgery, Jagiellonian University Medical College, 31-215 Krakow, Poland
| | - Karolina Radziun
- Cell Bank, Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.P.); (K.R.); (S.B.-W.); (E.Z.); (Z.M.)
| | - Sylwia Bobis-Wozowicz
- Cell Bank, Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.P.); (K.R.); (S.B.-W.); (E.Z.); (Z.M.)
| | - Agnieszka Waligórska
- Department of Cell Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
| | - Eliza Zimoląg
- Cell Bank, Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.P.); (K.R.); (S.B.-W.); (E.Z.); (Z.M.)
| | - Michał Nessler
- Malopolska Center for Burns and Plastic Surgery, The Ludwik Rydygier Hospital, 31-826 Krakow, Poland; (M.N.); (A.C.)
| | - Anna Chrapusta
- Malopolska Center for Burns and Plastic Surgery, The Ludwik Rydygier Hospital, 31-826 Krakow, Poland; (M.N.); (A.C.)
| | - Zbigniew Madeja
- Cell Bank, Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.P.); (K.R.); (S.B.-W.); (E.Z.); (Z.M.)
| | - Justyna Drukała
- Cell Bank, Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (M.P.); (K.R.); (S.B.-W.); (E.Z.); (Z.M.)
- Correspondence:
| |
Collapse
|
34
|
Sylakowski K, Bradshaw A, Wells A. Mesenchymal Stem Cell/Multipotent Stromal Cell Augmentation of Wound Healing: Lessons from the Physiology of Matrix and Hypoxia Support. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1370-1381. [PMID: 32294456 PMCID: PMC7369572 DOI: 10.1016/j.ajpath.2020.03.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/28/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022]
Abstract
Cutaneous wounds requiring tissue replacement are often challenging to treat and result in substantial economic burden. Many of the challenges inherent to therapy-mediated healing are due to comorbidities of disease and aging that render many wounds as chronic or nonhealing. Repeated failure to resolve chronic wounds compromises the reserve or functioning of localized reparative cells. Transplantation of mesenchymal stem cells/multipotent stromal cells (MSCs) has been proposed to augment the reparative capacity of resident cells within the wound bed to overcome stalled wound healing. However, MSCs face a variety of challenges within the wound micro-environment that curtail their survival after transplantation. MSCs are naturally pro-angiogenic and proreparative, and thus numerous techniques have been attempted to improve their survival and efficacy after transplantation, many with little impact. These setbacks have prompted researchers to re-examine the normal wound bed physiology, resulting in new approaches to MSC transplantation using extracellular matrix proteins and hypoxia preconditioning. These studies have also led to new insights on associated intracellular mechanisms, particularly autophagy, which play key roles in further regulating MSC survival and paracrine signaling. This review provides a brief overview of cutaneous wound healing with discussion on how extracellular matrix proteins and hypoxia can be utilized to improve MSC retention and therapeutic outcome.
