1
|
El Masri J, Fadlallah H, Al Sabsabi R, Afyouni A, Al-Sayegh M, Abou-Kheir W. Adipose-Derived Stem Cell Therapy in Spinal Cord Injury. Cells 2024; 13:1505. [PMID: 39273075 PMCID: PMC11394073 DOI: 10.3390/cells13171505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Spinal cord injury (SCI) is a serious condition accompanied by severe adverse events that affect several aspects of the patient's life, such as motor, sensory, and functional impairment. Despite its severe consequences, definitive treatment for these injuries is still missing. Therefore, researchers have focused on developing treatment strategies aimed at ensuring full recovery post-SCI. Accordingly, attention has been drawn toward cellular therapy using mesenchymal stem cells. Considering their wide availability, decreased immunogenicity, wide expansion capacity, and impressive effectiveness in many therapeutic approaches, adipose-derived stem cell (ADSC) injections in SCI cases have been investigated and showed promising results. In this review, SCI pathophysiology and ADSC transplantation benefits are discussed independently, together with SCI animal models and adipose stem cell preparation and application techniques. The mechanisms of healing in an SCI post-ADSC injection, the outcomes of this therapeutic approach, and current clinical trials are also deliberated, in addition to the challenges and future perspectives, aiming to encourage further research in this field.
Collapse
Affiliation(s)
- Jad El Masri
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
- Faculty of Medical Sciences, Lebanese University, Beirut 1533, Lebanon
| | - Hiba Fadlallah
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Rahaf Al Sabsabi
- Faculty of Medical Sciences, Lebanese University, Beirut 1533, Lebanon
| | - Ahmad Afyouni
- Faculty of Medical Sciences, Lebanese University, Beirut 1533, Lebanon
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi 2460, United Arab Emirates
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| |
Collapse
|
2
|
Eivazi Zadeh Z, Nour S, Kianersi S, Jonidi Shariatzadeh F, Williams RJ, Nisbet DR, Bruggeman KF. Mining human clinical waste as a rich source of stem cells for neural regeneration. iScience 2024; 27:110307. [PMID: 39156636 PMCID: PMC11326931 DOI: 10.1016/j.isci.2024.110307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Abstract
Neural diseases are challenging to treat and are regarded as one of the major causes of disability and morbidity in the world. Stem cells can provide a solution, by offering a mechanism to replace damaged circuitry. However, obtaining sufficient cell sources for neural regeneration remains a significant challenge. In recent years, waste-derived stem(-like) cells (WDS-lCs) extracted from both prenatal and adult clinical waste tissues/products, have gained increasing attention for application in neural tissue repair and remodeling. This often-overlooked pool of cells possesses favorable characteristics; including self-renewal, neural differentiation, secretion of neurogenic factors, cost-effectiveness, and low ethical concerns. Here, we offer a perspective regarding the biological properties, extraction protocols, and preclinical and clinical treatments where prenatal and adult WDS-lCs have been utilized for cell replacement therapy in neural applications, and the challenges involved in optimizing these approaches toward patient led therapies.
Collapse
Affiliation(s)
- Zahra Eivazi Zadeh
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Shirin Nour
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
- Polymer Science Group, Department of Chemical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sogol Kianersi
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences, University of Galway, Galway, Ireland
| | | | - Richard J. Williams
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
- iMPACT, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - David R. Nisbet
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU College of Health & Medicine, Canberra, ACT, Australia
- Research School of Chemistry, ANU College of Science, Canberra, ACT, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
- Founder and Scientific Advisory of Nano Status, Building 137, Sullivans Creek Rd, ANU, Acton, Canberra, ACT, Australia
| | - Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials Research, School of Engineering, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
3
|
Ullah MM, Collett JA, Monroe JC, Traktuev D, Coleman M, March KL, Basile DP. Subcutaneous injection of adipose stromal cell-secretome improves renal function and reduces inflammation in established acute kidney injury. Stem Cell Res Ther 2024; 15:119. [PMID: 38659070 PMCID: PMC11040889 DOI: 10.1186/s13287-024-03736-x] [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: 08/14/2023] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Adipose stromal cells (ASC) are a form of mesenchymal stromal cells that elicit effects primarily via secreted factors, which may have advantages for the treatment of injury or disease. Several previous studies have demonstrated a protective role for MSC/ASC on mitigating acute kidney injury but whether ASC derived factors could hasten recovery from established injury has not been evaluated. METHODS We generated a concentrated secretome (CS) of human ASC under well-defined conditions and evaluated its ability to improve the recovery of renal function in a preclinical model of acute kidney injury (AKI) in rats. 24 h following bilateral ischemia/reperfusion (I/R), rats were randomized following determination of plasma creatinine into groups receiving vehicle -control or ASC-CS treatment by subcutaneous injection (2 mg protein/kg) and monitored for evaluation of renal function, structure and inflammation. RESULTS Renal function, assessed by plasma creatinine levels, recovered faster in ASC-CS treated rats vs vehicle. The most prominent difference between the ASC-CS treated vs vehicle was observed in rats with the most severe degree of initial injury (Pcr > 3.0 mg/dl 24 h post I/R), whereas rats with less severe injury (Pcr < 2.9 mg/dl) recovered quickly regardless of treatment. The quicker recovery of ASC-treated rats with severe injury was associated with less tissue damage, inflammation, and lower plasma angiopoietin 2. In vitro, ASC-CS attenuated the activation of the Th17 phenotype in lymphocytes isolated from injured kidneys. CONCLUSIONS Taken together, these data suggest that ASC-CS represents a potent therapeutic option to improve established AKI.
Collapse
Affiliation(s)
- Md Mahbub Ullah
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Dr. MS 2063, Indianapolis, IN, 46202, USA
| | - Jason A Collett
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Dr. MS 2063, Indianapolis, IN, 46202, USA
| | - Jacob C Monroe
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Dr. MS 2063, Indianapolis, IN, 46202, USA
| | - Dmitry Traktuev
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
- Theratome Bio, Inc., Indianapolis, IN, USA
| | - Michael Coleman
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Keith L March
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
- Theratome Bio, Inc., Indianapolis, IN, USA
| | - David P Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Dr. MS 2063, Indianapolis, IN, 46202, USA.
| |
Collapse
|
4
|
Pagani A, Duscher D, Geis S, Klein S, Knoedler L, Panayi AC, Oliinyk D, Felthaus O, Prantl L. The Triple Adipose-Derived Stem Cell Exosome Technology as a Potential Tool for Treating Triple-Negative Breast Cancer. Cells 2024; 13:614. [PMID: 38607053 PMCID: PMC11011929 DOI: 10.3390/cells13070614] [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: 03/05/2024] [Revised: 03/26/2024] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Extracellular vesicles are pivotal mediators in intercellular communication, facilitating the exchange of biological information among healthy, pathological and tumor cells. Between the diverse subtypes of extracellular vesicles, exosomes have unique properties and clinical and therapeutical applications. Breast cancer ranks as one of the most prevalent malignancies across the globe. Both the tumor core and its surrounding microenvironment engage in a complex, orchestrated interaction that facilitates cancer's growth and spread. METHODS The most significant PubMed literature about extracellular vesicles and Adipose-Derived Stem Cell Exosomes and breast cancer was selected in order to report their biological properties and potential applications, in particular in treating triple-negative breast cancer. RESULTS Adipose-Derived Stem Cell Exosomes represent a potential tool in targeting triple-negative breast cancer cells at three main levels: the tumor core, the tumor microenvironment and surrounding tissues, including metastases. CONCLUSIONS The possibility of impacting triple-negative breast cancer cells with engineered Adipose-Derived Stem Cell Exosomes is real. The opportunity to translate our current in vitro analyses into a future in vivo scenario is even more challenging.
Collapse
Affiliation(s)
- Andrea Pagani
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany (S.K.); (O.F.); (L.P.)
| | - Dominik Duscher
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany (S.K.); (O.F.); (L.P.)
| | - Sebastian Geis
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany (S.K.); (O.F.); (L.P.)
| | - Silvan Klein
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany (S.K.); (O.F.); (L.P.)
| | - Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany (S.K.); (O.F.); (L.P.)
| | - Adriana C. Panayi
- Department of Plastic, Hand and Reconstructive Surgery, BG Klinik Ludwigshafen, University of Heidelberg, Ludwig-Guttmann-Straße 13, 67071 Ludwigshafen, Germany
| | - Dmytro Oliinyk
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany (S.K.); (O.F.); (L.P.)
| | - Oliver Felthaus
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany (S.K.); (O.F.); (L.P.)
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany (S.K.); (O.F.); (L.P.)
| |
Collapse
|
5
|
Hausen MDA, Moraes ADS, Pedrini F, Grabarz F, Camilli JA, Duek EADR. Crosslinked Collagen-Hyaluronic Acid Scaffold Enhances Interleukin-10 Under Co-Culture of Macrophages And Adipose-Derived Stem Cells. Macromol Biosci 2024; 24:e2300270. [PMID: 37700543 DOI: 10.1002/mabi.202300270] [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: 06/19/2023] [Revised: 08/25/2023] [Indexed: 09/14/2023]
Abstract
The skin, the human body's largest organ, possesses a protective barrier that renders it susceptible to various injuries, including burns. Following burn trauma, the inflammatory process triggers both innate and adaptive immune responses, leading to the polarization of macrophages into two distinct phenotypes: the pro-inflammatory M1 and the anti-inflammatory M2. This dual response sets the stage for wound healing and subsequent tissue regeneration. Contributing to this transition from M1 to M2 polarization are human adipose-derived stem cells (ASCs), which employ paracrine signaling and inflammation suppression to enhance the remodeling phase. ASCs, when combined with biocompatible polymers, can be integrated into functional scaffolds. This study introduces an 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide-crosslinked (EDC-crosslinked) collagen-hyaluronic acid (Col-HA) scaffold assembled with ASCs, designed as a natural biomaterial device to modulate macrophage behavior in vitro under co-culture conditions. This innovation aims to improve wound healing processes. The EDC-crosslinked Col-HA scaffold favored the release of anti-inflammatory cytokines by ASCs, which indicated the M2 prevalence. In tissue engineering, a critical objective lies in the development of functional biomaterials capable of guiding specific tissue responses, notably the control of inflammatory processes. Thus, this research not only presents original findings but also points toward a promising avenue within regenerative medicine.
Collapse
Affiliation(s)
- Moema de Alencar Hausen
- Surgery Department, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo, Postgraduate Program in Biomaterials and Regenerative Medicine (PPBMR), Laboratory of Biomaterials, Sorocaba, 18030-070, Brazil
| | - Ariana de Souza Moraes
- Surgery Department, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo, Postgraduate Program in Biomaterials and Regenerative Medicine (PPBMR), Laboratory of Biomaterials, Sorocaba, 18030-070, Brazil
| | - Flavia Pedrini
- Surgery Department, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo, Postgraduate Program in Biomaterials and Regenerative Medicine (PPBMR), Laboratory of Biomaterials, Sorocaba, 18030-070, Brazil
- Center of Sciences and Technology for Sustainability, Federal University of São Carlos, Postgraduate Program in Biotechnology and Environmental Monitoring, Sorocaba, 18052-780, Brazil
| | - Felipe Grabarz
- Biotechnology Center, Butantan Institute, Laboratory of Vaccine Development, Sorocaba, 05508-040, Brazil
| | - José Angelo Camilli
- Department of Functional and Structural Biology, University of Campinas, Institute of Biology, Laboratory of Bone Regeneration and Plasticity, São Paulo, 13083-970, Brazil
| | - Eliana Aparecida de Rezende Duek
- Surgery Department, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo, Postgraduate Program in Biomaterials and Regenerative Medicine (PPBMR), Laboratory of Biomaterials, Sorocaba, 18030-070, Brazil
- Center of Sciences and Technology for Sustainability, Federal University of São Carlos, Postgraduate Program in Biotechnology and Environmental Monitoring, Sorocaba, 18052-780, Brazil
| |
Collapse
|
6
|
Brenac C, Fieux M, Giroudon C, Gautherot Tournay N, Henry G, Person H, Ospital C, Mojallal A. Use of autologous adipose tissue in acute burn wound management: A systematic review. ANN CHIR PLAST ESTH 2024; 69:70-78. [PMID: 37770323 DOI: 10.1016/j.anplas.2023.09.006] [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: 08/11/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/30/2023]
Abstract
Fat transfer is increasingly used as part of our reconstructive armamentarium to address the challenges encountered in burn wounds and reconstructive surgery. The present systematic review aimed to evaluate the effectiveness of autologous fat transfer for acute burn wound management. A systematic review of the US National Library of Medicine, Cochrane Library, Web of Science, and Embase was conducted on October 15, 2022 (registration number CDR42022369726). A database watch was performed until submission of the manuscript. The review focused on wound healing. All studies reporting fat transfer in adult patients (at least 5 patients reported) with deep 2nd degree burn wounds were included. The database search yielded a total of 720 records and 367 patients were included from 3 studies. A statistically significant improvement in scar texture, scar appearance, and time to healing was reported in one study in the fat transfer group versus control (P<0.001). Similarly, scores for scar color, scar thickness, scar stiffness, and scar regularity increased significantly. The small number of included studies and their heterogeneity did not allow a meta-regression to be performed. This systematic review emphasizes the limited evidence currently available regarding the use of autologous fat transfer to improve burn wound healing in adult patients, even though it seems promising. Future search should focus on randomized controlled trials with a larger number of participants.
