151
|
Gong Q, Zeng J, Zhang X, Huang Y, Chen C, Quan J, Ling J. Effect of erythropoietin on angiogenic potential of dental pulp cells. Exp Ther Med 2021; 22:1079. [PMID: 34447472 PMCID: PMC8355638 DOI: 10.3892/etm.2021.10513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022] Open
Abstract
Erythropoietin (EPO) is a 34-kDa glycoprotein that possesses the potential for angiogenesis, as well as anti-inflammatory and anti-apoptotic properties. The present study aimed to examine the effect of EPO on the angiogenesis of dental pulp cells (DPCs) and to explore the underlying mechanisms of these effects. It was demonstrated that EPO not only promoted DPCs proliferation but also induced angiogenesis of DPCs in a paracrine fashion. EPO enhanced the angiogenic capacity by stimulating DPCs to secrete a series of angiogenic cytokines. ELISA confirmed that high concentrations of EPO increased the production of MMP-3 and angiopoietin-1 but decreased the secretion of IL-6. Furthermore, EPO activated the ERK1/2 and p38 signaling pathways in DPCs, while inhibition of these pathways diminished the angiogenesis capacity of DPCs. The present study suggested that EPO may have an important role in the repair and regeneration of dental pulp.
Collapse
Affiliation(s)
- Qimei Gong
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, P.R. China
| | - Junyu Zeng
- Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xufang Zhang
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, P.R. China
| | - Yihua Huang
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, P.R. China
| | - Chanchan Chen
- Department of Stomatology, Shenzhen Children's Hospital, Shenzhen, Guangdong 518038, P.R. China
| | - Jingjing Quan
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, P.R. China
| | - Junqi Ling
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
152
|
Valente S, Ciavarella C, Hernández-Aguilera A, Salvador FA, Buzzi M, Joven J, Pasquinelli G. Phenotypic, morphological, and metabolic characterization of vascular-spheres from human vascular mesenchymal stem cells. Microsc Res Tech 2021; 85:447-459. [PMID: 34448515 PMCID: PMC9290655 DOI: 10.1002/jemt.23918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/15/2021] [Accepted: 07/31/2021] [Indexed: 01/01/2023]
Abstract
The ability to form spheroids under non‐adherent conditions is a well‐known property of human mesenchymal stem cells (hMSCs), in addition to stemness and multilineage differentiation features. In the present study, we tested the ability of hMSCs isolated from the vascular wall (hVW‐MSCs) to grow as spheres, and provide a characterization of this 3D model. hVW‐MSCs were isolated from femoral arteries through enzymatic digestion. Spheres were obtained using ultra‐low attachment and hanging drop methods. Immunophenotype and pluripotent genes (SOX‐2, OCT‐4, NANOG) were analyzed by immunocytochemistry and real‐time PCR, respectively. Spheres histological and ultrastructural architecture were examined. Cell viability and proliferative capacity were measured using LIVE/DEATH assay and ki‐67 proliferation marker. Metabolomic profile was obtained with liquid chromatography–mass spectrometry. In 2D, hVW‐MSCs were spindle‐shaped, expressed mesenchymal antigens, and displayed mesengenic potential. 3D cultures of hVW‐MSCs were CD44+, CD105low, CD90low, exhibited a low propensity to enter the cell cycle as indicated by low percentage of ki‐67 expression and accumulated intermediate metabolites pointing to slowed metabolism. The 3D model of hVW‐MSCs exhibits stemness, dormancy and slow metabolism, typically observed in stem cell niches. This culture strategy can represent an accurate model to investigate hMSCs features for future clinical applications in the vascular field.
Collapse
Affiliation(s)
- Sabrina Valente
- DIMES - Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Carmen Ciavarella
- DIMES - Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Anna Hernández-Aguilera
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, IISPV, Universitat Rovira i Virgili, Reus, Spain.,Campus of International Excellence Southern Catalonia, Tarragona, Spain
| | - Fernández-Arroyo Salvador
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, IISPV, Universitat Rovira i Virgili, Reus, Spain.,Campus of International Excellence Southern Catalonia, Tarragona, Spain
| | - Marina Buzzi
- Emilia Romagna Cord Blood Bank - Transfusion Service, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, IISPV, Universitat Rovira i Virgili, Reus, Spain.,Campus of International Excellence Southern Catalonia, Tarragona, Spain
| | - Gianandrea Pasquinelli
- DIMES - Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Subcellular Nephro-Vascular Diagnostic Program, Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
153
|
Evens L, Beliën H, D’Haese S, Haesen S, Verboven M, Rummens JL, Bronckaers A, Hendrikx M, Deluyker D, Bito V. Combinational Therapy of Cardiac Atrial Appendage Stem Cells and Pyridoxamine: The Road to Cardiac Repair? Int J Mol Sci 2021; 22:ijms22179266. [PMID: 34502175 PMCID: PMC8431115 DOI: 10.3390/ijms22179266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/04/2022] Open
Abstract
Myocardial infarction (MI) occurs when the coronary blood supply is interrupted. As a consequence, cardiomyocytes are irreversibly damaged and lost. Unfortunately, current therapies for MI are unable to prevent progression towards heart failure. As the renewal rate of cardiomyocytes is minimal, the optimal treatment should achieve effective cardiac regeneration, possibly with stem cells transplantation. In that context, our research group identified the cardiac atrial appendage stem cells (CASCs) as a new cellular therapy. However, CASCs are transplanted into a hostile environment, with elevated levels of advanced glycation end products (AGEs), which may affect their regenerative potential. In this study, we hypothesize that pyridoxamine (PM), a vitamin B6 derivative, could further enhance the regenerative capacities of CASCs transplanted after MI by reducing AGEs’ formation. Methods and Results: MI was induced in rats by ligation of the left anterior descending artery. Animals were assigned to either no therapy (MI), CASCs transplantation (MI + CASCs), or CASCs transplantation supplemented with PM treatment (MI + CASCs + PM). Four weeks post-surgery, global cardiac function and infarct size were improved upon CASCs transplantation. Interstitial collagen deposition, evaluated on cryosections, was decreased in the MI animals transplanted with CASCs. Contractile properties of resident left ventricular cardiomyocytes were assessed by unloaded cell shortening. CASCs transplantation prevented cardiomyocyte shortening deterioration. Even if PM significantly reduced cardiac levels of AGEs, cardiac outcome was not further improved. Conclusion: Limiting AGEs’ formation with PM during an ischemic injury in vivo did not further enhance the improved cardiac phenotype obtained with CASCs transplantation. Whether AGEs play an important deleterious role in the setting of stem cell therapy after MI warrants further examination.
Collapse
Affiliation(s)
- Lize Evens
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Hanne Beliën
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Sarah D’Haese
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Sibren Haesen
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Maxim Verboven
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Jean-Luc Rummens
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
- UHasselt—Hasselt University, Faculty of Medicine and Life Sciences, Agoralaan, 3590 Diepenbeek, Belgium
| | - Annelies Bronckaers
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Marc Hendrikx
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Dorien Deluyker
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Virginie Bito
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
- Correspondence: ; Tel.: +32-11269285
| |
Collapse
|
154
|
Amanat M, Majmaa A, Zarrabi M, Nouri M, Akbari MG, Moaiedi AR, Ghaemi O, Zamani F, Najafi S, Badv RS, Vosough M, Hamidieh AA, Salehi M, Montazerlotfelahi H, Tavasoli AR, Heidari M, Mohebi H, Fatemi A, Garakani A, Ashrafi MR. Clinical and imaging outcomes after intrathecal injection of umbilical cord tissue mesenchymal stem cells in cerebral palsy: a randomized double-blind sham-controlled clinical trial. Stem Cell Res Ther 2021; 12:439. [PMID: 34362453 PMCID: PMC8343813 DOI: 10.1186/s13287-021-02513-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/08/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND This study assessed the safety and efficacy of intrathecal injection of umbilical cord tissue mesenchymal stem cells (UCT-MSC) in individuals with cerebral palsy (CP). The diffusion tensor imaging (DTI) was performed to evaluate the alterations in white-matter integrity. METHODS Participants (4-14 years old) with spastic CP were assigned in 1:1 ratio to receive either UCT-MSC or sham procedure. Single-dose (2 × 107) cells were administered in the experimental group. Small needle pricks to the lower back were performed in the sham-control arm. All individuals were sedated to prevent awareness. The primary endpoints were the mean changes in gross motor function measure (GMFM)-66 from baseline to 12 months after procedures. The mean changes in the modified Ashworth scale (MAS), pediatric evaluation of disability inventory (PEDI), and CP quality of life (CP-QoL) were also assessed. Secondary endpoints were the mean changes in fractional anisotropy (FA) and mean diffusivity (MD) of corticospinal tract (CST) and posterior thalamic radiation (PTR). RESULTS There were 36 participants in each group. The mean GMFM-66 scores after 12 months of intervention were significantly higher in the UCT-MSC group compared to baseline (10.65; 95%CI 5.39, 15.91) and control (β 8.07; 95%CI 1.62, 14.52; Cohen's d 0.92). The increase was also seen in total PEDI scores (vs baseline 8.53; 95%CI 4.98, 12.08; vs control: β 6.87; 95%CI 1.52, 12.21; Cohen's d 0.70). The mean change in MAS scores after 12 months of cell injection reduced compared to baseline (-1.0; 95%CI -1.31, -0.69) and control (β -0.72; 95%CI -1.18, -0.26; Cohen's d 0.76). Regarding CP-QoL, mean changes in domains including friends and family, participation in activities, and communication were higher than the control group with a large effect size. The DTI analysis in the experimental group showed that mean FA increased (CST 0.032; 95%CI 0.02, 0.03. PTR 0.024; 95%CI 0.020, 0.028) and MD decreased (CST -0.035 × 10-3; 95%CI -0.04 × 10-3, -0.02 × 10-3. PTR -0.045 × 10-3; 95%CI -0.05 × 10-3, -0.03 × 10-3); compared to baseline. The mean changes were significantly higher than the control group. CONCLUSIONS The UCT-MSC transplantation was safe and may improve the clinical and imaging outcomes. TRIAL REGISTRATION The study was registered with ClinicalTrials.gov ( NCT03795974 ).
Collapse
Affiliation(s)
- Man Amanat
- Department of Science and Research Branch, AJA University of Medical Sciences, Tehran, Iran
| | - Anahita Majmaa
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Zarrabi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Masoumeh Nouri
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Masood Ghahvechi Akbari
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Reza Moaiedi
- Department of Pediatric Neurology, Clinical Research Development Center of Children Hospital, Hormozgan University of Medical Sciences, Bandar Abass, Iran
| | - Omid Ghaemi
- Pediatrics Center of Excellence, Department of Radiology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Zamani
- Pediatrics Center of Excellence, Department of Radiology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sharif Najafi
- Clinical Biomechanics and Ergonomics Research Center, Department of Physical Medicine and Rehabilitation, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Shervin Badv
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatrics Center of Excellence Pediatric Hematology, Oncology and Stem Cell Transplantation Department, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Salehi
- Psychiatry and Psychology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hadi Montazerlotfelahi
- Department of Pediatrics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Reza Tavasoli
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Heidari
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Mohebi
- Department of Pediatric Neurology, AJA University of Medical Sciences, Tehran, Iran
| | - Ali Fatemi
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD, 21205, USA
- Department of Neurology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Amir Garakani
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mahmoud Reza Ashrafi
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
155
|
Vanorlé M, Lemaire A, di Pietrantonio L, Horckmans M, Communi D. UTP is a regulator of in vitro and in vivo angiogenic properties of cardiac adipose-derived stem cells. Purinergic Signal 2021; 17:681-691. [PMID: 34351588 DOI: 10.1007/s11302-021-09812-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/11/2021] [Indexed: 10/20/2022] Open
Abstract
The ability of cardiac adipose-derived stem cells (cADSC) to differentiate into multiple cell types has opened new perspectives in cardiac cell-based regenerative therapies. P2Y nucleotide receptors have already been described as regulators of adipogenic differentiation of cADSC and bone marrow-derived stem cells. In this study, we defined UTP as a regulator of cADSC endothelial differentiation. A daily UTP stimulation of cADSC during endothelial predifferentiation increased their capacity to form an endothelial network in matrigel. Additionally, pro-angiogenic UTP target genes such as epiregulin and hyaluronan synthase-1 were identified in predifferentiated cADSC by RNA sequencing experiments. Their regulation by UTP was confirmed by qPCR and ELISA experiments. We then evaluated the capacity of UTP-treated predifferentiated cADSC to increase post-ischemic revascularization in mice subjected to left anterior descending artery ligation. Predifferentiated cADSC treated or not with UTP were injected in the periphery of the infarcted zone, 3 days after ligation. We observed a significant increase of capillary density 14 and 30 days after UTP-treated predifferentiated cADSC injection, correlated with a reduction of cardiac fibrosis. This revascularization increase was not observed after injection of UTP-treated cADSC deficient for UTP and ATP nucleotide receptor P2Y2. The present study highlights the P2Y2 receptor as a regulator of cADSC endothelial differentiation and as a potential target for the therapeutic use of cADSC in post-ischemic heart revascularization.