Collapse
Affiliation(s)
- Kyle Sylakowski
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; R&D Service, VA Pittsburgh Health System, Pittsburgh, Pennsylvania
| | - Andrew Bradshaw
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; R&D Service, VA Pittsburgh Health System, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania; R&D Service, VA Pittsburgh Health System, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
35
|
Li P, Ma X, Jin W, Li X, Hu J, Jiang X, Guo X. Effects of local injection and intravenous injection of allogeneic bone marrow mesenchymal stem cells on the structure and function of damaged anal sphincter in rats. J Tissue Eng Regen Med 2020; 14:989-1000. [PMID: 32537834 DOI: 10.1002/term.3079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 04/09/2020] [Accepted: 05/11/2020] [Indexed: 12/21/2022]
Abstract
Anal sphincter injury leads to damage to the anal structure and functions and has been identified as a major risk factor for fecal incontinence. Bone marrow mesenchymal stem cells (BMSCs) with capacities of multidifferentiation, paracrine, and low immunogenicity have been widely used in tissue repair and regeneration. The primary objective of this research was to compare the effects of different injection therapies of BMSCs on the injured anal sphincters. Ninety-six Sprague-Dawley female rats were randomly divided into four groups (n = 24 each): intravenous injection, local injection, sham operation, and normal control. For the first three groups, 25% removal of the anal sphincter complex was performed and 0.3-ml phosphate-buffered saline (PBS) (containing 107 green fluorescent protein-labeled allogeneic BMSCs) was given accordingly to the treatment group 24 h after operation for 7 consecutive days. The sham operation group was injected with 0.3-ml PBS only. All cases had undergone evaluation in the 1st, 7th, 14th, and 28th postoperative days. The rats were sacrificed on the 28th postoperative day, and the anal sphincters were dissected to be analyzed by morphological examination. At 14 days postoperatively, local injection of BMSC significantly improved the peak contraction pressure, electromyography amplitude, and frequency of the injured anal sphincter compared with tail vein, but there was no significant difference in resting pressure until 28 days after sphincterectomy. Masson staining results confirmed that the local injection group had significantly more new muscles on the wound. BMSC could remarkably improve peak contraction pressure, electromyography amplitude, and muscle fibers on the wound, and local injection is superior to intravenous injection.
Collapse
Affiliation(s)
- Peng Li
- Department of Anorectal, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoying Ma
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wenqi Jin
- Department of Anorectal, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojia Li
- Department of Anorectal, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Hu
- Department of Anorectal, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoxue Jiang
- Department of Anorectal Surgery, Shanghai Eighth People's Hospital, Shanghai, China
| | - Xiutian Guo
- Department of Anorectal, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
36
|
Fang Z, Chen H, Teng W, Qiu P, Yu X, Luo C, Zhu Y, You Y, Fu Q, Ji J, Wang B, Yao K. Systemic Transplantation of Eyelid Adipose‐Derived Stem Cells for Antifibrotic Treatment. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Zhi Fang
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| | - Hui Chen
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| | - Wenqi Teng
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| | - Peijin Qiu
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| | - Xiaoning Yu
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| | - Chenqi Luo
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| | - Yanan Zhu
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| | - Yongsheng You
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| | - Qiuli Fu
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| | - Junfeng Ji
- Center of Stem Cell and Regenerative MedicineSchool of MedicineZhejiang University Hangzhou 310058 China
| | - Ben Wang
- Cancer Institute, Key Laboratory of Cancer Prevention and InterventionChina National Ministry of EducationThe Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
- Institute of Translational MedicineZhejiang University Hangzhou 310029 China
| | - Ke Yao
- Eye Institute of Zhejiang UniversityEye Center of the Second Affiliated HospitalSchool of MedicineZhejiang University Hangzhou 310009 China
| |
Collapse
|
37
|
Huang Y, Wang Y, Wang X, Lin L, Wang P, Sun J, Jiang L. The Effects of the Transforming Growth Factor-β1 (TGF-β1) Signaling Pathway on Cell Proliferation and Cell Migration are Mediated by Ubiquitin Specific Protease 4 (USP4) in Hypertrophic Scar Tissue and Primary Fibroblast Cultures. Med Sci Monit 2020; 26:e920736. [PMID: 32308208 PMCID: PMC7191961 DOI: 10.12659/msm.920736] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hypertrophic scar results from an abnormal repair response to trauma in the skin and involves fibroblasts proliferation with increased collagen deposition. Transforming growth factor-ß1 (TGF-ß1) and TGF-ß receptor type I (TGF-ßR1) are involved in tissue repair and are increased by ubiquitin-specific protease 4 (USP4). This study aimed to investigate the effects of TGF-ßR1 and USP4 in human tissue samples of hypertrophic scar and on cell proliferation and cell migration in primary fibroblast cultures in vitro. MATERIAL AND METHODS Skin excision tissue samples with adjacent normal skin were obtained from 15 patients with hypertrophic scar, which provided tissue sections and primary fibroblast culture for analysis. Immunohistochemistry detected the expression of USP4 and TGF-ßR1 in tissue sections. MicroRNA (miRNAs) expression levels were measured by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot was performed to measure protein expression levels. Cultured skin fibroblasts were investigated using immunofluorescence staining. Fibroblast proliferation, apoptosis, and migration were measured with the Cell Counting Kit-8 (CCK-8) assay, flow cytometry, and a wound-healing assay, respectively. RESULTS The expression of USP4 and TGF-ßR1 in hypertrophic scar were increased compared with normal skin. Fibroblasts cultured from hypertrophic scar tissue showed increased expression of of USP4 and TGF-ßR1. Fibroblast transfection with USP4 short-interfering RNA (siRNA) resulted in reduced fibroblast proliferation and migration, and increased apoptosis. Downregulation of USP4 inhibited the expression of TGF-ßR1 protein and increased the expression levels of Smad7 protein. CONCLUSIONS USP4 regulated the proliferation, migration, and apoptosis of hypertrophic scar fibroblasts by regulating the TGF-ß1 signaling pathway.