Collapse
Affiliation(s)
- C Brenac
- Department of Plastic, Reconstructive and Aesthetic Surgery, hospices civils de Lyon, Croix-Rousse Hospital, 69004 Lyon, France
| | - M Fieux
- Service d'ORL, d'otoneurochirurgie et de chirurgie cervicofaciale, centre hospitalier Lyon Sud, hospices civils de Lyon, 69310 Pierre-Bénite cedex, France; Université de Lyon, université Lyon 1, 69003 Lyon, France
| | - C Giroudon
- Service de la documentation centrale, hospices civils de Lyon, 69424 Lyon cedex, France
| | - N Gautherot Tournay
- Department of Plastic, Reconstructive and Aesthetic Surgery, hospices civils de Lyon, Croix-Rousse Hospital, 69004 Lyon, France
| | - G Henry
- Department of Plastic, Reconstructive and Aesthetic Surgery, hospices civils de Lyon, Croix-Rousse Hospital, 69004 Lyon, France
| | - H Person
- Service de chirurgie des brûlés, plastique, reconstructrice et esthétique, hospices civils de Lyon, hôpital Édouard-Herriot, 69003 Lyon, France
| | - C Ospital
- Department of Plastic, Reconstructive and Aesthetic Surgery, hospices civils de Lyon, Croix-Rousse Hospital, 69004 Lyon, France
| | - A Mojallal
- Department of Plastic, Reconstructive and Aesthetic Surgery, hospices civils de Lyon, Croix-Rousse Hospital, 69004 Lyon, France; Université de Lyon, université Lyon 1, 69003 Lyon, France; Service de chirurgie des brûlés, plastique, reconstructrice et esthétique, hospices civils de Lyon, hôpital Édouard-Herriot, 69003 Lyon, France.
| |
Collapse
|
7
|
Grun LK, Maurmann RM, Scholl JN, Fogaça ME, Schmitz CRR, Dias CK, Gasparotto J, Padoin AV, Mottin CC, Klamt F, Figueiró F, Jones MH, Filippi-Chiela EC, Guma FCR, Barbé-Tuana FM. Obesity drives adipose-derived stem cells into a senescent and dysfunctional phenotype associated with P38MAPK/NF-KB axis. Immun Ageing 2023; 20:51. [PMID: 37821967 PMCID: PMC10566105 DOI: 10.1186/s12979-023-00378-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Adipose-derived stem cells (ADSC) are multipotent cells implicated in tissue homeostasis. Obesity represents a chronic inflammatory disease associated with metabolic dysfunction and age-related mechanisms, with progressive accumulation of senescent cells and compromised ADSC function. In this study, we aimed to explore mechanisms associated with the inflammatory environment present in obesity in modulating ADSC to a senescent phenotype. We evaluated phenotypic and functional alterations through 18 days of treatment. ADSC were cultivated with a conditioned medium supplemented with a pool of plasma from eutrophic individuals (PE, n = 15) or with obesity (PO, n = 14), and compared to the control. RESULTS Our results showed that PO-treated ADSC exhibited decreased proliferative capacity with G2/M cycle arrest and CDKN1A (p21WAF1/Cip1) up-regulation. We also observed increased senescence-associated β-galactosidase (SA-β-gal) activity, which was positively correlated with TRF1 protein expression. After 18 days, ADSC treated with PO showed augmented CDKN2A (p16INK4A) expression, which was accompanied by a cumulative nuclear enlargement. After 10 days, ADSC treated with PO showed an increase in NF-κB phosphorylation, while PE and PO showed an increase in p38MAPK activation. PE and PO treatment also induced an increase in senescence-associated secretory phenotype (SASP) cytokines IL-6 and IL-8. PO-treated cells exhibited decreased metabolic activity, reduced oxygen consumption related to basal respiration, increased mitochondrial depolarization and biomass, and mitochondrial network remodeling, with no superoxide overproduction. Finally, we observed an accumulation of lipid droplets in PO-treated ADSC, implying an adaptive cellular mechanism induced by the obesogenic stimuli. CONCLUSIONS Taken together, our data suggest that the inflammatory environment observed in obesity induces a senescent phenotype associated with p38MAPK/NF-κB axis, which stimulates and amplifies the SASP and is associated with impaired mitochondrial homeostasis.
Collapse
Affiliation(s)
- L K Grun
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
| | - R M Maurmann
- Graduate Program in Cellular and Molecular Biology, School of Health, Sciences, and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - J N Scholl
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - M E Fogaça
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - C R R Schmitz
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - C K Dias
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - J Gasparotto
- Institute of Biomedical Sciences, Federal University at Alfenas, Alfenas, Brazil
| | - A V Padoin
- Graduate Program in Medicine and Health Sciences, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - C C Mottin
- Graduate Program in Medicine and Health Sciences, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - F Klamt
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - F Figueiró
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - M H Jones
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - E C Filippi-Chiela
- Institute of Basic Health Sciences, Department of Morphological Sciences, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Center for Biotechnology, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - F C R Guma
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - F M Barbé-Tuana
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Graduate Program in Cellular and Molecular Biology, School of Health, Sciences, and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
8
|
Cai F, Chen W, Zhao R, Liu Y. The capacity of exosomes derived from adipose-derived stem cells to enhance wound healing in diabetes. Front Pharmacol 2023; 14:1063458. [PMID: 37808198 PMCID: PMC10551633 DOI: 10.3389/fphar.2023.1063458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
The slow healing and nonhealing of diabetic wounds have long posed challenges for clinical practitioners. In the presence of elevated glucose levels, the body's regulatory mechanisms undergo alterations that impede normal wound healing processes, including cell proliferation, cytokine release, and growth factor activity. Consequently, the advancement of stem cell technology has sparked growing interest in utilizing stem cells and their derivatives as potential therapeutic agents to enhance diabetic wound healing. This paper aims to provide an academic review of the therapeutic effects of adipose-derived stem cell-EXOs (ADSC-EXOs) in diabetic wound healing. As a cell-free therapy, exosomes (EXOs) possess a multitude of proteins and growth factors that have been shown to be advantageous in promoting wound healing and mitigating the potential risks associated with stem cell therapy. By examining the current knowledge on ADSC-EXOs, this review seeks to offer insights and guidance for the potential application of EXOs in the treatment of diabetic wounds.
Collapse
Affiliation(s)
| | | | | | - Yi Liu
- Department of Burns and Plastic Surgery, and Wound Repair Surgery, The Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
9
|
Kun-Varga A, Gubán B, Miklós V, Parvaneh S, Guba M, Szűcs D, Monostori T, Varga J, Varga Á, Rázga Z, Bata-Csörgő Z, Kemény L, Megyeri K, Veréb Z. Herpes Simplex Virus Infection Alters the Immunological Properties of Adipose-Tissue-Derived Mesenchymal-Stem Cells. Int J Mol Sci 2023; 24:11989. [PMID: 37569367 PMCID: PMC10418794 DOI: 10.3390/ijms241511989] [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: 04/17/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 08/13/2023] Open
Abstract
The proper functioning of mesenchymal stem cells (MSCs) is of paramount importance for the homeostasis of the body. Inflammation and infection can alter the function of MSCs, which can also affect the regenerative potential and immunological status of tissues. It is not known whether human herpes simplex viruses 1 and 2 (HSV1 and HSV2), well-known human pathogens that can cause lifelong infections, can induce changes in MSCs. In non-healing ulcers, HSV infection is known to affect deeper tissue layers. In addition, HSV infection can recur after initially successful cell therapies. Our aim was to study the response of adipose-derived MSCs (ADMSCs) to HSV infection in vitro. After confirming the phenotype and differentiation capacity of the isolated cells, we infected the cells in vitro with HSV1-KOS, HSV1-532 and HSV2 virus strains. Twenty-four hours after infection, we examined the gene expression of the cells via RNA-seq and RT-PCR; detected secreted cytokines via protein array; and determined autophagy via Western blot, transmission electron microscopy (TEM) and fluorescence microscopy. Infection with different HSV strains resulted in different gene-expression patterns. In addition to the activation of pathways characteristic of viral infections, distinct non-immunological pathways (autophagy, tissue regeneration and differentiation) were also activated according to analyses with QIAGEN Ingenuity Pathway Analysis, Kyoto Encyclopedia of Genes and Genome and Genome Ontology Enrichment. Viral infections increased autophagy, as confirmed via TEM image analysis, and also increased levels of the microtubule-associated protein light chain 3 (LC3B) II protein. We identified significantly altered accumulation for 16 cytokines involved in tissue regeneration and inflammation. Our studies demonstrated that HSV infection can alter the viability and immunological status of ADMSCs, which may have implications for ADMSC-based cell therapies. Alterations in autophagy can affect numerous processes in MSCs, including the inhibition of tissue regeneration as well as pathological differentiation.
Collapse
Affiliation(s)
- Anikó Kun-Varga
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (A.K.-V.); (B.G.); (M.G.); (D.S.); (T.M.); (L.K.)
- Doctoral School of Clinical Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Barbara Gubán
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (A.K.-V.); (B.G.); (M.G.); (D.S.); (T.M.); (L.K.)
| | - Vanda Miklós
- Biobank, University of Szeged, H-6720 Szeged, Hungary;
| | - Shahram Parvaneh
- HCEMM-SZTE Skin Research Group, University of Szeged, H-6720 Szeged, Hungary; (S.P.); (Z.B.-C.)
| | - Melinda Guba
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (A.K.-V.); (B.G.); (M.G.); (D.S.); (T.M.); (L.K.)
- Interdisciplinary Research Development and Innovation Center of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Diána Szűcs
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (A.K.-V.); (B.G.); (M.G.); (D.S.); (T.M.); (L.K.)
- Interdisciplinary Research Development and Innovation Center of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Tamás Monostori
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (A.K.-V.); (B.G.); (M.G.); (D.S.); (T.M.); (L.K.)
- Interdisciplinary Research Development and Innovation Center of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - János Varga
- Dermatosurgery and Plastic Surgery Unit, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (J.V.); (Á.V.)
| | - Ákos Varga
- Dermatosurgery and Plastic Surgery Unit, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (J.V.); (Á.V.)
| | - Zsolt Rázga
- Department of Pathology, University of Szeged, H-6720 Szeged, Hungary;
| | - Zsuzsanna Bata-Csörgő
- HCEMM-SZTE Skin Research Group, University of Szeged, H-6720 Szeged, Hungary; (S.P.); (Z.B.-C.)
| | - Lajos Kemény
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (A.K.-V.); (B.G.); (M.G.); (D.S.); (T.M.); (L.K.)
- HCEMM-SZTE Skin Research Group, University of Szeged, H-6720 Szeged, Hungary; (S.P.); (Z.B.-C.)
- Interdisciplinary Research Development and Innovation Center of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Klára Megyeri
- Department of Medical Microbiology, University of Szeged, H-6720 Szeged, Hungary;
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary; (A.K.-V.); (B.G.); (M.G.); (D.S.); (T.M.); (L.K.)
- Biobank, University of Szeged, H-6720 Szeged, Hungary;
- Interdisciplinary Research Development and Innovation Center of Excellence, University of Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
10
|
Lazarev S, Uzhviyuk S, Rayev M, Timganova V, Bochkova M, Khaziakhmatova O, Malashchenko V, Litvinova L, Zamorina S. Interaction of Graphene Oxide Nanoparticles with Human Mesenchymal Stem Cells Visualized in the Cell-IQ System. Molecules 2023; 28:molecules28104148. [PMID: 37241889 DOI: 10.3390/molecules28104148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Graphene oxide is a promising nanomaterial with many potential applications. However, before it can be widely used in areas such as drug delivery and medical diagnostics, its influence on various cell populations in the human body must be studied to ensure its safety. We investigated the interaction of graphene oxide (GO) nanoparticles with human mesenchymal stem cells (hMSCs) in the Cell-IQ system, evaluating cell viability, mobility, and growth rate. GO nanoparticles of different sizes coated with linear or branched polyethylene glycol (P or bP, respectively) were used at concentrations of 5 and 25 μg/mL. Designations were the following: P-GOs (Ø 184 ± 73 nm), bP-GOs (Ø 287 ± 52 nm), P-GOb (Ø 569 ± 14 nm), and bP-GOb (Ø 1376 ± 48 nm). After incubating the cells with all types of nanoparticles for 24 h, the internalization of the nanoparticles by the cells was observed. We found that all GO nanoparticles used in this study exerted a cytotoxic effect on hMSCs when used at a high concentration (25 μg/mL), whereas at a low concentration (5 μg/mL) a cytotoxic effect was observed only for bP-GOb particles. We also found that P-GOs particles decreased cell mobility at a concentration of 25 μg/mL, whereas bP-GOb particles increased it. Larger particles (P-GOb and bP-GOb) increased the rate of movement of hMSCs regardless of concentration. There were no statistically significant differences in the growth rate of cells compared with the control group.