Collapse
Affiliation(s)
- Marion Vanorlé
- Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, ULB, Building C (5th floor), Campus Erasme, 808 Route de Lennik, 1070, Brussels, Belgium
| | - Anne Lemaire
- Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, ULB, Building C (5th floor), Campus Erasme, 808 Route de Lennik, 1070, Brussels, Belgium
| | - Larissa di Pietrantonio
- Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, ULB, Building C (5th floor), Campus Erasme, 808 Route de Lennik, 1070, Brussels, Belgium
| | - Michael Horckmans
- Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, ULB, Building C (5th floor), Campus Erasme, 808 Route de Lennik, 1070, Brussels, Belgium
| | - Didier Communi
- Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, ULB, Building C (5th floor), Campus Erasme, 808 Route de Lennik, 1070, Brussels, Belgium.
| |
Collapse
|
156
|
Nagelkerke A, Ojansivu M, van der Koog L, Whittaker TE, Cunnane EM, Silva AM, Dekker N, Stevens MM. Extracellular vesicles for tissue repair and regeneration: Evidence, challenges and opportunities. Adv Drug Deliv Rev 2021; 175:113775. [PMID: 33872693 DOI: 10.1016/j.addr.2021.04.013] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/20/2021] [Accepted: 04/15/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) are biological nanoparticles naturally secreted by cells, acting as delivery vehicles for molecular messages. During the last decade, EVs have been assigned multiple functions that have established their potential as therapeutic mediators for a variety of diseases and conditions. In this review paper, we report on the potential of EVs in tissue repair and regeneration. The regenerative properties that have been associated with EVs are explored, detailing the molecular cargo they carry that is capable of mediating such effects, the signaling cascades triggered in target cells and the functional outcome achieved. EV interactions and biodistribution in vivo that influence their regenerative effects are also described, particularly upon administration in combination with biomaterials. Finally, we review the progress that has been made for the successful implementation of EV regenerative therapies in a clinical setting.
Collapse
Affiliation(s)
- Anika Nagelkerke
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, P.O. Box 196, XB20, 9700 AD Groningen, the Netherlands.
| | - Miina Ojansivu
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.
| | - Luke van der Koog
- Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, P.O. Box 196, XB10, 9700 AD Groningen, the Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Thomas E Whittaker
- Department of Materials, Imperial College London, London, UK; Department of Bioengineering, Imperial College London, London, UK; Institute of Biomedical Engineering, Imperial College London, London, UK
| | - Eoghan M Cunnane
- Department of Materials, Imperial College London, London, UK; Department of Bioengineering, Imperial College London, London, UK; Institute of Biomedical Engineering, Imperial College London, London, UK.
| | - Andreia M Silva
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Niek Dekker
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Molly M Stevens
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; Department of Materials, Imperial College London, London, UK; Department of Bioengineering, Imperial College London, London, UK; Institute of Biomedical Engineering, Imperial College London, London, UK.
| |
Collapse
|
157
|
de Miguel-Gómez L, Romeu M, Pellicer A, Cervelló I. Strategies for managing asherman's syndrome and endometrial atrophy: Since the classical experimental models to the new bioengineering approach. Mol Reprod Dev 2021; 88:527-543. [PMID: 34293229 DOI: 10.1002/mrd.23523] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/11/2021] [Accepted: 06/30/2021] [Indexed: 11/07/2022]
Abstract
Endometrial function is essential for embryo implantation and pregnancy, but managing endometrial thickness that is too thin to support pregnancy or an endometrium of compromised functionality due to intrauterine adhesions is an ongoing challenge in reproductive medicine. Here, we review current and emerging therapeutic and experimental options for endometrial regeneration with a focus on animal models used to study solutions for Asherman's syndrome and endometrial atrophy, which both involve a damaged endometrium. A review of existing literature was performed that confirmed the lack of consensus on endometrial therapeutic options, though promising new alternatives have emerged in recent years (platelet-rich plasma, exosomes derived from stem cells, bioengineering-based techniques, endometrial organoids, among others). In the future, basic research using established experimental models of endometrial pathologies (combined with new high-tech solutions) and human clinical trials with large population sizes are needed to evaluate these emerging and new endometrial therapies.
Collapse
Affiliation(s)
- Lucía de Miguel-Gómez
- Fundación Instituto Valenciano de Infertilidad (FIVI), La Fe Health Research Institute, Valencia, Spain
- University of Valencia, Valencia, Spain
| | - Mónica Romeu
- Reproductive Medicine Research Group, La Fe Health Research Institute, La Fe University Hospital, Valencia, Spain
- Women's Health Area, Human Reproduction Unit, La Fe University Hospital, Valencia, Spain
| | | | - Irene Cervelló
- Fundación Instituto Valenciano de Infertilidad (FIVI), La Fe Health Research Institute, Valencia, Spain
| |
Collapse
|
158
|
Kraus L, Bryan C, Wagner M, Kino T, Gunchenko M, Jalal W, Khan M, Mohsin S. Bmi1 Augments Proliferation and Survival of Cortical Bone-Derived Stem Cells after Injury through Novel Epigenetic Signaling via Histone 3 Regulation. Int J Mol Sci 2021; 22:7813. [PMID: 34360579 PMCID: PMC8345961 DOI: 10.3390/ijms22157813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 01/18/2023] Open
Abstract
Ischemic heart disease can lead to myocardial infarction (MI), a major cause of morbidity and mortality worldwide. Multiple stem cell types have been safely transferred into failing human hearts, but the overall clinical cardiovascular benefits have been modest. Therefore, there is a dire need to understand the basic biology of stem cells to enhance therapeutic effects. Bmi1 is part of the polycomb repressive complex 1 (PRC1) that is involved in different processes including proliferation, survival and differentiation of stem cells. We isolated cortical bones stem cells (CBSCs) from bone stroma, and they express significantly high levels of Bmi1 compared to mesenchymal stem cells (MSCs) and cardiac-derived stem cells (CDCs). Using lentiviral transduction, Bmi1 was knocked down in the CBSCs to determine the effect of loss of Bmi1 on proliferation and survival potential with or without Bmi1 in CBSCs. Our data show that with the loss of Bmi1, there is a decrease in CBSC ability to proliferate and survive during stress. This loss of functionality is attributed to changes in histone modification, specifically histone 3 lysine 27 (H3K27). Without the proper epigenetic regulation, due to the loss of the polycomb protein in CBSCs, there is a significant decrease in cell cycle proteins, including Cyclin B, E2F, and WEE as well as an increase in DNA damage genes, including ataxia-telangiectasia mutated (ATM) and ATM and Rad3-related (ATR). In conclusion, in the absence of Bmi1, CBSCs lose their proliferative potential, have increased DNA damage and apoptosis, and more cell cycle arrest due to changes in epigenetic modifications. Consequently, Bmi1 plays a critical role in stem cell proliferation and survival through cell cycle regulation, specifically in the CBSCs. This regulation is associated with the histone modification and regulation of Bmi1, therefore indicating a novel mechanism of Bmi1 and the epigenetic regulation of stem cells.
Collapse
Affiliation(s)
- Lindsay Kraus
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (L.K.); (C.B.); (M.W.); (T.K.); (M.G.); (W.J.)
| | - Chris Bryan
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (L.K.); (C.B.); (M.W.); (T.K.); (M.G.); (W.J.)
| | - Marcus Wagner
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (L.K.); (C.B.); (M.W.); (T.K.); (M.G.); (W.J.)
| | - Tabito Kino
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (L.K.); (C.B.); (M.W.); (T.K.); (M.G.); (W.J.)
| | - Melissa Gunchenko
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (L.K.); (C.B.); (M.W.); (T.K.); (M.G.); (W.J.)
| | - Wassy Jalal
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (L.K.); (C.B.); (M.W.); (T.K.); (M.G.); (W.J.)
| | - Mohsin Khan
- Center for Metabolic Diseases, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA;
| | - Sadia Mohsin
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (L.K.); (C.B.); (M.W.); (T.K.); (M.G.); (W.J.)
| |
Collapse
|
159
|
Razavi M, Rezaee M, Telichko A, Inan H, Dahl J, Demirci U, Thakor AS. The Paracrine Function of Mesenchymal Stem Cells in Response to Pulsed Focused Ultrasound. Cell Transplant 2021; 29:963689720965478. [PMID: 33028105 PMCID: PMC7784560 DOI: 10.1177/0963689720965478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We studied the paracrine function of mesenchymal stem cells (MSCs) derived from various sources in response to pulsed focused ultrasound (pFUS). Human adipose tissue (AD), bone marrow (BM), and umbilical cord (UC) derived MSCs were exposed to pFUS at two intensities: 0.45 W/cm2 ISATA (310 kPa PNP) and 1.3 W/cm2 ISATA (540 kPa PNP). Following pFUS, the viability and proliferation of MSCs were assessed using a hemocytometer and confocal microscopy, and their secreted cytokine profile determined using a multiplex ELISA. Our findings showed that pFUS can stimulate the production of immunomodulatory, anti-inflammatory, and angiogenic cytokines from MSCs which was dependent on both the source of MSC being studied and the acoustic intensity employed. These important findings set the foundation for additional mechanistic and validation studies using this novel noninvasive and clinically translatable technology for modulating MSC biology.
Collapse
Affiliation(s)
- Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA.,BiionixTM (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, 6243University of Central Florida, Orlando, FL, USA.,Department of Materials Science and Engineering, 6243University of Central Florida, Orlando, FL, USA
| | - Melika Rezaee
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Arsenii Telichko
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Hakan Inan
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Jeremy Dahl
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Utkan Demirci
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| |
Collapse
|
160
|
Nagano H, Suematsu Y, Takuma M, Aoki S, Satoh A, Takayama E, Kinoshita M, Morimoto Y, Takeoka S, Fujie T, Kiyosawa T. Enhanced cellular engraftment of adipose-derived mesenchymal stem cell spheroids by using nanosheets as scaffolds. Sci Rep 2021; 11:14500. [PMID: 34262089 PMCID: PMC8280158 DOI: 10.1038/s41598-021-93642-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 06/28/2021] [Indexed: 02/07/2023] Open
Abstract
The short survival time of transplanted adipose-derived mesenchymal stem cells (ASCs) is a problem for skin wound healing. Transplantation after the formation of cellular spheroids has been investigated as a promising method for prolonging cellular survival. However, there have been technical restrictions for transplantation of spheroids in clinical practice. Here, we show an effective method for transplantation of ASC spheroids onto skin wounds in order to efficiently cure refractory ulcers. To assist anchoring of spheroids onto skin wounds, we used a 120-nm-thick free-standing film (nanosheet) that has a highly adhesive property. Bioluminescence imaging showed that ASC spheroids carried by the nanosheet survived for 14 days, which is about two-times longer than that previously reported. Wounds treated with a nanosheet carrying ASC spheroids were 4-times smaller than untreated wounds on day 14. This method for transplantation of spheroids could be applied to cell therapy for various refractory skin wounds.
Collapse
Affiliation(s)
- Hisato Nagano
- Department of Plastic and Reconstructive Surgery, National Defense Medical College, Tokorozawa, Saitama, 359-8513, Japan
| | - Yoshitaka Suematsu
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 162-8480, Japan
| | - Megumi Takuma
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, 226-8501, Japan
| | - Shimpo Aoki
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Ayano Satoh
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-0082, Japan
| | - Eiji Takayama
- Department of Oral Biochemistry, Asahi University School of Dentistry, Gifu, 501-0296, Japan
| | - Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Saitama, 359-8513, Japan
| | - Yuji Morimoto
- Department of Physiology, National Defense Medical College, Tokorozawa, Saitama, 359-8513, Japan
| | - Shinji Takeoka
- Institute for Advanced Research of Biosystem Dynamics, Research Institute for Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Toshinori Fujie
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, 226-8501, Japan
| | - Tomoharu Kiyosawa
- Department of Plastic and Reconstructive Surgery, National Defense Medical College, Tokorozawa, Saitama, 359-8513, Japan.
| |
Collapse
|
161
|
Beerts C, Brondeel C, Pauwelyn G, Depuydt E, Tack L, Duchateau L, Xu Y, Saunders JH, Peremans K, Spaas JH. Scintigraphic tracking of 99mTechnetium-labelled equine peripheral blood-derived mesenchymal stem cells after intravenous, intramuscular, and subcutaneous injection in healthy dogs. Stem Cell Res Ther 2021; 12:393. [PMID: 34256833 PMCID: PMC8278733 DOI: 10.1186/s13287-021-02457-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 06/14/2021] [Indexed: 01/02/2023] Open
Abstract
Background Mesenchymal stem cell treatments in dogs have been investigated as a potential innovative alternative to current conventional therapies for a variety of conditions. So far, the precise mode of action of the MSCs has yet to be determined. The aim of this study was to gain more insights into the pharmacokinetics of MSCs by evaluating their biodistribution in healthy dogs after different injection routes. Methods Three different studies were performed in healthy dogs to evaluate the biodistribution pattern of radiolabelled equine peripheral blood-derived mesenchymal stem cells following intravenous, intramuscular and subcutaneous administration in comparison with free 99mTechnetium. The labelling of the equine peripheral blood-derived mesenchymal stem cells was performed using stannous chloride as a reducing agent. Whole-body scans were obtained using a gamma camera during a 24-h follow-up. Results The labelling efficiency ranged between 59.58 and 83.82%. Free 99mTechnetium accumulation was predominantly observed in the stomach, thyroid, bladder and salivary glands, while following intravenous injection, the 99mTechnetium-labelled equine peripheral blood-derived mesenchymal stem cells majorly accumulated in the liver throughout the follow-up period. After intramuscular and subcutaneous injection, the injected dose percentage remained very high at the injection site. Conclusions A distinct difference was noted in the biodistribution pattern of the radiolabelled equine peripheral blood-derived mesenchymal stem cells compared to free 99mTechnetium indicating equine peripheral blood-derived mesenchymal stem cells have a specific pharmacokinetic pattern after systemic administration in healthy dogs. Furthermore, the biodistribution pattern of the used xenogeneic equine peripheral blood-derived mesenchymal stem cells appeared to be different from previously reported experiments using different sources of mesenchymal stem cells.