Collapse
Affiliation(s)
- Yong Huang
- Department of Plastic and Aesthetic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Yuting Wang
- Department of Plastic and Aesthetic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Xueming Wang
- Department of Plastic and Aesthetic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Lixin Lin
- Department of Plastic and Aesthetic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Peng Wang
- Department of Plastic and Aesthetic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Junjun Sun
- Department of Plastic and Aesthetic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Lei Jiang
- Department of Plastic and Aesthetic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| |
Collapse
|
38
|
Tao K, Bai X, Zhang D, Liu M, Zhang Y, Han F, Yang X, Han J, Hu D. Encapsulation of troglitazone and AVE0991 by gelation microspheres promotes epithelial transformation of adipose-derived stem cells. Mol Cell Probes 2020; 51:101543. [PMID: 32105703 DOI: 10.1016/j.mcp.2020.101543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/23/2020] [Indexed: 11/26/2022]
Abstract
Deformities in human soft tissue caused by trauma or burn present a difficult problem in plastic surgery. In this study, we encapsulated troglitazone and angiotensin 1-7 mimetic AVE0991 in gelation microspheres with the goal of inducing epithelial transformation for potential applications in tissue reconstruction. After troglitazone or AVE0991 were encapsulated to gelation microspheres, their release kinetics and bioactivity were examined. Surface morphology and diameter of the gelation microspheres were evaluated using light microscopy. The release of the drugs was assessed in the presence of human adipose-derived stem cells (ADSCs). Treatment with troglitazone microspheres increased cell viability and activated the β-catenin in ADSCs. Moreover, the AVE0991 microspheres also increased cell viability and C-myc expression of ADSCs. These results showed that troglitazone and AVE0991 microspheres promoted the activity of ADSCs. Furthermore, ADSCs were co-treated with troglitazone and AVE0991 microspheres. Western blot and immunofluorescent staining showed that co-treatment with troglitazone and AVE0991 microspheres elevated the expression of epithelialization associated protein CK14 in ADSCs. In conclusion, our findings indicate that microspheres with troglitazone and AVE0991 can significantly improve the viability and epithelialization of ADSCs, which provides a new approach for the construction of tissue-engineered skin.