Collapse
Affiliation(s)
- Sergey Lazarev
- Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences-Branch of Perm Federal Research Center, 614081 Perm, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State University, 614990 Perm, Russia
| | - Sofya Uzhviyuk
- Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences-Branch of Perm Federal Research Center, 614081 Perm, Russia
| | - Mikhail Rayev
- Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences-Branch of Perm Federal Research Center, 614081 Perm, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State University, 614990 Perm, Russia
| | - Valeria Timganova
- Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences-Branch of Perm Federal Research Center, 614081 Perm, Russia
| | - Maria Bochkova
- Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences-Branch of Perm Federal Research Center, 614081 Perm, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State University, 614990 Perm, Russia
| | - Olga Khaziakhmatova
- Department of Microbiology and Immunology, Faculty of Biology, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Vladimir Malashchenko
- Department of Microbiology and Immunology, Faculty of Biology, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Larisa Litvinova
- Department of Microbiology and Immunology, Faculty of Biology, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Svetlana Zamorina
- Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences-Branch of Perm Federal Research Center, 614081 Perm, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State University, 614990 Perm, Russia
| |
Collapse
|
11
|
Ibrahim HAM, Hussein AM, Gabr M, El-Saeed RA, Ammar OAA, Mosa AAH, Abdel-Aziz AAF. Effect of Melatonin on Alpha Synuclein and Autophagy in Dopaminergic Neuronal Differentiation of Adipose Mesenchymal Stem Cells. Rep Biochem Mol Biol 2023; 12:13-26. [PMID: 37724143 PMCID: PMC10505464 DOI: 10.52547/rbmb.12.1.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/04/2023] [Indexed: 09/20/2023]
Abstract
Background The current work investigated the effect of melatonin on differentiation of adipose mesenchymal stem cells (AD-MSCs) into dopamine producing cells and its effect on autophagy process and alpha-Synuclein (α-Syn) secretion. Methods AD-MSCs were characterized by flow cytometry and divided into 4 groups; i) control group (AD-MSCs without any treatment), ii) M+MSCs group (MSCs treated with 1 µM melatonin for 12 days), iii) DN group (MSCs cultured in neurobasal A medium and essential neuronal growth factors for 12 days) and iv) DN+M group (MSCs cultured in neurobasal A medium and 1µM melatonin for 12 days. By the end of experiments, the dopamine and α-Syn levels using ELISA, the expression of MAP-2, m-TOR and α-Syn genes at the level of mRNA and detection of autophagosomes formation using transmission electron microscope were performed. Results We found that the isolated cells were MSCs due to their positivity expression for CD105 and CD90 and negativity expression for CD34 and CD45. The concentration of dopamine was significantly higher and α-Syn concentration was significantly lower in DN+M group when compared to other groups (P< 0.005). Also, this group showed the highly expression for MAP-2 gene and less expression for m-TOR and α-Syn genes (P< 0.005). Moreover, there was significantly increase in autophagosomes formation in this group than another group (P< 0.005). Conclusions It is concluded that the melatonin promotes the differentiation of rat AD-MSCs into dopaminergic cells via induction of autophagy process and reduction of α-Syn secretion.
Collapse
Affiliation(s)
| | | | - Mahmoud Gabr
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt.
| | - Rasha Aly El-Saeed
- Division of Biochemistry, Chemistry Department, Faculty of Sciences, Mansoura University, Mansoura, Egypt.
| | | | | | | |
Collapse
|
12
|
Exosomes Derived from Adipose Stem Cells Enhance Bone Fracture Healing via the Activation of the Wnt3a/β-Catenin Signaling Pathway in Rats with Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24054852. [PMID: 36902283 PMCID: PMC10003369 DOI: 10.3390/ijms24054852] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Nonunion and delayed union are common complications of diabetes mellitus that pose a serious health threat to people. There are many approaches that have been used to improve bone fracture healing. Recently, exosomes have been regarded as promising medical biomaterials for improving fracture healing. However, whether exosomes derived from adipose stem cells can promote bone fracture healing in diabetes mellitus remains unclear. In this study, adipose stem cells (ASCs) and exosomes derived from adipose stem cells (ASCs-exos) are isolated and identified. Additionally, we evaluate the in vitro and in vivo effects of ASCs-exos on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and bone repair and the regeneration in a rat model of nonunion via Western blotting, immunofluorescence assay, ALP staining, alizarin red staining, radiographic examination and histological analysis. Compared with controls, ASCs-exos promoted BMSC osteogenic differentiation. Additionally, the results of Western blotting, radiographic examination and histological analysis show that ASCs-exos improve the ability for fracture repair in the rat model of nonunion bone fracture healing. Moreover, our results further proved that ASCs-exos play a role in activating the Wnt3a/β-catenin signaling pathway, which facilitates the osteogenic differentiation of BMSCs. All these results show that ASCs-exos enhance the osteogenic potential of BMSCs by activating the Wnt/β-catenin signaling pathway, and also facilitate the ability for bone repair and regeneration in vivo, which provides a novel direction for fracture nonunion in diabetes mellitus treatment.
Collapse
|
13
|
Wang T, Li T, Niu X, Hu L, Cheng J, Guo D, Ren H, Zhao R, Ji Z, Liu P, Li Y, Guo Y. ADSC-derived exosomes attenuate myocardial infarction injury by promoting miR-205-mediated cardiac angiogenesis. Biol Direct 2023; 18:6. [PMID: 36849959 PMCID: PMC9972746 DOI: 10.1186/s13062-023-00361-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/21/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Acute myocardial infarction is a major health problem and is the leading cause of death worldwide. Myocardial apoptosis induced by myocardial infarction injury is involved in the pathophysiology of heart failure. Therapeutic stem cell therapy has the potential to be an effective and favorable treatment for ischemic heart disease. Exosomes derived from stem cells have been shown to effectively repair MI injury-induced cardiomyocyte damage. However, the cardioprotective benefits of adipose tissue-derived mesenchymal stem cell (ADSC)-Exos remain unknown. This study aimed to investigate the protective effects of exosomes from ADSC on the hearts of MI-treated mice and to explore the underlying mechanisms. METHODS Cellular and molecular mechanisms were investigated using cultured ADSCs. On C57BL/6J mice, we performed myocardial MI or sham operations and assessed cardiac function, fibrosis, and angiogenesis 4 weeks later. Mice were intramyocardially injected with ADSC-Exos or vehicle-treated ADSCs after 25 min following the MI operation. RESULTS Echocardiographic experiments showed that ADSC-Exos could significantly improve left ventricular ejection fraction, whereas ADSC-Exos administration could significantly alleviate MI-induced cardiac fibrosis. Additionally, ADSC-Exos treatment has been shown to reduce cardiomyocyte apoptosis while increasing angiogenesis. Molecular experiments found that exosomes extracted from ADSCs can promote the proliferation and migration of microvascular endothelial cells, facilitate angiogenesis, and inhibit cardiomyocytes apoptosis through miRNA-205. We then transferred isolated exosomes from ADSCs into MI-induced mice and observed decreased cardiac fibrosis, increased angiogenesis, and improved cardiac function. We also observed increased apoptosis and decreased expression of hypoxia-inducible factor-1α and vascular endothelial growth factor in HMEC-1 transfected with a miRNA-205 inhibitor. CONCLUSION In summary, these findings show that ADSC-Exos can alleviate cardiac injury and promote cardiac function recovery in MI-treated mice via the miRNA-205 signaling pathway. ADSC-Exos containing miRNA205 have a promising therapeutic potential in MI-induced cardiac injury.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Tao Li
- Ultrasound Diagnostic and Treatment Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xiaolin Niu
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jin Cheng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - He Ren
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Ran Zhao
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhaole Ji
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Pengyun Liu
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Yanjie Guo
- Heart Hospital, Xi'an International Medical Center, Xi'an, 710038, China. .,Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
14
|
Adipose-Derived Mesenchymal Stromal Cells in Basic Research and Clinical Applications. Int J Mol Sci 2023; 24:ijms24043888. [PMID: 36835295 PMCID: PMC9962639 DOI: 10.3390/ijms24043888] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/09/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023] Open
Abstract
Adipose-derived mesenchymal stromal cells (AD-MSCs) have been extensively studied in recent years. Their attractiveness is due to the ease of obtaining clinical material (fat tissue, lipoaspirate) and the relatively large number of AD-MSCs present in adipose tissue. In addition, AD-MSCs possess a high regenerative potential and immunomodulatory activities. Therefore, AD-MSCs have great potential in stem cell-based therapies in wound healing as well as in orthopedic, cardiovascular, or autoimmune diseases. There are many ongoing clinical trials on AD-MSC and in many cases their effectiveness has been proven. In this article, we present current knowledge about AD-MSCs based on our experience and other authors. We also demonstrate the application of AD-MSCs in selected pre-clinical models and clinical studies. Adipose-derived stromal cells can also be the pillar of the next generation of stem cells that will be chemically or genetically modified. Despite much research on these cells, there are still important and interesting areas to explore.
Collapse
|
15
|
Hybrid Stromal Vascular Fraction (Hybrid-SVF): A New Paradigm in Mechanical Regenerative Cell Processing. Plast Reconstr Surg Glob Open 2022; 10:e4702. [PMID: 36601591 PMCID: PMC9803457 DOI: 10.1097/gox.0000000000004702] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 10/11/2022] [Indexed: 01/06/2023]
Abstract
Enzymatic digestion of extracellular matrix (ECM) from lipoaspirate is the conventional form of harvesting stromal vascular fraction (SVF) called enzymatically digested SVF (E-SVF). Mechanical SVF (M-SVF) isolation has emerged as an alternative method, but it has also some limitations in terms of lower cell viability and diminished cell counts. To enhance the SVF qualitatively and quantitatively, we propose a novel concept called "hybrid-SVF," in which we combine M-SVF with the concentrated parts of adipose tissue after centrifugation, which is called stromal vascular matrix (SVM). Methods Hybrid-SVF injection was applied as an adjunctive therapy to fat grafting in 88 patients and 11 samples were evaluated in the laboratory for cell count, viability and cell activity. Results Experimental results determined that SVM part showed higher cellular activity. SVM and M-SVF showed higher cellular potency than E-SVF. Clinically, none of the patients required an additional session for fat grafting since there was no significant graft resorption. However, seven patients asked for further volume augmentation due to their individual preferences. No major complication was encountered. Conclusions The usage of hybrid-SVF has a very high regenerative potential due to the ECM support and exceptionally high cell yield in addition to preserved cell potency. Although there are ongoing studies focusing on optimizing cell counts and further clinical applications, we believe that our preliminary results might create a paradigm shift in the area of regenerative fat grafting.
Collapse
|
16
|
Liang Y, Yang C, Ye F, Cheng Z, Li W, Hu Y, Hu J, Zou L, Jiang H. Repair of the Urethral Mucosa Defect Model Using Adipose-Derived Stem Cell Sheets and Monitoring the Fate of Indocyanine Green-Labeled Sheets by Near Infrared-II. ACS Biomater Sci Eng 2022; 8:4909-4920. [PMID: 36201040 DOI: 10.1021/acsbiomaterials.2c00695] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Treatment of urethral mucosa defects is a major challenge in urology. Synthetic materials or autologous mucosa does not provide satisfactory treatment options for long-term or large urethral mucosa defects. In response to this problem, we used autologous adipose-derived stem cells (ADSCs) to synthesize cell sheets in vitro for repairing urethral mucosa defect models. In order to monitor the localization and distribution of cell sheets in vivo, cells and sheets were labeled with indocyanine green (ICG) and the second near-infrared (NIR-II) fluorescence imaging was performed. ICG-based NIR-II imaging can successfully track ADSCs and sheets in vivo up to 8 W. Then, rabbit urethral mucosa defect models were repaired with ICG-ADSCs sheets. At 3 months after operation, retrograde urethrography showed that ADSC sheets could effectively repair urethral mucosa defect and restore urethral patency. Histological analysis showed that in ADSC sheet groups, continuous epithelial cells covered the urethra at the transplantation site, and a large number of vascular endothelial cells could also be seen. In the cell-free sheet group, there was no continuous epithelial cell coverage at the repair site of the urethra, and the expression of pro-inflammatory factor TNF-α was increased. It shows that the extracellular matrix alone without cells is not suitable for repairing urethral defects. Surviving ADSCs in the sheets may play a key role in the repair process. This study provides a new tracing method for tissue engineering to dynamically track grafts using an NIR-II imaging system. The ADSC sheets can effectively restore the structure and function of the urethra. It provides a new option for the repair of urethral mucosa defects.