Collapse
Affiliation(s)
- Charlotte Beerts
- Global Stem cell Technology NV, Noorwegenstraat 4, 9940, Evergem, Belgium.,Department of Medical Imaging and Orthopedics of Domestic Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Carlien Brondeel
- Department of Medical Imaging and Orthopedics of Domestic Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Glenn Pauwelyn
- Global Stem cell Technology NV, Noorwegenstraat 4, 9940, Evergem, Belgium
| | - Eva Depuydt
- Global Stem cell Technology NV, Noorwegenstraat 4, 9940, Evergem, Belgium
| | - Liesa Tack
- Global Stem cell Technology NV, Noorwegenstraat 4, 9940, Evergem, Belgium
| | - Luc Duchateau
- Biometrics Research Center, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Yangfeng Xu
- Department of Medical Imaging and Orthopedics of Domestic Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Jimmy H Saunders
- Department of Medical Imaging and Orthopedics of Domestic Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Kathelijne Peremans
- Department of Medical Imaging and Orthopedics of Domestic Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Jan H Spaas
- Global Stem cell Technology NV, Noorwegenstraat 4, 9940, Evergem, Belgium. .,Department of Medical Imaging and Orthopedics of Domestic Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium.
| |
Collapse
|
162
|
Chang D, Fan T, Gao S, Jin Y, Zhang M, Ono M. Application of mesenchymal stem cell sheet to treatment of ischemic heart disease. Stem Cell Res Ther 2021; 12:384. [PMID: 34233729 PMCID: PMC8261909 DOI: 10.1186/s13287-021-02451-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/07/2021] [Indexed: 12/29/2022] Open
Abstract
In recent years, mesenchymal stem cells (MSCs) have been used to improve cardiac function and attenuate adverse ventricular remodeling of the ischemic myocardium through paracrine effects and immunoregulation functions. In combination with cell sheet technology, MSCs could be more easily transplanted to the ischemic area. The long-term retention of MSCs in the affected area was realized and significantly improved the curative effect. In this review, we summarized the research and the applications of MSC sheets to the treatment of ischemic heart tissue. At present, many types of MSCs have been considered as multipotent cells in the treatment of heart failure, such as bone marrow-derived mesenchymal stem cells (BM-MSCs), adipose-derived mesenchymal stem cells (AD-MSCs), umbilical cord-derived mesenchymal stem cells (UC-MSCs), and skeletal myoblasts (SMs). Since UC-MSCs have few human leukocyte antigen-II and major histocompatibility complex class I molecules, and are easy to isolate and culture, UC-MSC sheets have been proposed as a candidate for clinical applications to ischemic heart disease.
Collapse
Affiliation(s)
- Dehua Chang
- Department of Cell Therapy in Regenerative Medicine, The University of Tokyo Hospital, 7-3-1 Honggo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Taibing Fan
- Children Heart Center, Fuwai Central China Cardiovascular Hospital, No.1 Fuwai Road, Zhengzhou, 450018, China
| | - Shuang Gao
- Research and Development Department, BOE Regenerative Medicine Technology Co., Ltd., NO.9 JiuXianQiao North Road, Beijing, 100015, China
| | - Yongqiang Jin
- Heart Center, First Hospital of Tsinghua University, NO.6 JiuXianQiao 1st Road, Beijing, 10016, China
| | - Mingkui Zhang
- Heart Center, First Hospital of Tsinghua University, NO.6 JiuXianQiao 1st Road, Beijing, 10016, China
| | - Minoru Ono
- Department of Cardiac Surgery, The University of Tokyo Hospital, 7-3-1 Honggo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
163
|
Pathipati P, Lecuyer M, Faustino J, Strivelli J, Phinney DG, Vexler ZS. Mesenchymal Stem Cell (MSC)-Derived Extracellular Vesicles Protect from Neonatal Stroke by Interacting with Microglial Cells. Neurotherapeutics 2021; 18:1939-1952. [PMID: 34235636 PMCID: PMC8609070 DOI: 10.1007/s13311-021-01076-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2021] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem cell (MSC)-based therapies are beneficial in models of perinatal stroke and hypoxia-ischemia. Mounting evidence suggests that in adult injury models, including stroke, MSC-derived small extracellular vesicles (MSC-sEV) contribute to the neuroprotective and regenerative effects of MSCs. Herein, we examined if MSC-sEV protect neonatal brain from stroke and if this effect is mediated via communication with microglia. MSC-sEV derived from bone marrow MSCs were characterized by size distribution (NanoSight™) and identity (protein markers). Studies in microglial cells isolated from the injured or contralateral cortex of postnatal day 9 (P9) mice subjected to a 3-h middle cerebral artery occlusion (tMCAO) and cultured (in vitro) revealed that uptake of fluorescently labeled MSC-sEV was significantly greater by microglia from the injured cortex vs. contralateral cortex. The cell-type-specific spatiotemporal distribution of MSC-sEV was also determined in vivo after tMCAO at P9. MSC-sEV administered at reperfusion, either by intracerebroventricular (ICV) or by intranasal (IN) routes, accumulated in the hemisphere ipsilateral to the occlusion, with differing spatial distribution 2 h, 18 h, and 72 h regardless of the administration route. By 72 h, MSC-sEV in the IN group was predominantly observed in Iba1+ cells with retracted processes and in GLUT1+ blood vessels in ischemic-reperfused regions. MSC-sEV presence in Iba1+ cells was sustained. MSC-sEV administration also significantly reduced injury volume 72 h after tMCAO in part via modulatory effects on microglial cells. Together, these data establish feasibility for MSC-sEV delivery to injured neonatal brain via a clinically relevant IN route, which affords protection during sub-acute injury phase.
Collapse
Affiliation(s)
- Praneeti Pathipati
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Matthieu Lecuyer
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Joel Faustino
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | | | - Donald G Phinney
- Department of Molecular Medicine, Scripps Research Institute, Jupiter, FL, USA
| | - Zinaida S Vexler
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
164
|
Fractionating stem cells secretome for Parkinson's disease modeling: Is it the whole better than the sum of its parts? Biochimie 2021; 189:87-98. [PMID: 34182001 DOI: 10.1016/j.biochi.2021.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 01/08/2023]
Abstract
Human mesenchymal stem cells (hMSCs) secretome has been have been at the forefront of a new wave of possible therapeutic strategies for central nervous system neurodegenerative disorders, as Parkinson's disease (PD). While within its protein fraction, several promising proteins were already identified with therapeutic properties on PD, the potential of hMSCs-secretome vesicular fraction remains to be elucidated. Such highlighting is important, since hMSCs secretome-derived vesicles can act as biological nanoparticles with beneficial effects in different pathological contexts. Therefore, in this work, we have isolated hMSCs secretome vesicular fraction, and assessed their impact on neuronal survival, and differentiation on human neural progenitors' cells (hNPCs), and in a 6-hydroxydopamine (6-OHDA) rat model of PD when compared to hMSCs secretome (as a whole) and its protein derived fraction. From the results, we have found hMSCs vesicular fraction as polydispersity source of vesicles, which when applied in vitro was able to induce hNPCs differentiation at the same levels as the whole secretome, while the protein separated fraction was not able to induce such effect. In the context of PD, although distinct effects were observed, hMSCs secretome and its derived fractions displayed a positive impact on animals' motor and histological performance, thereby indicating that hMSCs secretome and its different fractions may impact different mechanisms and pathways. Overall, we concluded that the use of the secretome collected from hMSCs and its different fractions might be active modulators of different neuroregeneration mechanisms, which could open new therapeutical opportunities for their future use as a treatment for PD.
Collapse
|
165
|
Nguyen-Truong M, Hematti P, Wang Z. Current status of myocardial restoration via the paracrine function of mesenchymal stromal cells. Am J Physiol Heart Circ Physiol 2021; 321:H112-H127. [PMID: 34085844 DOI: 10.1152/ajpheart.00217.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mesenchymal stromal cells (MSCs) have been studied for nearly two decades as a therapy for myocardial restoration. An emerging direction to repair myocardium is through their paracrine function, which includes the utilization of MSC-derived conditioned medium or extracellular vesicles. In this review, we go over the unique characteristics of MSCs that make it suitable for "off the shelf," cell-free regenerative therapy, current MSC-derived cell-free approaches including their advantages and disadvantages, and the known mechanisms of action of the paracrine effect of MSCs. With a summary of the clinical trials and preclinical studies of MSC-derived cell-free therapy, we classify the aforementioned mechanisms into angiogenesis, immunomodulation, extracellular matrix remodeling, antiapoptosis, and antioxidation. Particularly, we discuss on ways researchers have worked toward enhancing these desired properties to improve the therapeutic outcomes and the investigation of mechanobiology involved in MSC paracrine function. Lastly, we bring up the remaining challenges in this arising field and suggestions for future directions to improve our understanding and control over the potential of MSC paracrine function for myocardial restoration.
Collapse
Affiliation(s)
| | - Peiman Hematti
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
166
|
Yeganeh A, Alibhai FJ, Tobin SW, Lim F, Wu J, Li S, Weisel RD, Li RK. Age-related defects in autophagy alter the secretion of paracrine factors from bone marrow mononuclear cells. Aging (Albany NY) 2021; 13:14687-14708. [PMID: 34088884 PMCID: PMC8221303 DOI: 10.18632/aging.203127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/14/2021] [Indexed: 01/08/2023]
Abstract
Bone marrow mononuclear cell therapy improves cardiac repair after myocardial infarction (MI), in-part through signaling to resident cardiac cells, such as fibroblasts, which regulate scar formation. The efficacy of cell therapy declines with age, as aging of both donor and recipient cells decreases repair responses. Autophagy regulates the microenvironment by both extracellular vesicle (EV)-dependent and independent secretion pathways. We hypothesized that age-related autophagy changes in bone marrow cells (BMCs) alter paracrine signaling, contributing to lower cell therapy efficacy. Here, we demonstrate that young Sca-1+ BMCs exhibited a higher LC3II/LC3I ratio compared to old Sca-1+ BMCs, which was accentuated when BMCs were cultured under hypoxia. To examine the effect on paracrine signaling, old cardiac fibroblasts were cultured with conditioned medium (CM) from young and old Sca-1+ BMCs. Young, but not old CM, enhanced fibroblast proliferation, migration, and differentiation, plus reduced senescence. These beneficial effects were lost when autophagy or EV secretion in BMCs was blocked pharmacologically, or by siRNA knockdown of Atg7. Therefore, both EV-dependent and -independent paracrine signaling from young BMCs is responsible for paracrine stimulation of old cardiac fibroblasts. In vivo, bone marrow chimerism of old mice with young BMCs increased the number of LC3b+ cells in the heart compared to old mice reconstituted with old BMCs. These data suggest that the deterioration of autophagy with aging negatively impacts the paracrine effects of BMCs, and provide mechanistic insight into the age-related decline in cell therapy efficacy that could be targeted to improve the function of old donor cells.