Collapse
Affiliation(s)
- Ke Tao
- Department of Burn Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaan'xi, China
| | - Xiaozhi Bai
- Department of Burn Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaan'xi, China
| | - Dongliang Zhang
- Department of Burn Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaan'xi, China
| | - Mengdong Liu
- Department of Burn Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaan'xi, China
| | - Yue Zhang
- Department of Burn Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaan'xi, China
| | - Fu Han
- Department of Burn Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaan'xi, China
| | - Xuekang Yang
- Department of Burn Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaan'xi, China
| | - Juntao Han
- Department of Burn Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaan'xi, China
| | - Dahai Hu
- Department of Burn Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaan'xi, China.
| |
Collapse
|
39
|
Abdal-Wahab M, Abdel Ghaffar KA, Ezzatt OM, Hassan AAA, El Ansary MMS, Gamal AY. Regenerative potential of cultured gingival fibroblasts in treatment of periodontal intrabony defects (randomized clinical and biochemical trial). J Periodontal Res 2020; 55:441-452. [PMID: 32080858 DOI: 10.1111/jre.12728] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 11/22/2019] [Accepted: 12/11/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Defective cellular elements constitute an important challenge to achieve predictable periodontal regeneration. In an attempt to improve the cellularity of periodontal defects, gingival fibroblasts were implanted without their associated extracellular elements in periodontal defects to expose them to periodontal tissue mediators. In order to investigate the regenerative potential of gingival fibroblasts translocated into periodontal defects, the present study was designed to clinically and biochemically investigate the use of gingival fibroblasts (GF) and their associated mesenchymal stem cells (GMSC) in the treatment of intrabony periodontal defects. METHODS A total of 20 subjects were randomly divided into two groups (n = 20). Group I: ten patients were included with ten intrabony periodontal defects that received β-calcium triphosphate (β-TCP) followed by collagen membrane defect coverage, while group II: (10 patients) ten periodontal defects received cultured gingival fibroblasts (GF) on the β-TCP scaffold and covered by a collagen membrane. The clinical evaluation was carried out at the beginning and at 6 months. Gingival crevicular fluid (GCF) samples were collected directly from the test sites for the quantitative measurement of PDGF-BB and BMP-2 using the ELISA kit at 1, 7, 14, and 21 days after surgery. RESULTS Group II reported a significantly greater reduction in vertical pocket depth (VPD) and CAL gain compared with group I after 6 months. Radiographic bone gain was statistically higher in group II compared with group I. A significantly higher concentration of PDGF-BB was observed in group II on days 1, 3, and 7 compared with group I. CONCLUSIONS Translocation of gingival fibroblasts from gingival tissue to periodontal defects could be a promising option that increases cellular elements with regeneration potential. The concept of total isolation of gingival fibroblasts using occlusive membranes must be re-evaluated.
Collapse
Affiliation(s)
- Mahetab Abdal-Wahab
- Department of Periodontology, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| | | | - Ola M Ezzatt
- Department of Periodontology, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| | | | | | - Ahmed Y Gamal
- Department of Periodontology, Faculty of Dentistry, Nahda University, Cairo, Egypt
| |
Collapse
|
40
|
A Novel Method to Optimize Autologous Adipose Tissue Recovery with Extracellular Matrix Preservation. Processes (Basel) 2020. [DOI: 10.3390/pr8010088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
This work aims to characterize a new method to recover low-manipulated human adipose tissue, enriched with adipose tissue-derived mesenchymal stem cells (ATD-MSCs) for autologous use in regenerative medicine applications. Lipoaspirated fat collected from patients was processed through Lipocell, a Class II-a medical device for dialysis of adipose tissue, by varying filter sizes and washing solutions. ATD-MSC yield was measured with flow cytometry after stromal vascular fraction (SVF) isolation in fresh and cultured samples. Purification from oil and blood was measured after centrifugation with spectrophotometer analysis. Extracellular matrix preservation was assessed through hematoxylin and eosin (H&E) staining and biochemical assay for total collagen, type-2 collagen, and glycosaminoglycans (GAGs) quantification. Flow cytometry showed a two-fold increase of ATD-MSC yield in treated samples in comparison with untreated lipoaspirate; no differences where reported when varying filter size. The association of dialysis and washing thoroughly removed blood and oil from samples. Tissue architecture and extracellular matrix integrity were unaltered after Lipocell processing. Dialysis procedure associated with Ringer’s lactate preserves the proliferation ability of ATD-MSCs in cell culture. The characterization of the product showed that Lipocell is an efficient method for purifying the tissue from undesired byproducts and preserving ATD-MSC vitality and extracellular matrix (ECM) integrity, resulting in a promising tool for regenerative medicine applications.