Collapse
Affiliation(s)
- Yingchun Liang
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, 200040 Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Chen Yang
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, 200040 Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Fangdie Ye
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, 200040 Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Zhang Cheng
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, 200040 Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Weijian Li
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, 200040 Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Yun Hu
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, 200040 Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Jimeng Hu
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, 200040 Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Lujia Zou
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, 200040 Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, 200040 Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, 200040 Shanghai, China.,National Clinical Research Center for Aging and Medicine, Fudan University, 200040 Shanghai, China
| |
Collapse
|
17
|
Wu B, Feng J, Guo J, Wang J, Xiu G, Xu J, Ning K, Ling B, Fu Q, Xu J. ADSCs-derived exosomes ameliorate hepatic fibrosis by suppressing stellate cell activation and remodeling hepatocellular glutamine synthetase-mediated glutamine and ammonia homeostasis. Stem Cell Res Ther 2022; 13:494. [PMID: 36195966 PMCID: PMC9531400 DOI: 10.1186/s13287-022-03049-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/17/2022] [Indexed: 11/18/2022] Open
Abstract
Background Hepatic fibrosis is a common pathologic stage in chronic liver disease development, which might ultimately lead to liver cirrhosis. Accumulating evidence suggests that adipose-derived stromal cells (ADSCs)-based therapies show excellent therapeutic potential in liver injury disease owing to its superior properties, including tissue repair ability and immunomodulation effect. However, cell-based therapy still limits to several problems, such as engraftment efficiency and immunoreaction, which impede the ADSCs-based therapeutics development. So, ADSCs-derived extracellular vesicles (EVs), especially for exosomes (ADSC-EXO), emerge as a promise cell-free therapeutics to ameliorate liver fibrosis. The effect and underlying mechanisms of ADSC-EXO in liver fibrosis remains blurred. Methods Hepatic fibrosis murine model was established by intraperitoneal sequential injecting the diethylnitrosamine (DEN) for two weeks and then carbon tetrachloride (CCl4) for six weeks. Subsequently, hepatic fibrosis mice were administrated with ADSC-EXO (10 μg/g) or PBS through tail vein infusion for three times in two weeks. To evaluate the anti-fibrotic capacity of ADSC-EXO, we detected liver morphology by histopathological examination, ECM deposition by serology test and Sirius Red staining, profibrogenic markers by qRT-PCR assay. LX-2 cells treated with TGF-β (10 ng/ml) for 12 h were conducted for evaluating ADSC-EXO effect on activated hepatic stellate cells (HSCs). RNA-seq was performed for further analysis of the underlying regulatory mechanisms of ADSC-EXO in liver fibrosis. Results In this study, we obtained isolated ADSCs, collected and separated ADSCs-derived exosomes. We found that ADSC-EXO treatment could efficiently ameliorate DEN/CCl4-induced hepatic fibrosis by improving mice liver function and lessening hepatic ECM deposition. Moreover, ADSC-EXO intervention could reverse profibrogenic phenotypes both in vivo and in vitro, including HSCs activation depressed and profibrogenic markers inhibition. Additionally, RNA-seq analysis further determined that decreased glutamine synthetase (Glul) of perivenous hepatocytes in hepatic fibrosis mice could be dramatically up-regulated by ADSC-EXO treatment; meanwhile, glutamine and ammonia metabolism-associated key enzyme OAT was up-regulated and GLS2 was down-regulated by ADSC-EXO treatment in mice liver. In addition, glutamine synthetase inhibitor would erase ADSC-EXO therapeutic effect on hepatic fibrosis. Conclusions These findings demonstrated that ADSC-derived exosomes could efficiently alleviate hepatic fibrosis by suppressing HSCs activation and remodeling glutamine and ammonia metabolism mediated by hepatocellular glutamine synthetase, which might be a novel and promising anti-fibrotic therapeutics for hepatic fibrosis disease. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03049-x.
Collapse
Affiliation(s)
- Baitong Wu
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, People's Republic of China
| | - Jiuxing Feng
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Jingyi Guo
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, People's Republic of China
| | - Jian Wang
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, People's Republic of China
| | - Guanghui Xiu
- Department of Intensive Care Unit, Affiliated Hospital of Yunnan University (The Second People's Hospital of Yunnan Province), Yunnan University, Kunming, People's Republic of China
| | - Jiaqi Xu
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, People's Republic of China
| | - Ke Ning
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Bin Ling
- Department of Intensive Care Unit, Affiliated Hospital of Yunnan University (The Second People's Hospital of Yunnan Province), Yunnan University, Kunming, People's Republic of China.
| | - Qingchun Fu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China.
| | - Jun Xu
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, People's Republic of China.
| |
Collapse
|
18
|
Liu Q, Zhang P, Yuan X, Ya O, Li Q, Li J, Long Q. Investigate the stemness of adult adipose-derived stromal cells based on single-cell RNA-sequencing. Cell Biol Int 2022; 46:2118-2131. [PMID: 36150081 DOI: 10.1002/cbin.11898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/15/2022] [Accepted: 08/20/2022] [Indexed: 11/06/2022]
Abstract
The cellular heterogeneity and genetic features of stemness of adipose-derived stromal cells (ADSCs) remain unclear. Using single-cell RNA sequencing (scRNA-seq), we investigated the genomic features of the stemness gene in ADSCs with genetic variability. We cultured the ADSCs isolated from the fat waste of a healthy adult volunteers undergoing cosmetic plastic surgery to the third generation, used the BD Rhapsody platform to perform scRNA-seq, then used Monocle2 to analyze the growth and development trajectory of ADSCs, Cellular Trajectory Reconstruction Analysis Using Gene Counts and Expression (CytoTRACE) to evaluate the stemness gene characteristics in ADSCs clusters, and Beam to analyze the expression change characteristics of the main stemness related genes of ADSCs. According to the scRNA-seq data of 5325 ADSCs, they could be classified into nine cell clusters. According to CytoTRACE analysis, Cluster 3 of ADSCs had the highest stemness, whereas Cluster 8 had the lowest stemness. Pseudotime analysis revealed that Cluster 3 of ADSCs was primarily dispersed in the middle part of the growth and development trajectory, whereas Cluster 8 was primarily distributed at the end. We summarized the stemness of Cluster 3 in ADSCs with high expression of TPM1 and CCND1 genes in the metaphase of growth and development is the strongest, whereas the stemness of Cluster 8 with high expression of FICD, CREBRF, SDF2L1, HERPUD1, and HYOU1 genes in the telophase of growth and development is the weakest, providing a theoretical basis for screening and improving the therapeutic effect of ADSCs in cell transplantation research.
Collapse
Affiliation(s)
- Qing Liu
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan
| | - Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan
| | - Ou Ya
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan
| | - Qi Li
- Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, China
| | - Jing Li
- Radiology Department, Tangshan Maternal and Child Health Hospital, Tangshan, China
| | - Qingxi Long
- Department of Neurology, Kailuan General Hospital, Affiliated North China University of Science and Technology, Tangshan
| |
Collapse
|
19
|
Hendawy H, Metwally E, Elfadadny A, Yoshida T, Ma D, Shimada K, Hamabe L, Sasaki K, Tanaka R. Cultured versus freshly isolated adipose-derived stem cells in improvement of the histopathological outcomes in HCL-induced cystitis in a rat model. Biomed Pharmacother 2022; 153:113422. [DOI: 10.1016/j.biopha.2022.113422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/02/2022] Open
|
20
|
Lebeau G, Ah-Pine F, Daniel M, Bedoui Y, Vagner D, Frumence E, Gasque P. Perivascular Mesenchymal Stem/Stromal Cells, an Immune Privileged Niche for Viruses? Int J Mol Sci 2022; 23:ijms23148038. [PMID: 35887383 PMCID: PMC9317325 DOI: 10.3390/ijms23148038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) play a critical role in response to stress such as infection. They initiate the removal of cell debris, exert major immunoregulatory activities, control pathogens, and lead to a remodeling/scarring phase. Thus, host-derived ‘danger’ factors released from damaged/infected cells (called alarmins, e.g., HMGB1, ATP, DNA) as well as pathogen-associated molecular patterns (LPS, single strand RNA) can activate MSCs located in the parenchyma and around vessels to upregulate the expression of growth factors and chemoattractant molecules that influence immune cell recruitment and stem cell mobilization. MSC, in an ultimate contribution to tissue repair, may also directly trans- or de-differentiate into specific cellular phenotypes such as osteoblasts, chondrocytes, lipofibroblasts, myofibroblasts, Schwann cells, and they may somehow recapitulate their neural crest embryonic origin. Failure to terminate such repair processes induces pathological scarring, termed fibrosis, or vascular calcification. Interestingly, many viruses and particularly those associated to chronic infection and inflammation may hijack and polarize MSC’s immune regulatory activities. Several reports argue that MSC may constitute immune privileged sanctuaries for viruses and contributing to long-lasting effects posing infectious challenges, such as viruses rebounding in immunocompromised patients or following regenerative medicine therapies using MSC. We will herein review the capacity of several viruses not only to infect but also to polarize directly or indirectly the functions of MSC (immunoregulation, differentiation potential, and tissue repair) in clinical settings.
Collapse
Affiliation(s)
- Grégorie Lebeau
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Franck Ah-Pine
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Service Anatomo-Pathologie, CHU de la Réunion, 97400 Saint-Denis, France
| | - Matthieu Daniel
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Yosra Bedoui
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Damien Vagner
- Service de Médecine Interne, CHU de la Réunion, 97400 Saint-Denis, France;
| | - Etienne Frumence
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Philippe Gasque
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
- Correspondence:
| |
Collapse
|
21
|
Huang G, Xia B, Dai Z, Yang R, Chen R, Yang H. Comparative study of DFAT cell and ADSC sheets for periodontal tissue regeneration:
in vivo
and
in vitro
evidence. J Clin Periodontol 2022; 49:1289-1303. [PMID: 35851962 DOI: 10.1111/jcpe.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/29/2022] [Accepted: 06/30/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Guobin Huang
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| | - Bin Xia
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Oral and Maxillofacial Surgery The Affiliated Stomatological Hospital of Kunming Medical University Kunming Yunnan P.R. China
| | - Zichao Dai
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| | - Rongqiang Yang
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| | - Rui Chen
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| | - Hefeng Yang
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| |
Collapse
|
22
|
Schepici G, Gugliandolo A, Mazzon E. Serum-Free Cultures: Could They Be a Future Direction to Improve Neuronal Differentiation of Mesenchymal Stromal Cells? Int J Mol Sci 2022; 23:ijms23126391. [PMID: 35742836 PMCID: PMC9223839 DOI: 10.3390/ijms23126391] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are undifferentiated cells with multilinear potential, known for their immunomodulatory and regenerative properties. Although the scientific community is working to improve their application, concerns limit their use to repair tissues following neurological damage. One of these obstacles is represented by the use of culture media supplemented with fetal bovine serum (FBS), which, due to its xenogenic nature and the risk of contamination, has increased scientific, ethical and safety problems. Therefore, the use of serum-free media could improve MSC culture methods, avoiding infectious and immunogenic transmission problems as well as MSC bioprocesses, without the use of animal components. The purpose of our review is to provide an overview of experimental studies that demonstrate that serum-free cultures, along with the supplementation of growth factors or chemicals, can lead to a more defined and controlled environment, enhancing the proliferation and neuronal differentiation of MSCs.
Collapse
|
23
|
Wang S, Qiu Y, Qu L, Wang Q, Zhou Q. Hydrogels for Treatment of Different Degrees of Osteoarthritis. Front Bioeng Biotechnol 2022; 10:858656. [PMID: 35733529 PMCID: PMC9207401 DOI: 10.3389/fbioe.2022.858656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/18/2022] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) is a common disease that severely restricts human activities and degrades the quality of life. Every year, millions of people worldwide are diagnosed with osteoarthritis, placing a heavy burden on society. Hydrogels, a polymeric material with good biocompatibility and biodegradability, are a novel approach for the treatment of osteoarthritis. In recent years, this approach has been widely studied with the development of materials science and tissue engineering technology. We reviewed the research progress of hydrogels in the treatment of osteoarthritis in the past 3 years. We summarized the required hydrogel properties and current applications according to the development and treatment of osteoarthritis. Furthermore, we listed the challenges of hydrogels for different types of osteoarthritis and presented prospects for future development.
Collapse
Affiliation(s)
- Shuze Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yueyang Qiu
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Liu Qu
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Qing Zhou
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
- *Correspondence: Qing Zhou,
| |
Collapse
|
24
|
Xiao J, Zeng L, Ding S, Chen Y, Zhang X, Bian X, Tian G. Tumor-Tropic Adipose-Derived Mesenchymal Stromal Cell Mediated Bi 2 Se 3 Nano-Radiosensitizers Delivery for Targeted Radiotherapy of Non-Small Cell Lung Cancer. Adv Healthc Mater 2022; 11:e2200143. [PMID: 35195958 DOI: 10.1002/adhm.202200143] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/15/2022] [Indexed: 11/05/2022]
Abstract
With the successful marriage between nanotechnology and oncology, various high-Z element containing nanoparticles (NPs) are approved as radiosensitizers to overcome radiation resistance for enhanced radiotherapy (RT). Unfortunately, NPs themselves lack specificity to tumors. Due to the inherent tropism nature of malignant cells, mesenchymal stem cells (MSCs) emerge as cell-mediated delivery vehicles for functional NPs to improve their therapeutic index. Herein, radiosensitive bismuth selenide (Bi2 Se3 ) NPs-laden adipose-derived mesenchymal stromal cells (AD-MSCs/Bi2 Se3 ) are engineered for targeted RT of non-small cell lung cancer (NSCLC). The results reveal that the optimized intracellular loading strategy hardly affects cell viability, specific surface markers, or migration capability of AD-MSCs, and Bi2 Se3 NPs can be efficiently transported from AD-MSCs to tumor cells. In vivo biodistribution test shows that the Bi2 Se3 NPs accumulation in tumor is increased 20 times via AD-MSCs-mediated delivery. Therefore, AD-MSCs/Bi2 Se3 administration synchronized with X-ray irradiation controls the tumor progress well in orthotopic A549 tumor bearing mice. Considering that MSCs migrate better to irradiated tumor cells in comparison to nonirradiated ones and MSCs preferentially accumulate within lung tissues after systemic administration into accounts, the tumor-tropic MSCs/NPs system is feasible and promising for targeted RT treatment of NSCLC.