Collapse
Affiliation(s)
- Azadeh Yeganeh
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Faisal J. Alibhai
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Stephanie W. Tobin
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Fievel Lim
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Jun Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Shuhong Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Richard D. Weisel
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, Canada
| | - Ren-Ke Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
167
|
Kukumberg M, Zaw AM, Wong DHC, Toh CM, Chan BPL, Seet RCS, Wong PTH, Yim EKF. Characterization and Functional Assessment of Endothelial Progenitor Cells in Ischemic Stroke Patients. Stem Cell Rev Rep 2021; 17:952-967. [PMID: 33170433 PMCID: PMC7653671 DOI: 10.1007/s12015-020-10064-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 11/09/2022]
Abstract
Endothelial dysfunction has been implicated in atherosclerosis, ischemic heart disease, and stroke. Endothelial progenitor cells (EPCs), found in the bone marrow and peripheral blood as rare cell population, demonstrated a high proliferation and differentiation capacity. Understanding how such diseases influence the quantity and functionality of EPCs is essential for the development of novel therapies. This study aims to investigate the factors that affect the quantity and functionality of circulating EPCs in stroke patients and healthy controls. Blood samples were collected once from healthy donors (n = 30) and up to 3 times (within 7 days (baseline), 3 and 12 months post-stroke) from stroke patients (n = 207). EPC subpopulations were isolated with flow cytometry for characterization. The Matrigel tubular formation assay was performed as a measure of functionality. An increased amount of circulating EPCs was observed in stroke patients over 45 years when compared to age-matched healthy individuals. EPCs showed a rising trend in stroke patients over the 12-month post-stroke period, reaching statistical significance at 12 months post-stroke. Isolated CD34+KDR+ cells from stroke patients showed impairment in tubular formation capability when compared to cells from healthy donors. The quantity and vasculogenic function of circulating EPCs in peripheral blood have been effectively evaluated in stroke patients and healthy control donors in this study. Age and stroke are found to be 2 influencing factors on the angiogenic capacity. It is suggested that the increase in EPC number is triggered by the recovery response following ischemic stroke. Graphical abstract.
Collapse
Affiliation(s)
- Marek Kukumberg
- Mechanobiology Institute, National University of Singapore, #05-01 T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Aung Moe Zaw
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Daniel H C Wong
- Mechanobiology Institute, National University of Singapore, #05-01 T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16, Medical Drive, #04-01, Singapore, 117600, Singapore
| | - Chin Min Toh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16, Medical Drive, #04-01, Singapore, 117600, Singapore
| | - Bernard P L Chan
- Division of Neurology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Raymond C S Seet
- Division of Neurology, Department of Medicine, National University Hospital, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Level 10, NUHS Tower Block, 1E Kent Ride Road, Singapore, 119228, Singapore
| | - Peter T H Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16, Medical Drive, #04-01, Singapore, 117600, Singapore
| | - Evelyn K F Yim
- Mechanobiology Institute, National University of Singapore, #05-01 T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada.
- Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada.
- Centre for Biotechnology and Bioengineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada.
| |
Collapse
|
168
|
Human umbilical cord mesenchymal stem cells in type 2 diabetes mellitus: the emerging therapeutic approach. Cell Tissue Res 2021; 385:497-518. [PMID: 34050823 DOI: 10.1007/s00441-021-03461-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/11/2021] [Indexed: 12/14/2022]
Abstract
The umbilical cord has been proved to be an easy-access, reliable, and useful source of mesenchymal stem cells (MSC) for clinical applications due to its primitive, immunomodulatory, non-immunogenic, secretory and paracrine, migratory, proliferative, and multipotent properties. This set of characteristics has recently attracted great research interest in the fields of nanotechnology and regenerative medicine and cellular therapy. Accumulating evidence supports a pronounced therapeutic potential of MSC in many different pathologies, from hematology to immunology, wound-healing, tissue regeneration, and oncology. Diabetes mellitus, branded the epidemic of the century, is considered a chronic metabolic disorder, representing a major burden for health system sustainability and an important public health challenge to modern societies. The available treatments for type 2 diabetes mellitus (T2DM) still rely mainly on combinations of oral antidiabetic agents with lifestyle and nutritional adjustments. Despite the continuous development of novel and better hypoglycemic drugs, their efficacy is limited in the installment and progression of silent T2DM complications. T2DM comorbidities and mortality rates still make it a serious, common, costly, and long-term manageable disease. Recently, experimental models, preclinical observations, and clinical studies have provided some insights and preliminary promising results using umbilical cord MSCs to treat and manage diabetes. This review focuses on the latest research and applications of human-derived umbilical cord MSC in the treatment and management of T2DM, exploring and systematizing the key effects of both umbilical cord MSC and its factor-rich secretome accordingly with the major complications associated to T2DM.
Collapse
|
169
|
Hwang JW, Myeong SH, Lee NH, Kim H, Son HJ, Chang JW, Lee NK, Na DL. Immunosuppressant Drugs Mitigate Immune Responses Generated by Human Mesenchymal Stem Cells Transplanted into the Mouse Parenchyma. Cell Transplant 2021; 30:9636897211019025. [PMID: 34044601 PMCID: PMC8168027 DOI: 10.1177/09636897211019025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
It has been widely accepted that mesenchymal stem cells (MSCs) can evade the immune surveillance of the recipient. However, emerging research cast doubt on whether MSCs are intrinsically immune-privileged. Previously, we observed that the transplantation of human MSCs (hMSCs) into the mouse parenchyma attracted a high infiltration of leukocytes into the injection tract. Thus, in order to reduce the immune responses generated by hMSCs, the aim of this study was to assess which immunosuppressant condition (dexamethasone only, tacrolimus only, or dexamethasone and tacrolimus together) would not only reduce the overall immune response but also enhance the persistence of MSCs engrafted into the caudate putamen of wild-type C57BL/6 mice. According to immunohistochemical analysis, compared to the hMSC only group, the administration of immunosuppressants (for all three conditions) reduced the infiltration of CD45-positive leukocytes and neutrophils at the site of injection. The highest hMSC persistence was detected from the group that received combinatorial administrations of dexamethasone and tacrolimus. Moreover, compared to the immunocompetent WT mouse, higher MSC engraftment was observed from the immunodeficient BALB/c mice. The results of this study support the use of immunosuppressants to tackle MSC-mediated immune responses and to possibly prolong the engraftment of transplanted MSCs.
Collapse
Affiliation(s)
- Jung Won Hwang
- Department of Health Sciences and Technology, SAIHST, 35019Sungkyunkwan University, Gangnam-gu, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Su Hyeon Myeong
- Department of Health Sciences and Technology, SAIHST, 35019Sungkyunkwan University, Gangnam-gu, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Na-Hee Lee
- Department of Health Sciences and Technology, SAIHST, 35019Sungkyunkwan University, Gangnam-gu, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, 36626Samsung Medical Center, Seoul, Republic of Korea
| | - Hyeongseop Kim
- Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Stem Cell Institute, ENCell Co. Ltd., Seoul, Republic of Korea
| | - Hyo Jin Son
- Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, 36626Samsung Medical Center, Seoul, Republic of Korea.,School of Medicine, 35019Sungkyunkwan University, Seoul, Republic of Korea
| | - Jong Wook Chang
- Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Stem Cell Institute, ENCell Co. Ltd., Seoul, Republic of Korea
| | - Na Kyung Lee
- Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, 36626Samsung Medical Center, Seoul, Republic of Korea.,School of Medicine, 35019Sungkyunkwan University, Seoul, Republic of Korea
| | - Duk L Na
- Department of Health Sciences and Technology, SAIHST, 35019Sungkyunkwan University, Gangnam-gu, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, 36626Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Samsung Medical Center, 35019Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, 36626Samsung Medical Center, Seoul, Republic of Korea.,School of Medicine, 35019Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
170
|
Kim JY, Rhim WK, Seo HJ, Lee JY, Park CG, Han DK. Comparative Analysis of MSC-Derived Exosomes Depending on Cell Culture Media for Regenerative Bioactivity. Tissue Eng Regen Med 2021; 18:355-367. [PMID: 34047999 DOI: 10.1007/s13770-021-00352-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In order to produce and isolate the exosome derived from the cell of interests, a serum free environment (starvation) has been essential for excluding the unknown effect from serum-derived exosomes. Recently, serum-free culture media have been developed as a substitute for serum supplemented media so that MSC proliferates with maintaining the original characteristics of the cells in a serum free condition. Due to the different properties of the exosomes representing the states and characteristics of the origin cells, a study is needed to compare the properties of the cell-derived exosomes according to the cell culture media. METHODS To compare the cell culture condition on exosomes, human umbilical cord mesenchymal stem cells (UCMSCs) were cultured with two different media, serum containing media, 10% FBS supplemented DMEM (NM) and serum-free chemically defined media, CellCor™ CD MSC (CDM). To remove FBS-derived exosomes from UCMSC cultured with NM, the medium was replaced with FBS-free DMEM for starvation during exosome isolation. The production yield and expression levels of angiogenic and pro-inflammatory factors were compared. And, the subpopulations of exosome were classified depending on the surface properties and loaded cytokines. Finally, the wound healing and angiogenic effects have been evaluated using in vitro assays. RESULTS The UCMSC-derived exosomes under two different cell culture media could be classified into subpopulations according to the surface composition and loaded cytokines. Especially, exosome derived from UCMSC cultured with CDM showed higher expression levels of cytokines related to regenerative bioactivities which resulted in enhanced wound healing and angiogenesis. CONCLUSION CDM has the advantages to maintain cell proliferation even during the period of exosome isolations and eliminate unknown side effects caused by serum-derived exosomes. Additionally, exosomes derived from UCMSC cultured with CDM show better wound healing and angiogenic effects due to a lot of regeneration-related cytokines and less pro-inflammatory cytokines compared to with NM.
Collapse
Affiliation(s)
- Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.,Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea.,ntelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hyo Jeong Seo
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Joo Youn Lee
- Xcell Therapeutics, Hanhwa Biz metro Building, 242 Digital-ro, Guro-gu, Seoul, 08394, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea.,ntelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
171
|
Zhao Y, Shi Y, Lin H. Hypoxia Promotes Adipose-Derived Stem Cells to Protect Human Dermal Microvascular Endothelial Cells Against Hypoxia/Reoxygenation Injury. J Surg Res 2021; 266:230-235. [PMID: 34023579 DOI: 10.1016/j.jss.2021.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 02/25/2021] [Accepted: 04/10/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Microcirculation is important for regulating ischemia-reperfusion (I/R) injury associated with skin flap transplantation surgery. We investigated whether co-culture with adipose-derived stem cells (ADSCs) could protect human dermal microvascular endothelial cells (HDMECs) from I/R injury by inhibiting cell apoptosis and enhancing cell proliferation. We also investigated the effects of hypoxic preconditioning on ADSCs. MATERIALS AND METHODS HDMECs were divided into four groups, control, HDMECs in normoxic culture conditions; hypoxia/reoxygenation (H/R), HDMECs in a hypoxic incubator for 8 h then in saturated aerobic culture medium for 24 h; H/R + ADSC(N), HDMECs treated similar to the H/R group then co-cultured with normoxic ADSCs; and H/R + ADSC(H), HDMECs treated similar to the H/R group then co-cultured with hypoxia preconditioned ADSCs. RESULTS The rate of HDMECs apoptosis significantly increased in the H/R group, but decreased upon co-culture with ADSCs for 24 h, especially in the H/R + ADSC(H) group. Co-culture with ADSCs, especially hypoxia preconditioned ADSCs, significantly enhanced cell proliferation ability compared with that of the H/R group after 48 h and 72 h, but not after 24 h. Vascular endothelial growth factor levels were significantly higher in the H/R + ADSC(N) and H/R + ADSC(H) groups than in the H/R group. CONCLUSIONS ADSCs attenuated H/R injury in endothelial cells by promoting proliferation ability and reducing apoptosis, with an increase in Vascular endothelial growth factor level, especially in the context of hypoxic preconditioning. This approach suggests the potential for an easy and safe method to reduce I/R injury associated with skin flap transplantation surgery.
Collapse
Affiliation(s)
- Yinhua Zhao
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China, 100029
| | - Yanyu Shi
- Plastic and Reconstructive Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Huang Lin
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China, 100029.
| |
Collapse
|
172
|
van der Pol A, Bouten CVC. A Brief History in Cardiac Regeneration, and How the Extra Cellular Matrix May Turn the Tide. Front Cardiovasc Med 2021; 8:682342. [PMID: 34095264 PMCID: PMC8172606 DOI: 10.3389/fcvm.2021.682342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Abstract
Tissue homeostasis is perturbed by stressful events, which can lead to organ dysfunction and failure. This is particularly true for the heart, where injury resulting from myocardial infarction or ischemic heart disease can result in a cascading event ultimately ending with the loss of functional myocardial tissue and heart failure. To help reverse this loss of healthy contractile tissue, researchers have spent decades in the hopes of characterizing a cell source capable of regenerating the injured heart. Unfortunately, these strategies have proven to be ineffective. With the goal of truly understanding cardiac regeneration, researchers have focused on the innate regenerative abilities of zebrafish and neonatal mammals. This has led to the realization that although cells play an important role in the repair of the diseased myocardium, inducing cardiac regeneration may instead lie in the composition of the extra cellular milieu, specifically the extra cellular matrix. In this review we will briefly summarize the current knowledge regarding cell sources used for cardiac regenerative approaches, since these have been extensively reviewed elsewhere. More importantly, by revisiting innate cardiac regeneration observed in zebrafish and neonatal mammals, we will stress the importance the extra cellular matrix has on reactivating this potential in the adult myocardium. Finally, we will address how we can harness the ability of the extra cellular matrix to guide cardiac repair thereby setting the stage of next generation regenerative strategies.