Collapse
|
41
|
Lingzhi Z, Meirong L, Xiaobing F. Biological approaches for hypertrophic scars. Int Wound J 2019; 17:405-418. [PMID: 31860941 DOI: 10.1111/iwj.13286] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/01/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022] Open
Abstract
Scar formation is usually the pathological consequence of skin trauma. And hypertrophic scars (HSs) frequently occur in people after being injured deeply. HSs are unusually considered as the result of tissue contraction and excessive extracellular matrix component deposition. Myofibroblasts, as the effector cells, mainly differentiated from fibroblasts, play the crucial role in the pathophysiology of HSs. A number of growth factors, inflammatory cytokines involved in the process of HS occurrence. Currently, with in-depth exploration and clinical research of HSs, various creative and effective treatments budded. In here, we summarize the progress in the molecular mechanism of HSs, and review the available biotherapeutic methods for their pathophysiological characteristics. Additionally, we further prospected that the comprehensive therapy may be more suitable for HS treatment.
Collapse
Affiliation(s)
- Zhong Lingzhi
- Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| | - Li Meirong
- Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China.,Central Laboratory, Trauma Treatment Center, Chinese PLA General Hospital Hainan Branch, Sanya, China
| | - Fu Xiaobing
- Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
42
|
Li X, Wei Z, Li B, Li J, Lv H, Wu L, Zhang H, Yang B, Zhu M, Jiang J. In vivo migration of Fe 3O 4@polydopamine nanoparticle-labeled mesenchymal stem cells to burn injury sites and their therapeutic effects in a rat model. Biomater Sci 2019; 7:2861-2872. [PMID: 31070196 DOI: 10.1039/c9bm00242a] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cell (MSC)-based therapy has emerged as a promising therapeutic strategy for tissue regeneration and repair. However, efficient targeted delivery to specific tissues remains an open challenge. Here, we non-invasively monitored the migration of MSCs labeled with Fe3O4@polydopamine nanoparticles (Fe3O4@PDA NPs) toward laser burn injury sites in a living rat model and evaluated the effects of the labeled MSCs at the injury site. The Fe3O4@PDA NPs could be effectively incorporated into the MSCs without any negative effects on stem cell properties. Furthermore, they enhanced the migration ability of the MSCs by up-regulating the expression level of C-X-C chemokine receptor type 4 (CXCR4). They also increased the secretion of some cytokines and the expression of healing-related genes in comparison with unlabeled MSCs. Labeled MSCs were intravenously administered into injured rats, and live imaging was performed to monitor MSC migration. Fluorescent signals of the labeled MSCs appeared at burn injury lesions 1 day after injection and then gradually increased up to 7 days. After 7 days, the group injected with the labeled MSCs showed less inflammation compared with those injected with the unlabeled MSCs. Additionally, the labeled MSC group showed increased cytokines and reduced pro-inflammatory factors compared with the unlabeled MSC group. The Fe3O4@PDA NPs enhanced stromal cell-derived factor-1/CXCR4-mediated MSC migration in vivo. Thus, we demonstrated the safety, feasibility, and potential efficacy of using the Fe3O4@PDA NPs for optimizing MSC-based therapeutic strategies for burn wound healing.