Collapse
Affiliation(s)
- Jingfang Xiao
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 40038 P. R. China
| | - Lijuan Zeng
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 40038 P. R. China
| | - Shuaishuai Ding
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 40038 P. R. China
| | - Yemiao Chen
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 40038 P. R. China
- Biobank of The First Affiliated Hospital Third Military Medical University (Army Medical University) Chongqing 40038 P. R. China
- Clinical Research Center Chongqing Public Health Medical Center Chongqing 400036 P. R. China
| | - Xiao Zhang
- International Joint Research Center for Precision Biotherapy and Department of Stem Cell and Regenerative Medicine The First Affiliated Hospital Third Military Medical University (Army Medical University) Chongqing 400038 P. R. China
| | - Xiu‐wu Bian
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 40038 P. R. China
| | - Gan Tian
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology Ministry of Education of China Chongqing 40038 P. R. China
| |
Collapse
|
25
|
Smakaj A, De Mauro D, Rovere G, Pietramala S, Maccauro G, Parolini O, Lattanzi W, Liuzza F. Clinical Application of Adipose Derived Stem Cells for the Treatment of Aseptic Non-Unions: Current Stage and Future Perspectives-Systematic Review. Int J Mol Sci 2022; 23:ijms23063057. [PMID: 35328476 PMCID: PMC8950719 DOI: 10.3390/ijms23063057] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 12/18/2022] Open
Abstract
Fracture non-union is a challenging orthopaedic issue and a socio-economic global burden. Several biological therapies have been introduced to improve traditional surgical approaches. Among these, the latest research has been focusing on adipose tissue as a powerful source of mesenchymal stromal cells, namely, adipose-derived stem cells (ADSCs). ADSC are commonly isolated from the stromal vascular fraction (SVF) of liposuctioned hypodermal adipose tissue, and their applications have been widely investigated in many fields, including non-union fractures among musculoskeletal disorders. This review aims at providing a comprehensive update of the literature on clinical application of ADSCs for the treatment of non-unions in humans. The study was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). Only three articles met our inclusion criteria, with a total of 12 cases analyzed for demographics and harvesting, potential manufacturing and implantation of ADSCs. The review of the literature suggests that adipose derived cell therapy can represent a promising alternative in bone regenerative medicine for the enhancement of non-unions and bone defects. The low number of manuscripts reporting ADSC-based therapies for long bone fracture healing suggests some critical issues that are discussed in this review. Nevertheless, further investigations on human ADSC therapies are needed to improve the knowledge on their translational potential and to possibly achieve a consensus on their use for such applications.
Collapse
Affiliation(s)
- Amarildo Smakaj
- Department of Aging, Neurological, Orthopaedic and Head-Neck Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (A.S.); (D.D.M.); (G.R.); (S.P.); (G.M.)
- Department of Geriatrics and Orthopaedic Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Domenico De Mauro
- Department of Aging, Neurological, Orthopaedic and Head-Neck Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (A.S.); (D.D.M.); (G.R.); (S.P.); (G.M.)
- Department of Geriatrics and Orthopaedic Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giuseppe Rovere
- Department of Aging, Neurological, Orthopaedic and Head-Neck Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (A.S.); (D.D.M.); (G.R.); (S.P.); (G.M.)
- Department of Geriatrics and Orthopaedic Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Silvia Pietramala
- Department of Aging, Neurological, Orthopaedic and Head-Neck Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (A.S.); (D.D.M.); (G.R.); (S.P.); (G.M.)
- Department of Geriatrics and Orthopaedic Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giulio Maccauro
- Department of Aging, Neurological, Orthopaedic and Head-Neck Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (A.S.); (D.D.M.); (G.R.); (S.P.); (G.M.)
- Department of Geriatrics and Orthopaedic Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Fondazione Policlinico A. Gemelli IRCCS, 00168 Rome, Italy
| | - Wanda Lattanzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Fondazione Policlinico A. Gemelli IRCCS, 00168 Rome, Italy
- Correspondence: (W.L.); (F.L.); Tel.: +39-06-30154464 (W.L.); +39-339-719-1314 (F.L.)
| | - Francesco Liuzza
- Department of Aging, Neurological, Orthopaedic and Head-Neck Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (A.S.); (D.D.M.); (G.R.); (S.P.); (G.M.)
- Correspondence: (W.L.); (F.L.); Tel.: +39-06-30154464 (W.L.); +39-339-719-1314 (F.L.)
| |
Collapse
|
26
|
Ying L, Liang C, Zhang Y, Wang J, Wang C, Xia K, Shi K, Yu C, Yang B, Xu H, Zhang Y, Shu J, Huang X, Xing H, Li F, Zhou X, Chen Q. Enhancement of nucleus pulposus repair by glycoengineered adipose-derived mesenchymal cells. Biomaterials 2022; 283:121463. [DOI: 10.1016/j.biomaterials.2022.121463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 12/28/2022]
|
27
|
Liao S, Meng H, Zhao J, Lin W, Liu X, Tian Z, Lan L, Yang H, Zou Y, Xu Y, Gao X, Lu S, Peng J. Injectable adipose-derived stem cells-embedded alginate-gelatin microspheres prepared by electrospray for cartilage tissue regeneration. J Orthop Translat 2022; 33:174-185. [PMID: 35495963 PMCID: PMC9018217 DOI: 10.1016/j.jot.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 11/25/2022] Open
Abstract
Objective To prepare adipose-derived stem cells (ADSCs)-embedded alginate-gelatinemicrospheres (Alg-Gel-ADSCs MSs) by electrospray and evaluate their feasibility for cartilage tissue engineering. To observe the efficacy of Alg-Gel-ADSCs MSs in repairing articular cartilage defects in SD rats. Methods ADSCs were isolated and characterized by performing induced differentiation and flow cytometry assays. Alginate-gelatine microspheres with different gelatine concentrations were manufactured by electrospraying, and the appropriate alginate-gelatine concentration and ratio were determined by evaluating microsphere formation. Alg-Gel-ADSCs MSs were compared with Alg-ADSCs MSs through the induction of chondrogenic differentiation and culture. Their feasibility for cartilage tissue engineering was analysed by performing Live/Dead staining, cell proliferation analysis, toluidine blue staining and a glycosaminoglycan (GAG) content analysis. Alg-Gel-ADSCs MSs were implanted in the cartilage defects of SD rats, and the cartilage repair effect was evaluated at different time points. The evaluation included gross observations and histological evaluations, fluorescence imaging tracking, immunohistochemical staining, microcomputed tomography (micro-CT) and a CatWalk evaluation. Results The isolated ADSCs showed multidirectional differentiation and were used for cartilage tissue engineering. Using 1.5 w:v% alginate and 0.5 w:v% gelatine (Type B), we successfully prepared nearly spherical microspheres. Compared with alginate microspheres, alginate gel increased the viability of ADSCs and promoted the proliferation and chondrogenesis of ADSCs. In our experiments on knee cartilage defects in SD rats in vivo, the Alg-Gel-ADSCs MSs showed superior cartilage repair in cell resides, histology evaluation, micro-CT imaging and gait analysis. Conclusions Microspheres composed of 1.5 w:v% alginate-0.5 w:v% gelatine increase the viability of ADSCs and supported their proliferation and deposition of cartilage matrix components. ADSCs embedded in 1.5 w:v% alginate-0.5 w:v% gelatine microspheres show superior repair efficacy and prospective applications in cartilage tissue repair. The translational potential of this article In this study, injectable adipose-derived stem cells-embedded alginate-gelatin microspheres (Alg-Gel-ADSCs MSs) were prepared by the electrospray . Compared with the traditional alginate microspheres, its support ability for ADSCs is better and shows a better repair effect. This study provides a promising strategy for cartilage tissue regeneration.
Collapse
|
28
|
Litvinova LS, Shupletsova VV, Khaziakhmatova OG, Daminova AG, Kudryavtseva VL, Yurova KA, Malashchenko VV, Todosenko NM, Popova V, Litvinov RI, Korotkova EI, Sukhorukov GB, Gow AJ, Weissman D, Atochina-Vasserman EN, Khlusov IA. Human Mesenchymal Stem Cells as a Carrier for a Cell-Mediated Drug Delivery. Front Bioeng Biotechnol 2022; 10:796111. [PMID: 35284410 PMCID: PMC8909129 DOI: 10.3389/fbioe.2022.796111] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
A number of preclinical and clinical studies have demonstrated the efficiency of mesenchymal stromal cells to serve as an excellent base for a cell-mediated drug delivery system. Cell-based targeted drug delivery has received much attention as a system to facilitate the uptake a nd transfer of active substances to specific organs and tissues with high efficiency. Human mesenchymal stem cells (MSCs) are attracting increased interest as a promising tool for cell-based therapy due to their high proliferative capacity, multi-potency, and anti-inflammatory and immunomodulatory properties. In particular, these cells are potentially suitable for use as encapsulated drug transporters to sites of inflammation. Here, we studied the in vitro effects of incorporating synthetic polymer microcapsules at various microcapsule-to-cell ratios on the morphology, ultrastructure, cytokine profile, and migration ability of human adipose-derived MSCs at various time points post-phagocytosis. The data show that under appropriate conditions, human MSCs can be efficiently loaded with synthesized microcapsules without damaging the cell’s structural integrity with unexpressed cytokine secretion, retained motility, and ability to migrate through 8 μm pores. Thus, the strategy of using human MSCs as a delivery vehicle for transferring microcapsules, containing bioactive material, across the tissue–blood or tumor–blood barriers to facilitate the treatment of stroke, cancer, or inflammatory diseases may open a new therapeutic perspective.
Collapse
Affiliation(s)
- L. S. Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - V. V. Shupletsova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - O. G. Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - A. G. Daminova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Kazan Institute of Biochemistry and Biophysics, FRC KSC of RAS, Kazan, Russia
- Interdisciplinary Center for Analytical Microscopy, Kazan Federal University, Kazan, Russia
| | - V. L. Kudryavtseva
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - K. A. Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - V. V. Malashchenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - N. M. Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - V. Popova
- School of Earth Sciences and Engineering, National Research Tomsk Polytechnic University, Tomsk, Russia
| | - R. I. Litvinov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - E. I. Korotkova
- School of Earth Sciences and Engineering, National Research Tomsk Polytechnic University, Tomsk, Russia
| | - G. B. Sukhorukov
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - A. J. Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - D. Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - E. N. Atochina-Vasserman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: E. N. Atochina-Vasserman,
| | - I. A. Khlusov
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
- Department of Morphology and General Pathology, Siberian State Medical University, Tomsk, Russia
- Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, Russia
| |
Collapse
|
29
|
Mesenchymal Stromal Cells Preconditioning: A New Strategy to Improve Neuroprotective Properties. Int J Mol Sci 2022; 23:ijms23042088. [PMID: 35216215 PMCID: PMC8878691 DOI: 10.3390/ijms23042088] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Neurological diseases represent one of the main causes of disability in human life. Consequently, investigating new strategies capable of improving the quality of life in neurological patients is necessary. For decades, researchers have been working to improve the efficacy and safety of mesenchymal stromal cells (MSCs) therapy based on MSCs’ regenerative and immunomodulatory properties and multilinear differentiation potential. Therefore, strategies such as MSCs preconditioning are useful to improve their application to restore damaged neuronal circuits following neurological insults. This review is focused on preconditioning MSCs therapy as a potential application to major neurological diseases. The aim of our work is to summarize both the in vitro and in vivo studies that demonstrate the efficacy of MSC preconditioning on neuronal regeneration and cell survival as a possible application to neurological damage.
Collapse
|
30
|
Mouse Neural Stem Cell Differentiation and Human Adipose Mesenchymal Stem Cell Transdifferentiation Into Neuron- and Oligodendrocyte-like Cells With Myelination Potential. Stem Cell Rev Rep 2021; 18:732-751. [PMID: 34780018 DOI: 10.1007/s12015-021-10218-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 01/09/2023]
Abstract
Stem cell therapy is an interesting approach for neural repair, once it can improve and increase processes, like angiogenesis, neurogenesis, and synaptic plasticity. In this regard, adult neural stem cells (NSC) are studied for their mechanisms of proliferation, differentiation and functionality in neural repair. Here, we describe novel neural differentiation methods. NSC from adult mouse brains and human adipose-derived stem cells (hADSC) were isolated and characterized regarding their neural differentiation potential based on neural marker expression profiles. For both cell types, their capabilities of differentiating into neuron-, astrocyte- and oligodendrocytes-like cells (NLC, ALC and OLC, respectively) were analyzed. Our methodologies were capable of producing NLC, ALC and OLC from adult murine and human transdifferentiated NSC. NSC showed augmented gene expression of NES, TUJ1, GFAP and PDGFRA/Cnp. Following differentiation induction into NLC, OLC or ALC, specific neural phenotypes were obtained expressing MAP2, GalC/O4 or GFAP with compatible morphologies, respectively. Accordingly, immunostaining for nestin+ in NSC, GFAP+ in astrocytes and GalC/O4+ in oligodendrocytes was detected. Co-cultured NLC and OLC showed excitability in 81.3% of cells and 23.5% of neuron/oligodendrocyte marker expression overlap indicating occurrence of in vitro myelination. We show here that hADSC can be transdifferentiated into NSC and distinct neural phenotypes with the occurrence of neuron myelination in vitro, providing novel strategies for CNS regeneration therapy. Superior Part: Schematic organization of obtaining and generating hNSC from hADSC and differentiation processes and phenotypic expression of neuron, astrocyte and oligodendrocyte markers (MAP2, GFAP and O4, respectively) and stem cell marker (NES) of differentiating hNSC 14 days after induction. The nuclear staining in blue corresponds to DAPI. bar = 100 μm. Inferior part: Neural phenotype fates in diverse differentiation media. NES: nestin; GFAP: Glial fibrillary acidic protein. MAP2: Microtubule-associated protein 2. TUJ1: β-III tubulin. PDGFRA: PDGF receptor alpha. Two-way ANOVA with Bonferroni post-test with n = 3. * p < 0.05 and ** p < 0.01: (NSCiM1 NSC induction medium 1) vs differentiation media.