Collapse
Affiliation(s)
- Atze van der Pol
- Eindhoven University of Technology, Department of Biomedical Engineering, Eindhoven, Netherlands
| | | |
Collapse
|
173
|
Leong WMS, Samuel M, Chandran NS, Choi CEE, Yiong Huak C, Marsden H, Tam WWS. Interventions for inherited forms of epidermolysis bullosa. Hippokratia 2021. [DOI: 10.1002/14651858.cd014223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Wai Mun Sean Leong
- Department of Dermatology; National University Hospital; Singapore Singapore
| | - Miny Samuel
- Research Support Unit; NUS Yong Loo Lin School of Medicine; Singapore Singapore
| | | | - Ci En Ellie Choi
- Internal Medicine; National University Hospital Singapore; Singapore Singapore
| | - Chan Yiong Huak
- Biostatistics Unit; Yong Loo Lin School of Medicine; Singapore Singapore
| | - Harriet Marsden
- c/o Cochrane Skin Group; The University of Nottingham; Nottingham UK
| | - Wilson Wai San Tam
- Alice Lee Center for Nursing Studies, Yong Loo Lin School of Medicine; National University of Singapore, National University Health System; Singapore Singapore
| |
Collapse
|
174
|
Gupta AK, Renaud HJ, Rapaport JA. Platelet-rich Plasma and Cell Therapy: The New Horizon in Hair Loss Treatment. Dermatol Clin 2021; 39:429-445. [PMID: 34053596 DOI: 10.1016/j.det.2021.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Current medicinal therapies for treating hair loss have shortcomes due to variability and ineffectiveness, noncompliance, and adverse effects. The prevalence of hair loss and its associated negative psychological impact have driven research into regenerative medicine approaches, such as platelet-rich plasma (PRP) and cell-based therapies, in an attempt to find alternative, safe, effective, and reproducible treatments. Current research shows promising results from these therapies; however, more robust trials are needed to confirm the reported efficacies of PRP and cell-based therapies. Moreover, standardization of treatment preparation as well as dose and regimen are needed.
Collapse
Affiliation(s)
- Aditya K Gupta
- Division of Dermatology, Department of Medicine, University of Toronto School of Medicine, Toronto, Ontario, Canada; Mediprobe Research Inc., 645 Windermere Road, London, Ontario N5X 2P1, Canada.
| | - Helen J Renaud
- Mediprobe Research Inc., 645 Windermere Road, London, Ontario N5X 2P1, Canada
| | - Jeffrey A Rapaport
- Cosmetic Skin and Surgery Center, 333 Sylvan Avenue, Suite 207, Englewood Cliffs, NJ 07632, USA
| |
Collapse
|
175
|
Restorative Therapies for Erectile Dysfunction: Position Statement From the Sexual Medicine Society of North America (SMSNA). Sex Med 2021; 9:100343. [PMID: 34000480 PMCID: PMC8240368 DOI: 10.1016/j.esxm.2021.100343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Current non-invasive treatments for erectile dysfunction (ED) include oral medications, intracavernosal injections, and vacuum-assisted devices. Though these therapies work well for many, a subset of patients have contraindications or are unsatisfied with these options. Restorative therapies for ED are a new frontier of treatments focused on regenerating diseased tissue and providing a potential "cure" for ED. AIM The aim of this position statement is to examine existing clinical trial data for restorative therapies and identify elements that require further research before widespread adoption. METHODS A literature review was performed to identify all clinical trials performed with regenerative therapy for ED. This includes treatments such as stem cell therapy (SCT), platelet rich plasma (PRP), and restorative related technologies like low-intensity shockwave therapy (LiSWT). MAIN OUTCOME MEASURES Most clinical trials in restorative therapies were assessed for safety, feasibility, or efficacy. This included recording adverse events, changes in sexual function and erectile function questionnaires, and diagnostics measures. RESULTS To date there is an absence of robust clinical data supporting the efficacy of restorative therapies regarding ED, though technologies such as LiSWT have established relative safety. CONCLUSIONS Restorative therapies are a promising technology that represents a new frontier of treatment geared towards reversing disease pathology rather than just treating symptoms. However, current published clinical studies are limited. Future work needs to be adequately powered, multi-center, randomized, sham/placebo-controlled trials in well-characterized patient populations to ensure safety and demonstrate efficacy. Until these studies are done, restorative therapies should be reserved for clinical trials and not offered in routine clinical practice. Liu JL, Chu KY, Gabrielson AT, et al. Restorative Therapies for Erectile Dysfunction: Position Statement From the Sexual Medicine Society of North America (SMSNA). J Sex Med 2021;9:100343.
Collapse
|
176
|
Bose RJ, Ha K, McCarthy JR. Bio-inspired nanomaterials as novel options for the treatment of cardiovascular disease. Drug Discov Today 2021; 26:1200-1211. [PMID: 33561512 PMCID: PMC8205945 DOI: 10.1016/j.drudis.2021.01.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 11/28/2022]
Abstract
Cardiovascular disease (CVD) and its sequelae have long been the leading causes of death and disability in the developed world. Although mortality associated with CVD has been decreasing, due in large part to novel therapeutic options, the rate of decrease has flattened. Thus, there is a great need to investigate alternate therapeutic strategies that can increase efficacy while decreasing adverse effects. Nanomaterials have been widely investigated and have emerged as promising tools for both therapeutic and diagnostic purposes in oncology; however, the potential of nanomaterials has not been extensively explored for cardiovascular medicine. In this review, we focus on recent developments in the field of nanomedicines targeted for CVDs, with a special emphasis on cell membrane-coated nanoparticles (NPs) and their applications.
Collapse
Affiliation(s)
- Rajendran Jc Bose
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, USA
| | - Khan Ha
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, USA
| | - Jason R McCarthy
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, USA.
| |
Collapse
|
177
|
Kumar Mishra S, Khushu S, Gangenahalli G. Neuroprotective response and efficacy of intravenous administration of mesenchymal stem cells in traumatic brain injury mice. Eur J Neurosci 2021; 54:4392-4407. [PMID: 33932318 DOI: 10.1111/ejn.15261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022]
Abstract
Cellular transplantation of stem cells can be a beneficial treatment approach for neurodegenerative diseases such as traumatic brain injury (TBI). In this study, we investigated the proliferation and differentiation potential of infused mesenchymal stem cells (MSCs) after localisation at the injury site. We evaluated the appropriate homing of infused MSCs through immunohistochemistry, followed by Y-chromosome-specific polymerase chain reaction and fluorescent in situ hybridisation analyses. The proliferation and differentiation of infused MSCs were analysed using exogenous cell tracer 5'-bromo-2'-deoxyuridine (BrdU) labelling and neuronal specific markers, respectively. Structural and functional recovery in TBI mice were examined by performing magnetic resonance imaging and different behavioural assessments, respectively. Results demonstrated a significantly high number of BrdU-positive cells in the lesion region in the MSC-infused group compared with control and TBI groups. Infused MSCs were well differentiated into neural-like cells and expressed significantly more neural markers (neuronal nuclear antigen [NeuN], microtubule-associated protein 2 [MAP2] and glial fibrillary acid protein [GFAP]). Improved tissue abnormalities as well as functional behaviours were observed in MSC-infused TBI mice, implying the substantial proliferation and differentiation of infused MSCs. Our findings support the neuroprotective response and efficacy of MSCs after transplantation in TBI mice, and MSCs may serve as potential therapeutic candidates in regenerative medicine.
Collapse
Affiliation(s)
- Sushanta Kumar Mishra
- MRI Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Subash Khushu
- MRI Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Gurudutta Gangenahalli
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| |
Collapse
|
178
|
Yao C, Cao X, Yu B. Revascularization After Traumatic Spinal Cord Injury. Front Physiol 2021; 12:631500. [PMID: 33995118 PMCID: PMC8119644 DOI: 10.3389/fphys.2021.631500] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a complex pathological process. The initial mechanical damage is followed by a progressive secondary injury cascade. The injury ruptures the local microvasculature and disturbs blood-spinal cord barriers, exacerbating inflammation and tissue damage. Although endogenous angiogenesis is triggered, the new vessels are insufficient and often fail to function normally. Numerous blood vessel interventions, such as proangiogenic factor administration, gene modulation, cell transplantation, biomaterial implantation, and physical stimulation, have been applied as SCI treatments. Here, we briefly describe alterations and effects of the vascular system on local microenvironments after SCI. Therapies targeted at revascularization for SCI are also summarized.
Collapse
Affiliation(s)
- Chun Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xuemin Cao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
179
|
Salikhova D, Bukharova T, Cherkashova E, Namestnikova D, Leonov G, Nikitina M, Gubskiy I, Akopyan G, Elchaninov A, Midiber K, Bulatenco N, Mokrousova V, Makarov A, Yarygin K, Chekhonin V, Mikhaleva L, Fatkhudinov T, Goldshtein D. Therapeutic Effects of hiPSC-Derived Glial and Neuronal Progenitor Cells-Conditioned Medium in Experimental Ischemic Stroke in Rats. Int J Mol Sci 2021; 22:4694. [PMID: 33946667 PMCID: PMC8125106 DOI: 10.3390/ijms22094694] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/12/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Transplantation of various types of stem cells as a possible therapy for stroke has been tested for years, and the results are promising. Recent investigations have shown that the administration of the conditioned media obtained after stem cell cultivation can also be effective in the therapy of the central nervous system pathology (hypothesis of their paracrine action). The aim of this study was to evaluate the therapeutic effects of the conditioned medium of hiPSC-derived glial and neuronal progenitor cells in the rat middle cerebral artery occlusion model of the ischemic stroke. Secretory activity of the cultured neuronal and glial progenitor cells was evaluated by proteomic and immunosorbent-based approaches. Therapeutic effects were assessed by overall survival, neurologic deficit and infarct volume dynamics, as well as by the end-point values of the apoptosis- and inflammation-related gene expression levels, the extent of microglia/macrophage infiltration and the numbers of formed blood vessels in the affected area of the brain. As a result, 31% of the protein species discovered in glial progenitor cells-conditioned medium and 45% in neuronal progenitor cells-conditioned medium were cell type specific. The glial progenitor cell-conditioned media showed a higher content of neurotrophins (BDNF, GDNF, CNTF and NGF). We showed that intra-arterial administration of glial progenitor cells-conditioned medium promoted a faster decrease in neurological deficit compared to the control group, reduced microglia/macrophage infiltration, reduced expression of pro-apoptotic gene Bax and pro-inflammatory cytokine gene Tnf, increased expression of anti-inflammatory cytokine genes (Il4, Il10, Il13) and promoted the formation of blood vessels within the damaged area. None of these effects were exerted by the neuronal progenitor cell-conditioned media. The results indicate pronounced cytoprotective, anti-inflammatory and angiogenic properties of soluble factors secreted by glial progenitor cells.
Collapse
Affiliation(s)
- Diana Salikhova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (T.B.); (G.L.); (N.B.); (V.M.); (D.G.)
- Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (A.E.); (K.M.); (L.M.); (T.F.)
| | - Tatiana Bukharova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (T.B.); (G.L.); (N.B.); (V.M.); (D.G.)
| | - Elvira Cherkashova
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (E.C.); (D.N.); (I.G.); (A.M.); (V.C.)
- Radiology and Clinical Physiology Scientific Research Center, Federal State Budgetary Institution “Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency”, 117997 Moscow, Russia;
| | - Daria Namestnikova
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (E.C.); (D.N.); (I.G.); (A.M.); (V.C.)
- Radiology and Clinical Physiology Scientific Research Center, Federal State Budgetary Institution “Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency”, 117997 Moscow, Russia;
| | - Georgy Leonov
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (T.B.); (G.L.); (N.B.); (V.M.); (D.G.)
| | - Maria Nikitina
- Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (A.E.); (K.M.); (L.M.); (T.F.)
| | - Ilya Gubskiy
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (E.C.); (D.N.); (I.G.); (A.M.); (V.C.)