Collapse
Affiliation(s)
- Xiuying Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Emmerson S, Mukherjee S, Melendez-Munoz J, Cousins F, Edwards SL, Karjalainen P, Ng M, Tan KS, Darzi S, Bhakoo K, Rosamilia A, Werkmeister JA, Gargett CE. Composite mesh design for delivery of autologous mesenchymal stem cells influences mesh integration, exposure and biocompatibility in an ovine model of pelvic organ prolapse. Biomaterials 2019; 225:119495. [PMID: 31606680 DOI: 10.1016/j.biomaterials.2019.119495] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 08/20/2019] [Accepted: 09/11/2019] [Indexed: 12/21/2022]
Abstract
The widespread use of synthetic transvaginal polypropylene mesh for treating Pelvic Organ Prolapse (POP) has been curtailed due to serious adverse effects highlighted in 2008 and 2011 FDA warnings and subsequent legal action. We are developing new synthetic mesh to deliver endometrial mesenchymal stem cells (eMSC) to improve mesh biocompatibility and restore strength to prolapsed vaginal tissue. Here we evaluated knitted polyamide (PA) mesh in an ovine multiparous model using transvaginal implantation and matched for the degree of POP. Polyamide mesh dip-coated in gelatin and stabilised with 0.5% glutaraldehyde (PA/G) were used either alone or seeded with autologous ovine eMSC (eMSC/PA/G), which resulted in substantial mesh folding, poor tissue integration and 42% mesh exposure in the ovine model. In contrast, a two-step insertion protocol, whereby the uncoated PA mesh was inserted transvaginally followed by application of autologous eMSC in a gelatin hydrogel onto the mesh and crosslinked with blue light (PA + eMSC/G), integrated well with little folding and no mesh exposure. The autologous ovine eMSC survived 30 days in vivo but had no effect on mesh integration. The stiff PA/G constructs provoked greater myofibroblast and inflammatory responses in the vaginal wall, disrupted the muscularis layer and reduced elastin fibres compared to PA + eMSC/G constructs. This study identified the superiority of a two-step protocol for implanting synthetic mesh in cellular compatible composite constructs and simpler surgical application, providing additional translational value.
Collapse
Affiliation(s)
- S Emmerson
- Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, Australia; Department of Obstetrics and Gynaecology, Monash University, Wellington Road, Clayton, Melbourne, Australia
| | - S Mukherjee
- Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, Australia; Department of Obstetrics and Gynaecology, Monash University, Wellington Road, Clayton, Melbourne, Australia
| | | | - F Cousins
- Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, Australia
| | - S L Edwards
- CSIRO Manufacturing, Research Way, Clayton, Melbourne, Australia
| | - P Karjalainen
- Monash Health, Centre Road, Moorabbin, Melbourne, Australia
| | - M Ng
- Singapore Bioimaging Consortium, 1 Agency for Science, Technology and Research (A*STAR), 1 Biopolis Way, Singapore
| | - K S Tan
- Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, Australia
| | - S Darzi
- Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, Australia; Department of Obstetrics and Gynaecology, Monash University, Wellington Road, Clayton, Melbourne, Australia
| | - K Bhakoo
- Singapore Bioimaging Consortium, 1 Agency for Science, Technology and Research (A*STAR), 1 Biopolis Way, Singapore
| | - A Rosamilia
- Department of Obstetrics and Gynaecology, Monash University, Wellington Road, Clayton, Melbourne, Australia; Monash Health, Centre Road, Moorabbin, Melbourne, Australia
| | - J A Werkmeister
- Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, Australia; Department of Obstetrics and Gynaecology, Monash University, Wellington Road, Clayton, Melbourne, Australia
| | - C E Gargett
- Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, Australia; Department of Obstetrics and Gynaecology, Monash University, Wellington Road, Clayton, Melbourne, Australia.
| |
Collapse
|
44
|
Bradshaw A, Sylakowski K, Wells A. The Pro-reparative Engine: Stem Cells Aid Healing by Dampening Inflammation. CURRENT PATHOBIOLOGY REPORTS 2018; 6:109-115. [PMID: 30271682 DOI: 10.1007/s40139-018-0167-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Purpose of Review Stem cells have been proposed as sources for tissue replacement when healing does not occur. These cells could contribute directly to skin structures via differentiation, or via producing trophic factors that would 'educate' the micro-environment to encourage tissue repair. Studies in animals have supported both mechanisms, but translation to humans has been challenged by poor cell survival after transplantation. However, the improvement noted with even transient existence suggests another new possibility, that of suppressing the inflammatory response that limits regenerative healing. Herein, we will propose that this immunomodulatory aspect holds promise for promoting skin healing. Recent Findings We have found that stem cell transplantation into wounds can dampen both acute and chronic inflammation, leading to more regenerative-like healing and diminished scarring. Summary Wound healing could be improved by dampening inflammation both initially to allow for tissue replacement to proceed and late to reduce scarring.