Collapse
|
31
|
Bian Y, Yu H, Jin M, Gao X. Repigmentation by combined narrow‑band ultraviolet B/adipose‑derived stem cell transplantation in the mouse model: Role of Nrf2/HO‑1‑mediated Ca 2+ homeostasis. Mol Med Rep 2021; 25:6. [PMID: 34751412 PMCID: PMC8600419 DOI: 10.3892/mmr.2021.12522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 07/16/2021] [Indexed: 11/06/2022] Open
Abstract
Vitiligo is a depigmentation disease commonly seen in clinical practice, mainly involving loss of functional epidermal pigment cells and hair follicle melanocytes. Narrow‑band ultraviolet B (NB‑UVB) has emerged as the first choice of treatment for vitiligo, but long‑term exposure may have serious consequences. Recently, it was reported that adipose‑derived stem cells (ADSCs) improve melanocyte growth and the efficacy of melanocyte transplantation. The present study aimed to examine the efficacy of NB‑UVB/ADSC‑transplantation combined therapy on a mouse vitiligo model and explore the underlying mechanisms by focusing on endoplasmic reticulum stress and cellular calcium (Ca2+) homeostasis. Vitiligo mice models were established by applying 40% monobenzone (MBZ) cream twice daily and treated with NB‑UVB/ADSC combination therapy. Some treated mice were also given ML385, a nuclear factor erythroid 2 like 2 (Nr2) inhibitor. Histopathological changes were evaluated using a depigmentation evaluation score and observed with hematoxylin and eosin staining on skin tissues. ELISA was used to measure diagnostic markers in plasma. Flow cytometric assay was performed to quantify CD3+, CD4+ and CD8+ levels. Expression levels of associated proteins were detected with western blot and immunofluorescence. Treatment of mice with MBZ‑induced depigmentation patches on the skin was accompanied with loss of redox balance and disruption of cellular Ca2+ homeostasis. Oxidative stress and Ca2+ unbalancing were improved after the mice were treated by NB‑UVB/ADSCs transplantation combination therapy. ML385, strongly negated the protective effect of NB‑UVB/ADSC transplantation combination therapy, indicating the critical role of Nr2 signaling. The findings improved the understanding of the pathogenesis of vitiligo and will guide future development of therapeutic strategies against it.
Collapse
Affiliation(s)
- Yuanyuan Bian
- Department of Dermatovenereal Disease, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hao Yu
- Department of Endocrinology, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Mingzhu Jin
- Department of Dermatovenereal Disease, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xinghua Gao
- Department of Dermatovenereal Disease, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
32
|
Ma J, Zhang Z, Wang Y, Shen H. Investigation of miR-126-3p loaded on adipose stem cell-derived exosomes for wound healing of full-thickness skin defects. Exp Dermatol 2021; 31:362-374. [PMID: 34694648 DOI: 10.1111/exd.14480] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 09/01/2021] [Accepted: 10/16/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the function of miR-126-3p loaded on adipose stem cell (ADSC)-derived exosomes (ADSC-Exos) in wound healing of full-thickness skin defects. METHODS ADSCs transfected with miR-126-3p mimic, miR-126-3p inhibitor or pcDNA3.1-PIK3R2, or PKH26-marked ADSC-Exos were cultured with fibroblasts or human umbilical vein endothelial cells (HUVECs). The proliferation and migration rates of fibroblasts and angiogenesis of HUVECs were measured. Rats with full-thickness skin defects were injected with ADSC-Exos or exosomes extracted from ADSCs transfected with miR-126-3p inhibitor and the wound healing rates were measured. The wound bed, collagen deposition and angiogenesis in injured rats were assessed. RESULTS ADSC-Exos could be ingested by fibroblasts and HUVECs. Co-incubation with ADSCs or ADSC-Exos promoted the proliferation and migration of fibroblasts and angiogenesis of HUVECs, which was further enhanced by miR-126-3p overexpression. Inhibition of ADSC-Exos or miR-126-3p or PIK3R2 overexpression suppressed the proliferation and migration of fibroblasts and angiogenesis of HUVECs. ADSC-derived exosomal miR-126-3p increased wound healing rate, collagen deposition and newly formed vessels in injured rats. CONCLUSION ADSC-derived exosomal miR-126-3p promotes wound healing of full-thickness skin defects by targeting PIK3R2.
Collapse
Affiliation(s)
- Jie Ma
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Zhaofeng Zhang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Yinmin Wang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Hua Shen
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| |
Collapse
|
33
|
A Comparative Study of the Effect of Anatomical Site on Multiple Differentiation of Adipose-Derived Stem Cells in Rats. Cells 2021; 10:cells10092469. [PMID: 34572123 PMCID: PMC8465004 DOI: 10.3390/cells10092469] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived from adipose tissue are evolved into various cell-based regenerative approaches. Adipose-derived stem cells (ASCs) isolated from rats are commonly used in tissue engineering studies. Still, there is a gap in knowledge about how the harvest locations influence and guide cell differentiation. This study aims to investigate how the harvesting site affects stem-cell-specific surface markers expression, pluripotency, and differentiation potential of ASCs in female Sprague Dawley rats. ASCs were extracted from the adipose tissue of the peri-ovarian, peri-renal, and mesenteric depots and were compared in terms of cell morphology. MSCs phenotype was validated by cell surfaces markers using flow cytometry. Moreover, pluripotent gene expression of Oct4, Nanog, Sox2, Rex-1, and Tert was evaluated by reverse transcriptase-polymerase chain reaction (RT-PCR). ASCs multipotency was evaluated by specific histological stains, and the results were confirmed by quantitative polymerase chain reaction (RT-qPCR) expression analysis of specific genes. There was a non-significant difference detected in the cell morphology and immunophenotype between different harvesting sites. ASCs from multiple locations were significantly varied in their capacity to differentiate into adipocytes, osteoblastic cells, and chondrocytes. To conclude, depot selection is a critical element that should be considered when using ASCs in tissue-specific cell-based regenerative therapies research.
Collapse
|
34
|
Yang J, Li S, He L, Chen M. Adipose-derived stem cells inhibit dermal fibroblast growth and induce apoptosis in keloids through the arachidonic acid-derived cyclooxygenase-2/prostaglandin E2 cascade by paracrine. BURNS & TRAUMA 2021; 9:tkab020. [PMID: 34514006 PMCID: PMC8430279 DOI: 10.1093/burnst/tkab020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/14/2020] [Indexed: 11/14/2022]
Abstract
Background The clinical features of keloids consist of aberrant proliferation, secretion, differentiation and apoptosis of keloid dermis-derived fibroblasts (KFBs). Notably, the apoptosis rate of KFBs is lower than the proliferation rate. Though the anti-fibrotic effect of adipose-derived stem cells (ADSCs) on keloids has become a hot topic of research, the exact anti-fibrotic mechanism of the paracrine effect remains unclear. This study aimed to find out how the conditioned medium of ADSCs (ADSC-CM) exerts an anti-fibrotic effect in KFBs. Methods KFBs and ADSCs were extracted and cultured. Then, ADSC-CM was prepared. Whether ADSC-CM could inhibit KFB growth and induce apoptosis was verified by the use of a cell counting kit-8, an 5-Ethynyl-2-deoxyuridine (Edu) kit and flow cytometry. The expressions of cyclooxygenase-1 (COX-1), COX-2, caspase 3 and B-cell lymphoma-2 (Bcl-2) in ADSC-CM-cultured KFBs were tested by real-time PCR and western blotting. To clarify the role of COX-2 in ADSC-CM-induced KFB apoptosis, a specific COX-2 inhibitor, celecoxib, was applied to KFBs cultured in ADSC-CM. Moreover, we tested the production of arachidonic acid (AA) and prostaglandin E2 (PGE2) by ELISA. Then, we established a keloid transplantation model in a nude mouse to validate the therapeutic effect in vivo. Results The proliferation ability of KFBs cultured in ADSC-CM was found to be weakened and apoptosis was significantly increased. Caspase 3 expression was significantly upregulated and Bcl-2 was downregulated in ADSC-CM-cultured KFBs. Furthermore, ADSC-CM strikingly elevated COX-2 mRNA and protein expressions, but COX-1 expression was unaltered. COX-2 inhibitors reduced ADSC-CM-induced apoptosis. Additionally, COX-2 inhibition blocked the elevation of caspase 3 and reversed the decrease in Bcl-2 expression. ADSC-CM increased PGE2 levels by 1.5-fold and this effect was restrained by COX-2 inhibition. In the nude mouse model, expressions of AA, COX-2 and PGE2 were higher in the translated keloid tissues after ADSC-CM injection than in the controls. Conclusions We showed activation of the COX-2/PGE2 cascade in KFBs in response to ADSC-CM. By employing a specific COX-2 inhibitor, COX-2/PGE2 cascade activation played a crucial role in mediating the ADSC-CM-induced KFB apoptosis and anti-proliferation effects.
Collapse
Affiliation(s)
- Jinxiu Yang
- Department of Burn and Plastic Surgery, the Fourth Medical Centre, Chinese People's Liberation Army General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100038, China
| | - Shiyi Li
- Department of Burn and Plastic Surgery, the Fourth Medical Centre, Chinese People's Liberation Army General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100038, China
| | - Leren He
- 7th Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.33 Ba Dachu Road, Shi Jingshan District, Beijing, 100144, China
| | - Minliang Chen
- Department of Burn and Plastic Surgery, the Fourth Medical Centre, Chinese People's Liberation Army General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100038, China
| |
Collapse
|
35
|
Construction of transplantable artificial vascular tissue based on adipose tissue-derived mesenchymal stromal cells by a cell coating and cryopreservation technique. Sci Rep 2021; 11:17989. [PMID: 34504254 PMCID: PMC8429436 DOI: 10.1038/s41598-021-97547-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 08/26/2021] [Indexed: 02/07/2023] Open
Abstract
Prevascularized artificial three-dimensional (3D) tissues are effective biomaterials for regenerative medicine. We have previously established a scaffold-free 3D artificial vascular tissue from normal human dermal fibroblasts (NHDFs) and umbilical vein-derived endothelial cells (HUVECs) by layer-by-layer cell coating technique. In this study, we constructed an artificial vascular tissue constructed by human adipose tissue-derived stromal cells (hASCs) and HUVECs (ASCVT) by a modified technique with cryopreservation. ASCVT showed a higher thickness with more dense vascular networks than the 3D tissue based on NHDFs. Correspondingly, 3D-cultured ASCs showed higher expression of several angiogenesis-related factors, including vascular endothelial growth factor-A and hepatic growth factor, compared to that of NHDFs. Moreover, perivascular cells in ASCVT were detected by pericyte markers, suggesting the differentiation of hASCs into pericyte-like cells. Subcutaneous transplantation of ASCVTs to nude mice resulted in an engraftment with anastomosis of host's vascular structures at 2 weeks after operation. In the engrafted tissue, the vascular network was surrounded by mural-like structure-forming hASCs, in which some parts developed to form vein-like structures at 4 weeks, suggesting the generation of functional vessel networks. These results demonstrated that cryopreserved human cells, including hASCs, could be used directly to construct the artificial transplantable tissue for regenerative medicine.
Collapse
|
36
|
Gkantsinikoudis N, Kapetanakis S, Magras I, Tsiridis E, Kritis A. Tissue-Engineering of Human Intervertebral Disc: A Concise Review. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:848-860. [PMID: 34409867 DOI: 10.1089/ten.teb.2021.0090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intervertebral disc (IVD) represents a structure of crucial structural and functional importance for human spine. Pathology of IVD institutes a frequently encountered condition in current clinical practice. Degenerative Disc Disease (DDD), the principal clinical representative of IVD pathology, constitutes an increasingly diagnosed spinal disorder associated with substantial morbidity and mortality in recent years. Despite the considerable incidence and socioeconomic burden of DDD, existing treatment modalities including conservative and surgical methods have been demonstrated to provide a limited therapeutic effect, being not capable of interrupting or reversing natural progress of underlying disease. These limitations underline the requirement for development of novel, innovative and more effective therapeutic strategies for DDD management. Within this literature framework, compromised IVD replacement with a viable IVD construct manufactured with Tissue-Engineering (TE) methods has been recommended as a promising therapeutic strategy for DDD. Existing preliminary preclinical data demonstrate that proper combination of cells from various sources, different scaffold materials and appropriate signaling molecules renders manufacturing of whole-IVD tissue-engineered constructs a technically feasible process. Aim of this narrative review is to critically summarize current published evidence regarding particular aspects of IVD-TE, primarily emphasizing in providing researchers in this field with practicable knowledge in order to enhance clinical translatability of their research and informing clinical practitioners about the features and capabilities of innovative TE science in the field of IVD-TE.