- Radiology and Clinical Physiology Scientific Research Center, Federal State Budgetary Institution “Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency”, 117997 Moscow, Russia;
| | - Gevorg Akopyan
- Radiology and Clinical Physiology Scientific Research Center, Federal State Budgetary Institution “Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency”, 117997 Moscow, Russia;
| | - Andrey Elchaninov
- Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (A.E.); (K.M.); (L.M.); (T.F.)
| | - Konstantin Midiber
- Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (A.E.); (K.M.); (L.M.); (T.F.)
| | - Natalia Bulatenco
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (T.B.); (G.L.); (N.B.); (V.M.); (D.G.)
| | - Victoria Mokrousova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (T.B.); (G.L.); (N.B.); (V.M.); (D.G.)
| | - Andrey Makarov
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (E.C.); (D.N.); (I.G.); (A.M.); (V.C.)
| | - Konstantin Yarygin
- Institute of Biomedical Chemistry, 119121 Moscow, Russia;
- Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
| | - Vladimir Chekhonin
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (E.C.); (D.N.); (I.G.); (A.M.); (V.C.)
| | - Liudmila Mikhaleva
- Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (A.E.); (K.M.); (L.M.); (T.F.)
| | - Timur Fatkhudinov
- Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (A.E.); (K.M.); (L.M.); (T.F.)
- Department of Histology, Cytology and Embryology, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| | - Dmitry Goldshtein
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (T.B.); (G.L.); (N.B.); (V.M.); (D.G.)
- Department of Histology, Cytology and Embryology, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| |
Collapse
|
180
|
Kim JY, Rhim WK, Yoo YI, Kim DS, Ko KW, Heo Y, Park CG, Han DK. Defined MSC exosome with high yield and purity to improve regenerative activity. J Tissue Eng 2021; 12:20417314211008626. [PMID: 33959246 PMCID: PMC8060739 DOI: 10.1177/20417314211008626] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/21/2021] [Indexed: 12/14/2022] Open
Abstract
Exosomes derived from mesenchymal stem cells (MSCs) have been studied as vital
components of regenerative medicine. Typically, various isolation methods of
exosomes from cell culture medium have been developed to increase the isolation
yield of exosomes. Moreover, the exosome-depletion process of serum has been
considered to result in clinically active and highly purified exosomes from the
cell culture medium. Our aim was to compare isolation methods, ultracentrifuge
(UC)-based conventional method, and tangential flow filtration (TFF)
system-based method for separation with high yield, and the bioactivity of the
exosome according to the purity of MSC-derived exosome was determined by the
ratio of Fetal bovine serum (FBS)-derived exosome to MSC-derived exosome
depending on exosome depletion processes of FBS. The TFF-based isolation yield
of exosome derived from human umbilical cord MSC (UCMSC) increased two orders
(92.5 times) compared to UC-based isolation method. Moreover, by optimizing the
process of depleting FBS-derived exosome, the purity of UCMSC-derived exosome,
evaluated using the expression level of MSC exosome surface marker (CD73), was
about 15.6 times enhanced and the concentration of low-density
lipoprotein-cholesterol (LDL-c), known as impurities resulting from FBS, proved
to be negligibly detected. The wound healing and angiogenic effects of highly
purified UCMSC-derived exosomes were improved about 23.1% and 71.4%,
respectively, with human coronary artery endothelial cells (HCAEC). It suggests
that the defined MSC exosome with high yield and purity could increase
regenerative activity.
Collapse
Affiliation(s)
- Jun Yong Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, Republic of Korea.,Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, Republic of Korea.,Department of Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, Republic of Korea
| | - Won-Kyu Rhim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, Republic of Korea
| | - Yong-In Yoo
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, Republic of Korea.,School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Kyoung-Won Ko
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, Republic of Korea
| | - Yun Heo
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, Republic of Korea.,Department of Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, Republic of Korea
| |
Collapse
|
181
|
Lee HL, Yeum CE, Lee H, Oh J, Kim JT, Lee WJ, Ha Y, Yang YI, Kim KN. Peripheral Nerve-Derived Stem Cell Spheroids Induce Functional Recovery and Repair after Spinal Cord Injury in Rodents. Int J Mol Sci 2021; 22:ijms22084141. [PMID: 33923671 PMCID: PMC8072978 DOI: 10.3390/ijms22084141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 01/09/2023] Open
Abstract
Stem cell therapy is one of the most promising candidate treatments for spinal cord injury. Research has shown optimistic results for this therapy, but clinical limitations remain, including poor viability, engraftment, and differentiation. Here, we isolated novel peripheral nerve-derived stem cells (PNSCs) from adult peripheral nerves with similar characteristics to neural-crest stem cells. These PNSCs expressed neural-crest specific markers and showed multilineage differentiation potential into Schwann cells, neuroglia, neurons, and mesodermal cells. In addition, PNSCs showed therapeutic potential by releasing the neurotrophic factors, including glial cell-line-derived neurotrophic factor, insulin-like growth factor, nerve growth factor, and neurotrophin-3. PNSC abilities were also enhanced by their development into spheroids which secreted neurotrophic factors several times more than non-spheroid PNSCs and expressed several types of extra cellular matrix. These features suggest that the potential for these PNSC spheroids can overcome their limitations. In an animal spinal cord injury (SCI) model, these PNSC spheroids induced functional recovery and neuronal regeneration. These PNSC spheroids also reduced the neuropathic pain which accompanies SCI after remyelination. These PNSC spheroids may represent a new therapeutic approach for patients suffering from SCI.
Collapse
Affiliation(s)
- Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Chung-Eun Yeum
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - HyeYeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Jinsoo Oh
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Jong-Tae Kim
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - Won-Jin Lee
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Korea
| | - Young-Il Yang
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
- Correspondence: (Y.-I.Y.); (K.-N.K.)
| | - Keung-Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
- Correspondence: (Y.-I.Y.); (K.-N.K.)
| |
Collapse
|
182
|
Koh AEH, Subbiah SK, Farhana A, Alam MK, Mok PL. Mitigation of Sodium Iodate-Induced Cytotoxicity in Retinal Pigment Epithelial Cells in vitro by Transgenic Erythropoietin-Expressing Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:652065. [PMID: 33937251 PMCID: PMC8082501 DOI: 10.3389/fcell.2021.652065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/15/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSC) have shown promise in restoring the vision of patients in clinical trials. However, this therapeutic effect is not observed in every treated patient and is possibly due to the inefficacies of cell delivery and high cell death following transplantation. Utilizing erythropoietin can significantly enhance the regenerative properties of MSCs and hence improve retinal neuron survivability in oxidative stress. Hence, this study aimed to investigate the efficacy of conditioned medium (CM) obtained from transgenic human erythropoietin-expressing MSCs (MSC EPO ) in protecting human retinal pigment epithelial cells from sodium iodate (NaIO3)-induced cell death. Human MSC and MSC EPO were first cultured to obtain conditioned media (CM). The IC50 of NaIO3 in the ARPE-19 culture was then determined by an MTT assay. After that, the efficacy of both MSC-CM and MSC-CM EPO in ARPE-19 cell survival were compared at 24 and 48 h after NaIO3 treatment with MTT. The treatment effects on mitochondrial membrane potential was then measured by a JC-1 flow cytometric assay. The MTT results indicated a corresponding increase in cell survivability (5-58%) in the ARPE-19 cell cultures. In comparison to MSC-CM, the use of conditioned medium collected from the MSC-CM EPO further enhanced the rate of ARPE-19 survivability at 24 h (P < 0.05) and 48 h (P < 0.05) in the presence of NaIO3. Furthermore, more than 90% were found viable with the JC-1 assay after MSC-CM EPO treatment, showing a positive implication on the mitochondrial dynamics of ARPE-19. The MSC-CM EPO provided an enhanced mitigating effect against NaIO3-induced ARPE-19 cell death over that of MSC-CM alone during the early phase of the treatment, and it may act as a future therapy in treating retinal degenerative diseases.
Collapse
Affiliation(s)
- Avin Ee-Hwan Koh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia
| | - Suresh Kumar Subbiah
- Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia.,Genetics and Regenerative Medicine Research Group, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia.,Department of Biotechnology, Bharath Institute of Higher Education and Research, Chennai, India
| | - Aisha Farhana
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | | | - Pooi Ling Mok
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia.,Genetics and Regenerative Medicine Research Group, Universiti Putra Malaysia, UPM, Seri Kembangan, Malaysia.,Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
183
|
3D cell sheet structure augments mesenchymal stem cell cytokine production. Sci Rep 2021; 11:8170. [PMID: 33854167 PMCID: PMC8046983 DOI: 10.1038/s41598-021-87571-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) secrete paracrine factors that play crucial roles during tissue regeneration. An increasing body of evidence suggests that this paracrine function is enhanced by MSC cultivation in three-dimensional (3D) tissue-like microenvironments. Toward this end, this study explored scaffold-free cell sheet technology as a new 3D platform. MSCs cultivated on temperature-responsive culture dishes to a confluent 2D monolayer were harvested by temperature reduction from 37 to 20 °C that induces a surface wettability transition from hydrophobic to hydrophilic. Release of culture-adherent tension induced spontaneous cell sheet contraction, reducing the diameter 2.4-fold, and increasing the thickness 8.0-fold to render a 3D tissue-like construct with a 36% increase in tissue volume. This 2D-to-3D transition reorganized MSC actin cytoskeleton from aligned to multidirectional, corresponding to a cell morphological change from elongated in 2D monolayers to rounded in 3D cell sheets. 3D culture increased MSC gene expression of cell interaction proteins, β-catenin, integrin β1, and connexin 43, and of pro-tissue regenerative cytokines, vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and interleukin-10 (IL-10), and increased VEGF secretion per MSC 2.1-fold relative to 2D cultures. Together, these findings demonstrate that MSC therapeutic potency can be enhanced by 3D cell sheet tissue structure.
Collapse
|
184
|
Nuge T, Liu X, Tshai KY, Lim SS, Nordin N, Hoque ME, Liu Z. Accelerated wound closure: Systematic evaluation of cellulose acetate effects on biologically active molecules release from amniotic fluid stem cells. Biotechnol Appl Biochem 2021; 69:906-919. [PMID: 33826152 DOI: 10.1002/bab.2162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/26/2021] [Indexed: 01/07/2023]
Abstract
Despite a lot of intensive research on cell-scaffold interaction, the focus is mainly on the capacity of construct scaffolds to regulate cell mobility, migration, and cytotoxicity. The effect of the scaffold's topographical and material properties on the expression of biologically active compounds from stem cells is not well understood. In this study, the influence of cellulose acetate (CA) on the electrospinnability of gelatin and the roles of gelatin-cellulose acetate (Ge-CA) on modulating the release of biologically active compounds from amniotic fluid stem cells (AFSCs) is emphasized. It was found that the presence of a small amount of CA could provide a better microenvironment that mimics AFSCs' niche. However, a large amount of CA exhibited no significant effect on AFSCs migration and infiltration. Further study on the effect of surface topography and mechanical properties on AFSCs showed that the tailored microenvironment provided by the Ge-CA scaffolds had transduced physical cues to biomolecules released into the culture media. It was found that the AFSCs seeded on electrospun scaffolds with less CA proportions have profound effects on the secretion of metabolic compounds compared to those with higher CA contained and gelatin coating. The enhanced secretion of biologically active molecules by the AFSCs on the electrospun scaffolds was proven by the accelerated wound closure on the injured human dermal fibroblast (HDF) model. The rapid HDF cell migration could be anticipated due to a higher level of paracrine factors in AFSCs media. Our study demonstrates that the fibrous topography and mechanical properties of the scaffold are a key material property that modulates the high expression of biologically active compounds from the AFSCs. The discovery elucidates a new aspect of material functions and scaffolds material-AFSC interaction for regulating biomolecules release to promote tissue regeneration/repair. To the best of our knowledge, this is the first report describing the scaffolds material-AFSC interaction and the efficacy of scratch assays on quantifying the cell migration in response to the AFSCs metabolic products.
Collapse
Affiliation(s)
- Tamrin Nuge
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Xiaoling Liu
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Kim Yeow Tshai
- Faculty of Science and Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih, Malaysia
| | - Siew Shee Lim
- Faculty of Science and Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih, Malaysia
| | - Norshariza Nordin
- Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Malaysia
| | - Md Enamul Hoque
- Department of Biomedical Engineering, Military Institute of Science and Technology, Dhaka, Bangladesh
| | - Ziqian Liu
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| |
Collapse
|
185
|
Rangasami VK, Nawale G, Asawa K, Kadekar S, Samanta S, Nilsson B, Ekdahl KN, Miettinen S, Hilborn J, Teramura Y, Varghese OP, Oommen OP. Pluronic Micelle-Mediated Tissue Factor Silencing Enhances Hemocompatibility, Stemness, Differentiation Potential, and Paracrine Signaling of Mesenchymal Stem Cells. Biomacromolecules 2021; 22:1980-1989. [PMID: 33813822 PMCID: PMC8154246 DOI: 10.1021/acs.biomac.1c00070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Mesenchymal stem/stromal
cells (MSCs) evoke great excitement for
treating different human diseases due to their ability to home inflamed
tissues, suppress inflammation, and promote tissue regeneration. Despite
great promises, clinical trial results are disappointing as allotransplantation
of MSCs trigger thrombotic activity and are damaged by the complement
system, compromising their survival and function. To overcome this,
a new strategy is presented by the silencing of tissue factor (TF),
a transmembrane protein that mediates procoagulant activity. Novel Pluronic-based micelles are designed
with the pendant pyridyl disulfide group, which are used to conjugate
TF-targeting siRNA by the thiol-exchange reaction. This nanocarrier
design effectively delivered the payload to MSCs resulting in ∼72%
TF knockdown (KD) without significant cytotoxicity. Hematological
evaluation of MSCs and TF-KD MSCs in an ex vivo human whole blood
model revealed a significant reduction in an instant-blood-mediated-inflammatory
reaction as evidenced by reduced platelet aggregation (93% of free
platelets in the TF-KD group, compared to 22% in untreated bone marrow-derived
MSCs) and thrombin–antithrombin complex formation. Effective
TF silencing induced higher MSC differentiation in osteogenic and
adipogenic media and showed stronger paracrine suppression of proinflammatory
cytokines in macrophages and higher stimulation in the presence of
endotoxins. Thus, TF silencing can produce functional cells with higher
fidelity, efficacy, and functions.