Collapse
Affiliation(s)
- Andrew Bradshaw
- Departments of Pathology and Bioengineering, and the McGowan, Institute for Regenerative Medicine, University of Pittsburgh, and VA Pittsburgh Health System, Pittsburgh PA 15213 USA
| | - Kyle Sylakowski
- Departments of Pathology and Bioengineering, and the McGowan, Institute for Regenerative Medicine, University of Pittsburgh, and VA Pittsburgh Health System, Pittsburgh PA 15213 USA
| | - Alan Wells
- Departments of Pathology and Bioengineering, and the McGowan, Institute for Regenerative Medicine, University of Pittsburgh, and VA Pittsburgh Health System, Pittsburgh PA 15213 USA
| |
Collapse
|
45
|
Exosomal MicroRNAs Derived from Human Amniotic Epithelial Cells Accelerate Wound Healing by Promoting the Proliferation and Migration of Fibroblasts. Stem Cells Int 2018; 2018:5420463. [PMID: 30147728 PMCID: PMC6083635 DOI: 10.1155/2018/5420463] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/27/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023] Open
Abstract
Previous work in our laboratory demonstrated that exosomes derived from human amniotic epithelial cells (hAECs) accelerated wound healing by promoting the proliferation and migration of fibroblasts. It is reported that exosomes, which are carriers of the microRNAs (miRNAs) and proteins, play an important role in the regulation of cell-to-cell communication. However, it is still unclear precisely which molecule or which group of molecules carried within hAEC-derived exosomes (hAEC-Exos) mediated wound healing. Here, we explored purified hAEC-Exos together with either proteinase K (PROse) or RNase A on the effect of fibroblasts and cutaneous wound healing. Our experiments demonstrated that hAEC-Exos were positive for exosomal markers CD9, CD63, and CD81. Also, we found that hAEC-Exos could be internalized by fibroblasts and then stimulated cell migration and proliferation. However, the promotive effect of hAEC-Exos was abolished by pretreating hAEC-Exos with RNase A, not PROse. Importantly, in vivo wound healing assay showed that local injection of hAEC-Exos or PROse pretreated hAEC-Exos at skin wounds significantly accelerated wound healing. Our findings revealed an important role of exosomal miRNAs in wound healing.
Collapse
|
46
|
Munir H, Ward LSC, McGettrick HM. Mesenchymal Stem Cells as Endogenous Regulators of Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:73-98. [PMID: 30155623 DOI: 10.1007/978-3-319-78127-3_5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This chapter discusses the regulatory role of endogenous mesenchymal stem cells (MSC) during an inflammatory response. MSC are a heterogeneous population of multipotent cells that normally contribute towards tissue maintenance and repair but have garnered significant scientific interest for their potent immunomodulatory potential. It is through these physicochemical interactions that MSC are able to exert an anti-inflammatory response on neighbouring stromal and haematopoietic cells. However, the impact of the chronic inflammatory environment on MSC function remains to be determined. Understanding the relationship of MSC between resolution of inflammation and autoimmunity will both offer new insights in the use of MSC as a therapeutic, and also their involvement in the pathogenesis of inflammatory disorders.
Collapse
Affiliation(s)
- Hafsa Munir
- MRC Cancer Unit/Hutchison, University of Cambridge, Cambridge, UK
| | | | - Helen M McGettrick
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.
| |
Collapse
|