Collapse
Affiliation(s)
- Nikolaos Gkantsinikoudis
- School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th.), Department of Physiology and Pharmacology , Thessaloniki, Greece.,School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, Thessaloniki, Greece;
| | - Stylianos Kapetanakis
- Interbalkan European Medical Center, Spine Department and Deformities, Thessaloniki, Greece;
| | - Ioannis Magras
- AHEPA University General Hospital, Aristotle University of Thessaloniki, Department of Neurosurgery, Thessaloniki, Greece;
| | - Eleftherios Tsiridis
- Papageorgiou General Hospital, Aristotle University Medical School, Academic Orthopaedic Department, Thessaloniki Ring Road, Nea Efkarpia, Greece.,Aristotle University Thessaloniki, Balkan Center, Buildings A & B, Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center of Interdisciplinary Research and Innovation (C.I.R.I.), Thessaloniki, 10th km Thessaloniki-Thermi Rd, Greece;
| | - Aristeidis Kritis
- School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th.), Department of Physiology and Pharmacology , Thessaloniki, Greece.,School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, Thessaloniki, Greece;
| |
Collapse
|
37
|
Khazaei S, Khademi A, Nasr Esfahani MH, Khazaei M, Nekoofar MH, Dummer PMH. Isolation and Differentiation of Adipose-Derived Stem Cells into Odontoblast-Like Cells: A Preliminary In Vitro Study. CELL JOURNAL 2021; 23:288-293. [PMID: 34308571 PMCID: PMC8286457 DOI: 10.22074/cellj.2021.7325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/21/2020] [Indexed: 11/04/2022]
Abstract
Objective The aim of present study was to isolate and differentiate human adipose-derived stem cells (ASCs) into odontoblast-like cells. Materials and Methods In this experimental study, human adipose tissues were taken from the buccal fat pad of three individuals (mean age: 24.6 ± 2.1 years). The tissues were transferred to a laboratory in a sterile culture medium, divided into small pieces and digested by collagenase I (2 mg/mL, 60-90 minutes). ASCs were isolated by passing the cell suspension through cell strainers (70 and 40 μm), followed by incubation at 37ºC and 5% CO2 in Dulbecco's modified eagle medium (DMEM) supplemented with fetal bovine serum (FBS 5%) and penicillin/streptomycin (P/S). After three passages, the ASCs were harvested. Subsequently, flow cytometry and reverse transcriptase polymerase chain reaction (RT-PCR) were used to detect expression levels of NANOG and OCT4 to evaluate stemness. Then, a differentiation medium that included high-glucose DMEM supplemented with 10% FBS, dexamethasone (10 nM), sodium β-glycerophosphate (5 mM) and ascorbic acid (100 μM) was added. The cells were cultivated for four weeks, and the odontogenic medium was changed every two days. Cell differentiation was evaluated with Alizarin red staining and expressions of collagen I (COL1A1), dentin sialophosphoprotein (DSPP) and dentin matrix protein-1 (DMP1). Results The ASCs were effectively and easily isolated. They were negative for CD45 and positive for the CD105 and CD73 markers. The ASCs expressed OCT4 and NANOG. Differentiated cells highly expressed DSPP, COL1A1 and DMP1. Alizarin red staining revealed a positive reaction for calcium deposition. Conclusion ASCs were isolated successfully in high numbers from the buccal fat pad of human volunteers and were differentiated into odontoblast-like cells. These ASCs could be considered a new source of cells for use in regenerative endodontic treatments.
Collapse
Affiliation(s)
- Saber Khazaei
- Department of Endodontics, School of Dentistry and Dental Research Centre, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Endodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Abbasali Khademi
- Department of Endodontics, School of Dentistry and Dental Research Centre, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammad Hossein Nasr Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Centre, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | | | - Paul M H Dummer
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
38
|
Lindsay SL, Barnett SC. Therapeutic Potential of Niche-Specific Mesenchymal Stromal Cells for Spinal Cord Injury Repair. Cells 2021; 10:cells10040901. [PMID: 33919910 PMCID: PMC8070966 DOI: 10.3390/cells10040901] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
The use of mesenchymal stem/stromal cells (MSCs) for transplant-mediated repair represents an important and promising therapeutic strategy after spinal cord injury (SCI). The appeal of MSCs has been fuelled by their ease of isolation, immunosuppressive properties, and low immunogenicity, alongside the large variety of available tissue sources. However, despite reported similarities in vitro, MSCs sourced from distinct tissues may not have comparable biological properties in vivo. There is accumulating evidence that stemness, plasticity, immunogenicity, and adaptability of stem cells is largely controlled by tissue niche. The extrinsic impact of cellular niche for MSC repair potential is therefore important, not least because of its impact on ex vivo expansion for therapeutic purposes. It is likely certain niche-targeted MSCs are more suited for SCI transplant-mediated repair due to their intrinsic capabilities, such as inherent neurogenic properties. In addition, the various MSC anatomical locations means that differences in harvest and culture procedures can make cross-comparison of pre-clinical data difficult. Since a clinical grade MSC product is inextricably linked with its manufacture, it is imperative that cells can be made relatively easily using appropriate materials. We discuss these issues and highlight the importance of identifying the appropriate niche-specific MSC type for SCI repair.
Collapse
|
39
|
MiR-105 enhances osteogenic differentiation of hADSCs via the targeted regulation of SOX9. Tissue Cell 2021; 72:101540. [PMID: 33838353 DOI: 10.1016/j.tice.2021.101540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To investigate whether miR-105 can regulate the osteogenic differentiation of human adipose-derived mesenchymal stem cells (hADSCs) by targeting SOX9. METHODS The hADSCs were grouped for subsequent transfection and induction of osteogenic differentiation as follows: control, miR-NC, miR-105 mimics, miR-105 inhibitors, SOX9, SOX9 siRNA, miR-105 mimics + SOX9 and miR-105 inhibitors + SOX9 siRNA groups. Next, hADSCs were stained for alkaline phosphatase (ALP), and Alizarin Red S staining (ARS) was performed. Osteogenic differentiation-related genes and miR-105 expression were assessed by qRT-PCR, while SOX9 protein expression was determined by Western blotting. RESULTS MiR-105 expression was increased and SOX9 protein expression was decreased during the osteogenic differentiation of hADSCs. A dual-luciferase reporter assay confirmed SOX9 to be a target gene of miR-105. Compared with the control group, the miR-105 mimics and SOX9 siRNA groups had elevated BMP2, OPN, OCN, BSP, Osx and Runx2 mRNA expression with reduced SOX9 expression, as well as increased ARS intensity and ALP activity. After transfection of miR-105 inhibitors/SOX9 into hADSCs, the results were the opposite. Overexpressing SOX9 reversed the effect of miR-105 in promoting the osteogenic differentiation of hADSCs. CONCLUSION MiR-105 could target SOX9 to improve the expression of osteogenic differentiation genes and thus enhance the osteogenic differentiation of hADSCs.
Collapse
|
40
|
Photobiomodulation: An Effective Approach to Enhance Proliferation and Differentiation of Adipose-Derived Stem Cells into Osteoblasts. Stem Cells Int 2021; 2021:8843179. [PMID: 33833810 PMCID: PMC8012132 DOI: 10.1155/2021/8843179] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/07/2021] [Accepted: 03/11/2021] [Indexed: 01/07/2023] Open
Abstract
Osteoporosis is regarded as the most common chronic metabolic bone condition in humans. In osteoporosis, bone mesenchymal stem cells (MSCs) have reduced cellular function. Regenerative medicine using adipose-derived stem cell (ADSC) transplantation can promote the growth and strength of new bones, improve bone stability, and reduce the risk of fractures. Various methods have been attempted to differentiate ADSCs to functioning specialized cells for prospective clinical application. However, commonly used therapies have resulted in damage to the donor site and morbidity, immune reactions, carcinogenic generation, and postoperative difficulties. Photobiomodulation (PBM) improves ADSC differentiation and proliferation along with reducing clinical difficulties such as treatment failures to common drug therapies and late initiation of treatment. PBM is a noninvasive, nonthermal treatment that encourages cells to produce more energy and to undergo self-repair by using visible green and red and invisible near-infrared (NIR) radiation. The use of PBM for ADSC proliferation and differentiation has been widely studied with multiple outcomes observed due to laser fluence and wavelength dependence. In this article, the potential for differentiating ADSCs into osteoblasts and the various methods used, including biological induction, chemical induction, and PBM, will be addressed. Likewise, the optimal laser parameters that could improve the proliferation and differentiation of ADSC, translating into clinical success, will be commented on.
Collapse
|
41
|
Cheng J, Zou Q, Xue Y, Sun C, Zhang D. Mechanical stretch promotes antioxidant responses and cardiomyogenic differentiation in P19 cells. J Tissue Eng Regen Med 2021; 15:453-462. [PMID: 33743188 DOI: 10.1002/term.3184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/26/2021] [Accepted: 02/14/2021] [Indexed: 11/07/2022]
Abstract
Accumulating evidence has suggested that mechanical stimuli play a crucial role in regulating the lineage-specific differentiation of stem cells through fine-tuning redox balance. We aimed to investigate the effects of cyclic tensile strain (CTS) on the expression of antioxidant enzymes and cardiac-specific genes in P19 cells, a widely characterized tool for cardiac differentiation research. A stretching device was applied to generate different magnitude and duration of cyclic strains on P19 cells. The messenger RNA and protein levels of targeted genes were determined by real-time polymerase chain reaction and Western blot assays, respectively. Proper magnitude and duration of cognitive stimulation therapy (CST) stimulation substantially enhanced the expression of both antioxidant enzymes and cardiac-specific genes in P19 cells. Sirtuin 1 (SIRT1) played an essential role in the CTS-induced cardiomyogenic differentiation of P19, as evidenced by changes in the expression of antioxidant enzymes and cardiac-specific genes. Mechanical loading promoted the cardiomyogenic differentiation of P19 cells. SIRT1 was involved in CST-mediated P19 differentiation, implying that SIRT1 might serve as an important target for developing methods to promote cardiomyogenic differentiation of stem cells.
Collapse
Affiliation(s)
- Jin Cheng
- Department of Cardiology, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Qing Zou
- Department of Cardiology, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Yugang Xue
- Department of Cardiology, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Chuang Sun
- Department of Cardiology, Xi'An International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Dongwei Zhang
- Department of Cardiology, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
42
|
Capella-Monsonís H, Zeugolis DI. Decellularized xenografts in regenerative medicine: From processing to clinical application. Xenotransplantation 2021; 28:e12683. [PMID: 33709410 DOI: 10.1111/xen.12683] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/28/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022]
Abstract
Decellularized xenografts are an inherent component of regenerative medicine. Their preserved structure, mechanical integrity and biofunctional composition have well established them in reparative medicine for a diverse range of clinical indications. Nonetheless, their performance is highly influenced by their source (ie species, age, tissue) and processing (ie decellularization, crosslinking, sterilization and preservation), which govern their final characteristics and determine their success or failure for a specific clinical target. In this review, we provide an overview of the different sources and processing methods used in decellularized xenografts fabrication and discuss their effect on the clinical performance of commercially available decellularized xenografts.
Collapse
Affiliation(s)
- Héctor Capella-Monsonís
- 1Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.,Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Dimitrios I Zeugolis
- 1Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.,Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.,Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland
| |
Collapse
|
43
|
Sotoodehnejadnematalahi F, Moghadasali R, Hajinasrollah M, Ehsani E, Hajizadeh-Saffar E, Sodeifi N, Saidi R, Zarrabi M, Farzanehkhah M, Sadeghi B, Baharvand H, Aghdami N. Immunomodulatory Activity of Human Bone Marrow and Adipose-Derived Mesenchymal Stem Cells Prolongs Allogenic Skin Graft Survival in Nonhuman Primates. CELL JOURNAL 2021; 23:1-13. [PMID: 33650815 PMCID: PMC7944119 DOI: 10.22074/cellj.2021.6895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/09/2019] [Indexed: 11/04/2022]
Abstract
Objective In the present study, we examined the tolerance-inducing effects of human adipose-derived mesenchymal stem cells (hAD-MSCs) and bone marrow-derived MSCs (hBM-MSCs) on a nonhuman primate model of skin transplantation. Materials and Methods In this experimental study, allogenic and xenogeneic of immunomodulatory properties of human AD-MSCs and BM-MSCs were evaluated by mixed lymphocyte reaction (MLR) assays. Human MSCs were obtained from BM or AD tissues (from individuals of either sex with an age range of 35 to 65 years) and intravenously injected (2×106 MSCs/kg) after allogeneic skin grafting in a nonhuman primate model. The skin sections were evaluated by H and E staining for histopathological evaluations, particularly inflammation and rejection reaction of grafts after 96 hours of cell injection. At the mRNA and protein levels, cellular mediators of inflammation, such as CD4+IL-17+ (T helper 17; Th17) and CD4+INF-γ+ (T helper 1, Th1) cells, along with CD4+FoxP3+ cells (Treg), as the mediators of immunomodulation, were measured by RT-PCR and flow cytometry analyses. Results A significant Treg cells expansion was observed in MSCs-treated animals which reached the zenith at 24 hours and remained at a high concentration for 96 hours; however, Th1 and Th17 cells were significantly decreased. Our results showed that human MSCs significantly decrease Th1 and Th17 cell proliferation by decreasing interleukin-17 (IL-17) and interferon-γ (INF-γ) production and significantly increase Treg cell proliferation by increasing FoxP3 production. They also extend the allogenic skin graft survival in nonhuman primates. Histological evaluations showed no obvious presence of inflammatory cells or skin redness or even bulging after MSCs injection up to 96 hours, compared to the group without MSCs. There were no significant differences between hBM-MSCs and hAD-MSCs in terms of histopathological scores and inflammatory responses (P<0.05). Conclusion It seems that MSCs could be regarded as a valuable immunomodulatory tool to reduce the use of immunosuppressive agents.