Collapse
Affiliation(s)
- Vignesh K Rangasami
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, Tampere 33720, Finland
| | - Ganesh Nawale
- Translational Chemical Biology Laboratory, Department of Chemistry, Ångström Laboratory, Uppsala University, Uppsala 751 21, Sweden
| | - Kenta Asawa
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Sandeep Kadekar
- Translational Chemical Biology Laboratory, Department of Chemistry, Ångström Laboratory, Uppsala University, Uppsala 751 21, Sweden
| | - Sumanta Samanta
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, Tampere 33720, Finland
| | - Bo Nilsson
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-75105, Sweden
| | - Kristina N Ekdahl
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-75105, Sweden.,Department of Chemistry and Biomedical Sciences, Faculty of Health and Life Sciences, Linnaeus University, Kalmar SE-391 82, Sweden
| | - Susanna Miettinen
- Adult Stem Cells Group, Faculty of Medicine and Health Technologies, Tampere University, Tampere 33014, Finland.,Research, Development and Innovation Center, Tampere University Hospital, Tampere 33520, Finland
| | - Jöns Hilborn
- Polymer Chemistry, Department of Chemistry-Ångström Laboratory, Uppsala University, Uppsala 751 21, Sweden
| | - Yuji Teramura
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-75105, Sweden
| | - Oommen P Varghese
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Oommen P Oommen
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, Tampere 33720, Finland
| |
Collapse
|
186
|
Fan Y, Sun J, Zhang Q, Lai D. Transplantation of human amniotic epithelial cells promotes morphological and functional regeneration in a rat uterine scar model. Stem Cell Res Ther 2021; 12:207. [PMID: 33762002 PMCID: PMC7992833 DOI: 10.1186/s13287-021-02260-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/01/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Cesarean scar defect (CSD) is characterized by the presence of fibrotic tissue and decreased muscular density which is induced by cesarean section. Serious CSD may eventually result in infertility or obstetrical complications. Human amniotic epithelial cells (hAECs) have shown great promise in tissue regeneration. This study aims to investigate whether hAEC transplantation has the therapeutic effects on the rat uterine scar following full-thickness injury. METHODS A rat uterine scar model was established by excising the full-thickness uterine wall of about 1.0 cm in length and 1/2-2/3 of the total circumference in width. At day 30 post-surgery, hAECs were transplanted into the uterine scar. At day 30 and 60 post-transplantation, hematoxylin and eosin (H&E) staining, Masson staining, and IHC staining for vWF, VEGFA, α-SMA, and MMP-8 were performed to evaluate the regeneration of the scarred uterus and the underlying mechanism. Pregnancy outcomes were assessed at day 60 after hAEC transplantation. Finally, hAECs were incubated with hydrogen peroxide to verify the paracrine effect of hAECs. RESULTS Collagen deposition, thin myometrium, and injured endometrium were observed in the rat uterine scar model. After hAEC transplantation, collagen deposition in the uterine scar decreased, and myometrial and endometrial recovery was facilitated. hAEC transplantation also increased the fetus number implanted within the scarred area. Moreover, we found hAECs promoted angiogenesis via upregulation of VEGFA and decreased collagen deposition by upregulating MMP-8 in the uterine scar. The in vitro studies further demonstrated an increase in the expression level of MMP-8 in hAECs cultured with hydrogen peroxide. CONCLUSIONS These results suggested that hAEC transplantation may be efficacious in the functional repair and collagen degradation of uterine scars, which provides a new therapeutic strategy to CSD.
Collapse
Affiliation(s)
- Yihui Fan
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Junyan Sun
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qiuwan Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Dongmei Lai
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
- Shanghai Key Clinical Speciality, Shanghai, 200030, China.
| |
Collapse
|
187
|
Zhao X, Liu Y, Jia P, Cheng H, Wang C, Chen S, Huang H, Han Z, Han ZC, Marycz K, Chen X, Li Z. Chitosan hydrogel-loaded MSC-derived extracellular vesicles promote skin rejuvenation by ameliorating the senescence of dermal fibroblasts. Stem Cell Res Ther 2021; 12:196. [PMID: 33743829 PMCID: PMC7981922 DOI: 10.1186/s13287-021-02262-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/01/2021] [Indexed: 12/27/2022] Open
Abstract
Background The senescence of dermal fibroblasts (DFLs) leads to an imbalance in the synthesis and degradation of extracellular matrix (ECM) proteins, presenting so-called senescence-associated secretory phenotype (SASP), which ultimately leads to skin aging. Recently, mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have been recognized as a promising cell-free therapy for degenerative diseases, which opens a new avenue for skin aging treatment. Methods In this study, we utilized chitosan (CS) hydrogel for effective loading and sustained release of EVs. In vitro, we explored the rejuvenation effects of CS hydrogel-incorporated EVs (CS-EVs) on replicative senescence DFLs through a series of experiments such as senescence-associated β-galactosidase (SA-β-gal) staining, RT-PCR, and Western blot analysis. Besides, we employed local multi-site subcutaneous injection to treat skin aging of naturally aged mice with CS-EVs and DiI fluorescent dye was used to label EVs to achieve in vivo real-time tracking. Results CS-EVs can significantly improve the biological functions of senescent fibroblasts, including promoting their proliferation, enhancing the synthesis of ECM proteins, and inhibiting the overexpression of matrix metalloproteinases (MMPs). Moreover, CS hydrogel could prolong the release of EVs and significantly increase the retention of EVs in vivo. After CS-EVs subcutaneous injection treatment, the aging skin tissues showed a rejuvenation state, manifested explicitly as the enhanced expression of collagen, the decreased expression of SASP-related factors, and the restoration of tissue structures. Conclusions CS hydrogel-encapsulated EVs could delay the skin aging processes by ameliorating the function of aging DFLs. Our results also highlight the potential of CS hydrogel-encapsulated EVs as a novel therapeutic strategy for improving aging skin to rejuvenation.
Collapse
Affiliation(s)
- Xiangnan Zhao
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China.,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, The College of Life Sciences, Tianjin, 300071, China
| | - Yue Liu
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China
| | - Pingping Jia
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hui Cheng
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China
| | - Chen Wang
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China
| | - Shang Chen
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China
| | - Haoyan Huang
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China
| | - Zhibo Han
- Jiangxi Engineering Research Center for Stem Cell, Shangrao, 334109, Jiangxi, China.,Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, 300457, China.,Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co., Beijing, 100176, China
| | - Zhong-Chao Han
- Jiangxi Engineering Research Center for Stem Cell, Shangrao, 334109, Jiangxi, China.,Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, 300457, China.,Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co., Beijing, 100176, China
| | - Krzysztof Marycz
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Xiaoniao Chen
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Zongjin Li
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China. .,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, The College of Life Sciences, Tianjin, 300071, China. .,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
188
|
Zha K, Li X, Yang Z, Tian G, Sun Z, Sui X, Dai Y, Liu S, Guo Q. Heterogeneity of mesenchymal stem cells in cartilage regeneration: from characterization to application. NPJ Regen Med 2021; 6:14. [PMID: 33741999 PMCID: PMC7979687 DOI: 10.1038/s41536-021-00122-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
Articular cartilage is susceptible to damage but hard to self-repair due to its avascular nature. Traditional treatment methods are not able to produce satisfactory effects. Mesenchymal stem cells (MSCs) have shown great promise in cartilage repair. However, the therapeutic effect of MSCs is often unstable partly due to their heterogeneity. Understanding the heterogeneity of MSCs and the potential of different types of MSCs for cartilage regeneration will facilitate the selection of superior MSCs for treating cartilage damage. This review provides an overview of the heterogeneity of MSCs at the donor, tissue source and cell immunophenotype levels, including their cytological properties, such as their ability for proliferation, chondrogenic differentiation and immunoregulation, as well as their current applications in cartilage regeneration. This information will improve the precision of MSC-based therapeutic strategies, thus maximizing the efficiency of articular cartilage repair.
Collapse
Affiliation(s)
- Kangkang Zha
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhen Yang
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Guangzhao Tian
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiqiang Sun
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xiang Sui
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
| | - Yongjing Dai
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
| | - Shuyun Liu
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China.
| | - Quanyi Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China.
| |
Collapse
|
189
|
Shirazi S, Huang CC, Kang M, Lu Y, Ravindran S, Cooper LF. The importance of cellular and exosomal miRNAs in mesenchymal stem cell osteoblastic differentiation. Sci Rep 2021; 11:5953. [PMID: 33723364 PMCID: PMC7960990 DOI: 10.1038/s41598-021-85306-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 02/12/2021] [Indexed: 12/19/2022] Open
Abstract
The differentiation of osteoblasts is under complex regulation that includes autocrine and paracrine signaling from MSCs. Exosomes are important components of the MSC secretome and their cargo contains numerous miRNAs. In this study, the importance of MSC miRNAs in modulation of osteoblastic differentiation was examined by global reduction of miRNA biosynthesis in Dicer knock down hMSCs. We additionally impaired hMSC responses to miRNAs by knockdown of Argonaute 2 expression. Knockdown of Dicer and Argonaute 2 both reduced osteoblastic differentiation of hMSCs. This was observed at the levels of hMSC culture mineralization and osteoblastic gene expression. The treatment of Dicer deficient hMSCs with wild type hMSC exosomes effectively recovered the impaired osteoblastic differentiation. Dicer knockdown reduced the quantity and diversity of miRNAs present in hMSC exosomes. miRSeq data and KEGG analysis implicated the miRNA-dependent effects on multiple osteoinductive pathways in Dicer deficient cells, including the Hippo signaling and TGF-beta signaling pathways. Treatment of hMSCs with mimics of miRNAs significantly downregulated in Dicer knockdown cells recovered functions of exosome-mediated signaling in hMSCs. These results indicate that hMSC exosomes exert miRNA-dependent control that contributes to osteoblastic differentiation.
Collapse
Affiliation(s)
- Sajjad Shirazi
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA
| | - Chun-Chieh Huang
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA
| | - Miya Kang
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA
| | - Yu Lu
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA
| | - Sriram Ravindran
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA.
| | - Lyndon F Cooper
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA.
| |
Collapse
|
190
|
Koprivec S, Novak M, Bernik S, Voga M, Mohorič L, Majdič G. Treatment of cranial cruciate ligament injuries in dogs using a combination of tibial tuberosity advancement procedure and autologous mesenchymal stem cells/multipotent mesenchymal stromal cells - A pilot study. Acta Vet Hung 2021; 68:405-412. [PMID: 33656452 DOI: 10.1556/004.2020.00063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022]
Abstract
In the present pilot study, we evaluated different supplemental therapies using autologous multipotent mesenchymal stromal cells (MMSCs) for the treatment of cranial cruciate ligament defects in dogs. We used tibial tuberosity advancement (TTA) and augmented it by supportive therapy with MMSCs in three patient groups. In the first patient group, the dogs were injected with MMSCs directly into the treated stifle one month after surgery. In the second group, MMSCs were delivered in a silk fibroin scaffold which was placed in the osteotomy gap during surgery. In the third group, MMSCs were first mixed with bone tissue and blood from the patient and delivered into the osteotomy gap during surgery. In the control group, patients underwent the TTA procedure but did not receive MMSC treatment. In the group of patients who received cells in the silk fibroin scaffold during surgery, the osteotomy gap did not heal, presumably due to the low absorption of silk fibroin. Patients who received MMSCs mixed with bone tissue and blood during surgery into the osteotomy gap recovered clinically faster and had better healing of the osteotomy gap than dogs from the other two treated groups and from the control group, as assessed by clinical examination and quantification of radiographs. In conclusion, dogs that received stem cells directly into the osteotomy gap (Group 3) recovered faster compared to dogs from Groups 1 (MMSCs injected into the joint one month after surgery), 2 (cells implanted into the osteotomy gap in a silk fibroin scaffold), and the control group that did not receive additional MMSCs treatment.