Collapse
Affiliation(s)
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Department of Regenerative Medicine, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Mostafa Hajinasrollah
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ehsan Ehsani
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Biology, Roudehen Branch, Islamic Azad University, Roudehen, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Sodeifi
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Reza Saidi
- Department of Surgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Zarrabi
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | | | - Bahareh Sadeghi
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Hossein Baharvand
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Nasser Aghdami
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
44
|
He T, Yang J, Liu P, Xu L, Lü Q, Tan Q. [Research progress of adipose-derived stem cells in skin scar prevention and treatment]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2021; 35:234-240. [PMID: 33624480 DOI: 10.7507/1002-1892.202007083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Objective To review the research progress of adipose-derived stem cells (ADSCs) in skin scar prevention and treatment. Methods The related literature was extensively reviewed and analyzed. The recent in vitroand in vivo experiments and clinical studies on the role of ADSCs in skin scar prevention and treatment, and the possible mechanisms and biomaterials to optimize the effect of ADSCs were summarized. Results As demonstrated by in vitro and in vivo experiments and clinical studies, ADSCs participate in the whole process of skin wound healing and may prevent and treat skin scars by reducing inflammation, promoting angiogenesis, or inhibiting (muscle) fibroblasts activity to reduce collagen deposition through the p38/mitogen-activated protein kinase, peroxisome proliferator activated receptor γ, transforming growth factor β 1/Smads pathways. Moreover, bioengineered materials such as hydrogel from acellular porcine adipose tissue, porcine small-intestine submucosa, and poly (3-hydroxybutyrate-co-hydroxyvalerate) scaffold may further enhance the efficacy of ADSCs in preventing and treating skin scars. Conclusion Remarkable progress has been made in the application of ADSCs in skin scar prevention and treatment. While, further studies are still needed to explore the application methods of ADSCs in the clinic.
Collapse
Affiliation(s)
- Tao He
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China;Laboratory of Stem Cell and Tissue Engineering, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Jiqiao Yang
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Pengcheng Liu
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China;Laboratory of Stem Cell and Tissue Engineering, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Li Xu
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China;Laboratory of Stem Cell and Tissue Engineering, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Qing Lü
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Qiuwen Tan
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China;Laboratory of Stem Cell and Tissue Engineering, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| |
Collapse
|
45
|
Abstract
Background: Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease of upper and lower motor neurons with high burden on society. Despite tremendous efforts over the last several decades, there is still no definite cure for ALS. Up to now, only two disease-modifying agents, riluzole and edaravone, are approved by U.S. Food and Drug Administration (FDA) for ALS treatment, which only modestly improves survival and disease progression. Major challenging issues to find an effective therapy are heterogeneity in the pathogenesis and genetic variability of ALS. As such, stem cell therapy has been recently a focus of both preclinical and clinical investigations of ALS. This is because stem cells have multifaceted features that can potentially target multiple pathogenic mechanisms in ALS even though its underlying mechanisms are not completely elucidated. Methods & Results: Here, we will have an overview of stem cell therapy in ALS, including their therapeutic mechanisms, the results of recent clinical trials as well as ongoing clinical trials. In addition, we will further discuss complications and limitations of stem cell therapy in ALS. Conclusion: The determination of whether stem cells offer a viable treatment strategy for ALS rests on well-designed and appropriately powered future clinical trials. Randomized, double-blinded, and sham-controlled studies would be valuable.
Collapse
Affiliation(s)
- Goun Je
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA USA
| | - Kiandokht Keyhanian
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA USA
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA USA
| |
Collapse
|
46
|
Al-Ghadban S, Bunnell BA. Adipose Tissue-Derived Stem Cells: Immunomodulatory Effects and Therapeutic Potential. Physiology (Bethesda) 2021; 35:125-133. [PMID: 32027561 DOI: 10.1152/physiol.00021.2019] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived stem cells (ASCs) can self-renew and differentiate along multiple cell lineages. ASCs are also potently anti-inflammatory due to their inherent ability to regulate the immune system by secreting anti-inflammatory cytokines and growth factors that play a crucial role in the pathology of many diseases, including multiple sclerosis, diabetes mellitus, Crohn's, SLE, and graft-versus-host disease. The immunomodulatory effects and mechanisms of action of ASCs on pathological conditions are reviewed here.
Collapse
Affiliation(s)
- Sara Al-Ghadban
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Bruce A Bunnell
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana.,Department of Pharmacology, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
47
|
Giduthuri AT, Theodossiou SK, Schiele NR, Srivastava SK. Dielectrophoretic Characterization of Tenogenically Differentiating Mesenchymal Stem Cells. BIOSENSORS 2021; 11:50. [PMID: 33669223 PMCID: PMC7919818 DOI: 10.3390/bios11020050] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
Tendons are collagenous musculoskeletal tissues that connect muscles to bones and transfer the forces necessary for movement. Tendons are susceptible to injury and heal poorly, with long-term loss of function. Mesenchymal stem cell (MSC)-based therapies are a promising approach for treating tendon injuries but are challenged by the difficulties of controlling stem cell fate and of generating homogenous populations of stem cells optimized for tenogenesis (differentiation toward tendon). To address this issue, we aim to explore methods that can be used to identify and ultimately separate tenogenically differentiated MSCs from non-tenogenically differentiated MSCs. In this study, baseline and tenogenically differentiating murine MSCs were characterized for dielectric properties (conductivity and permittivity) of their outer membrane and cytoplasm using a dielectrophoretic (DEP) crossover technique. Experimental results showed that unique dielectric properties distinguished tenogenically differentiating MSCs from controls after three days of tenogenic induction. A single shell model was used to quantify the dielectric properties and determine membrane and cytoplasm conductivity and permittivity. Together, cell responses at the crossover frequency, cell morphology, and shell models showed that changes potentially indicative of early tenogenesis could be detected in the dielectric properties of MSCs as early as three days into differentiation. Differences in dielectric properties with tenogenesis indicate that the DEP-based label-free separation of tenogenically differentiating cells is possible and avoids the complications of current label-dependent flow cytometry-based separation techniques. Overall, this work illustrates the potential of DEP to generate homogeneous populations of differentiated stem cells for applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
| | | | | | - Soumya K. Srivastava
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, ID 83844-1021, USA; (A.T.G.); (S.K.T.); (N.R.S.)
| |
Collapse
|
48
|
Research Progress on Stem Cell Therapies for Articular Cartilage Regeneration. Stem Cells Int 2021; 2021:8882505. [PMID: 33628274 PMCID: PMC7895563 DOI: 10.1155/2021/8882505] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Injury of articular cartilage can cause osteoarthritis and seriously affect the physical and mental health of patients. Unfortunately, current surgical treatment techniques that are commonly used in the clinic cannot regenerate articular cartilage. Regenerative medicine involving stem cells has entered a new stage and is considered the most promising way to regenerate articular cartilage. In terms of theories on the mechanism, it was thought that stem cell-mediated articular cartilage regeneration was achieved through the directional differentiation of stem cells into chondrocytes. However, recent evidence has shown that the stem cell secretome plays an important role in biological processes such as the immune response, inflammation regulation, and drug delivery. At the same time, the stem cell secretome can effectively mediate the process of tissue regeneration. This new theory has attributed the therapeutic effect of stem cells to their paracrine effects. The application of stem cells is not limited to exogenous stem cell transplantation. Endogenous stem cell homing and in situ regeneration strategies have received extensive attention. The application of stem cell derivatives, such as conditioned media, extracellular vesicles, and extracellular matrix, is an extension of stem cell paracrine theory. On the other hand, stem cell pretreatment strategies have also shown promising therapeutic effects. This article will systematically review the latest developments in these areas, summarize challenges in articular cartilage regeneration strategies involving stem cells, and describe prospects for future development.
Collapse
|
49
|
The Effect of Conditioned Media From Human Adipocyte-Derived Mesenchymal Stem Cells on Androgenetic Alopecia After Nonablative Fractional Laser Treatment. Dermatol Surg 2021; 46:1698-1704. [PMID: 32769526 DOI: 10.1097/dss.0000000000002518] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The conditioned media from adipocyte-derived mesenchymal stem cells-conditioned media (ADSC-CM) contains cytokines and growth factors that stimulate hair regeneration. OBJECTIVE We evaluated the efficacy and safety of human ADSC-CM treatment on patients who underwent nonablative fractional laser for the treatment of androgenetic alopecia (AGA). MATERIALS AND METHODS Thirty patients who underwent nonablative fractional laser treatment were topically administered either ADSC-CM or placebo solution. As a primary outcome, phototrichograms were taken to measure changes in hair density at each visit. In addition, global improvement scores (GISs) were compared by clinical digital photographs, which were taken at the initial and final visits, and assessed by 2 independent dermatologists. Finally, the investigator's improvement score was measured by questionnaire response during the final visit. RESULTS Hair density comparisons during the treatment period revealed that the ADSC-CM group had significantly higher final densities compared with the placebo group. The GIS of the ADSC-CM group was also significantly higher than the placebo group. Finally, no adverse effects associated with the application of ADSC-CM were noted during the study. CONCLUSION The application of ADSC-CM after nonablative fractional laser treatment accelerated increases in hair density and volume in AGA patients.
Collapse
|
50
|
Wang L, Li H, Lin J, He R, Chen M, Zhang Y, Liao Z, Zhang C. CCR2 improves homing and engraftment of adipose-derived stem cells in dystrophic mice. Stem Cell Res Ther 2021; 12:12. [PMID: 33413615 PMCID: PMC7791736 DOI: 10.1186/s13287-020-02065-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/02/2020] [Indexed: 02/17/2023] Open
Abstract
Background Dystrophinopathy, a common neuromuscular disorder caused by the absence of dystrophin, currently lacks effective treatments. Systemic transplantation of adipose-derived stem cells (ADSCs) is a promising treatment approach, but its low efficacy remains a challenge. Chemokine system-mediated stem cell homing plays a critical role in systemic transplantation. Here, we investigated whether overexpression of a specific chemokine receptor could improve muscle homing and therapeutic effects of ADSC systemic transplantation in dystrophic mice. Methods We analysed multiple microarray datasets from the Gene Expression Omnibus to identify a candidate chemokine receptor and then evaluated the protein expression of target ligands in different tissues and organs of dystrophic mice. The candidate chemokine receptor was overexpressed using the lentiviral system in mouse ADSCs, which were used for systemic transplantation into the dystrophic mice, followed by evaluation of motor function, stem cell muscle homing, dystrophin expression, and muscle pathology. Results Chemokine-profile analysis identified C–C chemokine receptor (CCR)2 as the potential target for improving ADSC homing. We found that the levels of its ligands C–C chemokine ligand (CCL)2 and CCL7 were higher in muscles than in other tissues and organs of dystrophic mice. Additionally, CCR2 overexpression improved ADSC migration ability and maintained their multilineage-differentiation potentials. Compared with control ADSCs, transplantation of those overexpressing CCR2 displayed better muscle homing and further improved motor function, dystrophin expression, and muscle pathology in dystrophic mice. Conclusions These results demonstrated that CCR2 improved ADSC muscle homing and therapeutic effects following systemic transplantation in dystrophic mice.
Collapse
Affiliation(s)
- Liang Wang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, GD, China.,National Key Clinical Department and Key Discipline of Neurology, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, No. 58 Zhongshan Road 2, Guangzhou, GD, 510080, China
| | - Huan Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, GD, China.,National Key Clinical Department and Key Discipline of Neurology, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, No. 58 Zhongshan Road 2, Guangzhou, GD, 510080, China
| | - Jinfu Lin
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, GD, China.,National Key Clinical Department and Key Discipline of Neurology, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, No. 58 Zhongshan Road 2, Guangzhou, GD, 510080, China
| | - Ruojie He
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, GD, China.,National Key Clinical Department and Key Discipline of Neurology, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, No. 58 Zhongshan Road 2, Guangzhou, GD, 510080, China
| | - Menglong Chen
- Department of Neurology, Guangzhou Overseas Chinese Hospital, No. 613 Huangpu Road, Guangzhou, GD, 510630, China
| | - Yu Zhang
- Department of Neurology, Guangzhou Overseas Chinese Hospital, No. 613 Huangpu Road, Guangzhou, GD, 510630, China
| | - Ziyu Liao
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, GD, China.,National Key Clinical Department and Key Discipline of Neurology, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, No. 58 Zhongshan Road 2, Guangzhou, GD, 510080, China
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, GD, China. .,National Key Clinical Department and Key Discipline of Neurology, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, No. 58 Zhongshan Road 2, Guangzhou, GD, 510080, China.
| |
Collapse
|