Collapse
Affiliation(s)
| | | | | | - Metka Voga
- 2Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - Luka Mohorič
- 3Animacel Biotechnology Ltd., Ljubljana, Slovenia
| | - Gregor Majdič
- 2Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| |
Collapse
|
191
|
Chu W, Liu Z, Gan Y, Chang Y, Jiao X, Jiang W, Dai K. Use of a novel Screen-Enrich-Combine(-biomaterials) Circulating System to fill a 3D-printed open Ti6Al4V frame with mesenchymal stem cells/β-tricalcium phosphate to repair complex anatomical bone defects in load-bearing areas. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:454. [PMID: 33850851 PMCID: PMC8039683 DOI: 10.21037/atm-20-6689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background Repairing complex anatomical load-bearing bone defects is difficult because it requires the restoration of the load-bearing function, reconstructing the anatomical shape, and repair by regenerated bone. We previously developed a Screen–Enrich–Combine(-biomaterials) Circulating System (SECCS) for rapid intraoperative enrichment of autologous bone marrow mesenchymal stem cells (MSCs) to enhance the osteogenic ability of porous bone substitutes. In this study, we prepared a 3D-printed Ti6A14V macroporous frame matching the defect shape to provide early load-bearing support and evaluated the efficacy of filling the frame with SECCS-processed MSCs/beta tricalcium phosphate (β-TCP) for long-term bone growth. Methods Fifteen 2-year-old goats were involved in this study, and the lateral part of their distal femur was removed by an electric saw and was fitted by a matching electron beam melting technology-prepared (EBM) Ti6Al4V frame. Three types of frames, filled with nothing, pure porous β-TCP, or SECCS-processed MSCs/β-TCP, were fixed onto the defect site. Repair efficacy was evaluated by X-ray radiography, computed tomography (CT), histology, and histomorphometry. Results In the basic regular hexagon printing unit, the combined side width (w) and the inscribed circle diameter (d) determines the printing frame’s mechanical strength. The compressive load was significantly higher for w=1.9 mm, d=4.4 mm than for w=1.7 mm, d=4.0 mm or w=2.0 mm, d=5.0 mm (P<0.05). The EBM-prepared Ti6Al4V defect-matched frame was well maintained 9 months after implantation. The MSCs successfully adhered to the wall of the porous β-TCP in the SECCS-processed group and had spread fully in the test samples. Each goat in the MSCs/β-TCP–the filled group, had approximately 31,321.7±22,554.7 of MSCs and a larger area of new bone growth inside the frame than the control and blank areas groups. Conclusions Filling the 3D-printed Ti6Al4V large-aperture frame with osteogenic materials achieved biological reconstruction over a larger area of regenerated bone to repair complex anatomical weight-bearing bone defects under the condition of early frame-supported load bearing. MSCs/β-TCP prepared by SECCS can be used as a filling material for this type of bone defect to obtain more efficacious bone repair.
Collapse
Affiliation(s)
- Wenxiang Chu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhiqing Liu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaokai Gan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongyun Chang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Jiao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenbo Jiang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
192
|
Lohmann S, Eijken M, Møldrup U, Møller BK, Hunter J, Moers C, Leuvenink H, Ploeg RJ, Clahsen-van Groningen MC, Hoogduijn M, Baan CC, Keller AK, Jespersen B. Ex Vivo Administration of Mesenchymal Stromal Cells in Kidney Grafts Against Ischemia-reperfusion Injury-Effective Delivery Without Kidney Function Improvement Posttransplant. Transplantation 2021; 105:517-528. [PMID: 32956281 DOI: 10.1097/tp.0000000000003429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Mesenchymal stromal cell (MSC) therapy may improve renal function after ischemia-reperfusion injury in transplantation. Ex vivo renal intraarterial administration is a targeted delivery method, avoiding the lung vasculature, a known barrier for cellular therapies. In a randomized and blinded study, we tested the feasibility and effectiveness of MSC therapy in a donation after circulatory death autotransplantation model to improve posttransplant kidney function, using an ex vivo MSC delivery method similar to the clinical standard procedure of pretransplant cold graft flush. METHODS Kidneys exposed to 75 minutes of warm ischemia and 16 hours of static cold storage were intraarterially infused ex vivo with 10 million male porcine MSCs (Tx-MSC, n = 8) or vehicle (Tx-control, n = 8). Afterwards, the kidneys were autotransplanted after contralateral nephrectomy. Biopsies an hour after reperfusion confirmed the presence of MSCs in the renal cortex. Animals were observed for 14 days. RESULTS Postoperatively, peak plasma creatinine was 1230 and 1274 µmol/L (Tx-controls versus Tx-MSC, P = 0.69). During follow-up, no significant differences over time were detected between groups regarding plasma creatinine, plasma neutrophil gelatinase-associated lipocalin, or urine neutrophil gelatinase-associated lipocalin/creatinine ratio. At day 14, measured glomerular filtration rates were 40 and 44 mL/min, P = 0.66. Renal collagen content and fibrosis-related mRNA expression were increased in both groups but without significant differences between the groups. CONCLUSIONS We demonstrated intraarterial MSC infusion to transplant kidneys as a safe and effective method to deliver MSCs to the graft. However, we could not detect any positive effects of this cell treatment within 14 days of observation.
Collapse
Affiliation(s)
- Stine Lohmann
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Marco Eijken
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Ulla Møldrup
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Bjarne K Møller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - James Hunter
- Nuffield Department of Surgical Sciences, Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Cyril Moers
- Department of Surgery-Organ Donation and Transplantation, University of Medical Center Groningen, Groningen, the Netherlands
| | - Henri Leuvenink
- Department of Surgery-Organ Donation and Transplantation, University of Medical Center Groningen, Groningen, the Netherlands
| | - Rutger J Ploeg
- Nuffield Department of Surgical Sciences, Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | | | - Martin Hoogduijn
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Anna Krarup Keller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Bente Jespersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
193
|
Ra K, Oh HJ, Kim EY, Kang SK, Ra JC, Kim EH, Park SC, Lee BC. Comparison of Anti-Oxidative Effect of Human Adipose- and Amniotic Membrane-Derived Mesenchymal Stem Cell Conditioned Medium on Mouse Preimplantation Embryo Development. Antioxidants (Basel) 2021; 10:antiox10020268. [PMID: 33572334 PMCID: PMC7916131 DOI: 10.3390/antiox10020268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 01/08/2023] Open
Abstract
Oxidative stress is a major cause of damage to the quantity and quality of embryos produced in vitro. Antioxidants are usually supplemented to protect embryos from the suboptimal in vitro culture (IVC) environment. Amniotic membrane-derived mesenchymal stem cells (AMSC) have emerged as a promising regenerative therapy, and their paracrine factors with anti-oxidative effects are present in AMSC conditioned medium (CM). We examined the anti-oxidative potential of human AMSC-CM treatment during IVC on mouse preimplantation embryo development and antioxidant gene expression in the forkhead box O (FoxO) pathway. AMSC-CM (10%) was optimal for overall preimplantation embryo developmental processes and upregulated the expression of FoxOs and their downstream antioxidants in blastocysts (BL). Subsequently, compared to adipose-derived mesenchymal stem cell (ASC)-CM, AMSC-CM enhanced antioxidant gene expression and intracellular GSH levels in the BL. Total antioxidant capacity and SOD activity were greater in AMSC-CM than in ASC-CM. Furthermore, SOD and catalase were more active in culture medium supplemented with AMSC-CM than in ASC-CM. Lastly, the anti-apoptotic effect of AMSC-CM was observed with the regulation of apoptosis-related genes and mitochondrial membrane potential in BL. In conclusion, the present study established AMSC-CM treatment at an optimal concentration as a novel antioxidant intervention for assisted reproduction.
Collapse
Affiliation(s)
- Kihae Ra
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (K.R.); (H.J.O.); (E.H.K.)
| | - Hyun Ju Oh
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (K.R.); (H.J.O.); (E.H.K.)
- Research and Development Center, MKbiotech Co., Ltd., 99 Daehak-ro, Daejeon 34134, Korea
| | - Eun Young Kim
- Biostar Stem Cell Research Institute, R Bio Co., Ltd., Seoul 08506, Korea; (E.Y.K.); (S.K.K.); (J.C.R.)
| | - Sung Keun Kang
- Biostar Stem Cell Research Institute, R Bio Co., Ltd., Seoul 08506, Korea; (E.Y.K.); (S.K.K.); (J.C.R.)
| | - Jeong Chan Ra
- Biostar Stem Cell Research Institute, R Bio Co., Ltd., Seoul 08506, Korea; (E.Y.K.); (S.K.K.); (J.C.R.)
| | - Eui Hyun Kim
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (K.R.); (H.J.O.); (E.H.K.)
| | - Se Chang Park
- Laboratory of Aquatic Biomedicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
- Correspondence: (S.C.P.); (B.C.L.)
| | - Byeong Chun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (K.R.); (H.J.O.); (E.H.K.)
- Correspondence: (S.C.P.); (B.C.L.)
| |
Collapse
|
194
|
Casati S, Giannasi C, Niada S, Bergamaschi RF, Orioli M, Brini AT. Bioactive Lipids in MSCs Biology: State of the Art and Role in Inflammation. Int J Mol Sci 2021; 22:1481. [PMID: 33540695 PMCID: PMC7867257 DOI: 10.3390/ijms22031481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Lipidomics is a lipid-targeted metabolomics approach that aims to the comprehensive analysis of lipids in biological systems in order to highlight the specific functions of lipid species in health and disease. Lipids play pivotal roles as they are major structural components of the cellular membranes and energy storage molecules but also, as most recently shown, they act as functional and regulatory components of intra- and intercellular signaling. Herein, emphasis is given to the recently highlighted roles of specific bioactive lipids species, as polyunsaturated fatty acids (PUFA)-derived mediators (generally known as eicosanoids), endocannabinoids (eCBs), and lysophospholipids (LPLs), and their involvement in the mesenchymal stem cells (MSCs)-related inflammatory scenario. Indeed, MSCs are a heterogenous population of multipotent cells that have attracted much attention for their potential in regulating inflammation, immunomodulatory capabilities, and reparative roles. The lipidomics of the inflammatory disease osteoarthritis (OA) and the influence of MSCs-derived lipids have also been addressed.
Collapse
Affiliation(s)
- Sara Casati
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
| | - Chiara Giannasi
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
| | | | - Roberta F. Bergamaschi
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
| | - Marica Orioli
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
| | - Anna T. Brini
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
| |
Collapse
|
195
|
Forbes S, Bond AR, Thirlwell KL, Burgoyne P, Samuel K, Noble J, Borthwick G, Colligan D, McGowan NWA, Lewis PS, Fraser AR, Mountford JC, Carter RN, Morton NM, Turner ML, Graham GJ, Campbell JDM. Human umbilical cord perivascular cells improve human pancreatic islet transplant function by increasing vascularization. Sci Transl Med 2021; 12:12/526/eaan5907. [PMID: 31941825 DOI: 10.1126/scitranslmed.aan5907] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/24/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Islet transplantation is an efficacious therapy for type 1 diabetes; however, islets from multiple donor pancreata are required, and a gradual attrition in transplant function is seen. Here, we manufactured human umbilical cord perivascular mesenchymal stromal cells (HUCPVCs) to Good Manufacturing Practice (GMP) standards. HUCPVCs showed a stable phenotype while undergoing rapid ex vivo expansion at passage 2 (p2) to passage 4 (p4) and produced proregenerative factors, strongly suppressing T cell responses in the resting state and in response to inflammation. Transplanting an islet equivalent (IEQ):HUCPVC ratio of 1:30 under the kidney capsule in diabetic NSG mice demonstrated the fastest return to normoglycemia by 3 days after transplant: Superior glycemic control was seen at both early (2.7 weeks) and later stages (7, 12, and 16 weeks) versus ratios of 1:0, 1:10, and 1:50, respectively. Syngeneic islet transplantation in immunocompetent mice using the clinically relevant hepatic portal route with a marginal islet mass showed that mice transplanted with an IEQ:HUCPVC ratio of 1:150 had superior glycemic control versus ratios of 1:0, 1:90, and 1:210 up to 6 weeks after transplant. Immunodeficient mice transplanted with human islets (IEQ:HUCPVC ratio of 1:150) exhibited better glycemic control for 7 weeks after transplant versus islet transplant alone, and islets transplanted via the hepatic portal vein in an allogeneic mouse model using a curative islet mass demonstrated delayed rejection of islets when cotransplanted with HUCPVCs (IEQ:HUCPVC ratio of 1:150). The immunosuppressive and proregenerative properties of HUCPVCs demonstrated long-term positive effects on graft function in vivo, indicating that they may improve long-term human islet allotransplantation outcomes.
Collapse
Affiliation(s)
- Shareen Forbes
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK. .,Clinical Islet Transplantation Programme, Royal Infirmary of Edinburgh, Edinburgh EH16 4SU, UK
| | - Andrew R Bond
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Kayleigh L Thirlwell
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK.,Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Paul Burgoyne
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.,Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK.,Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Kay Samuel
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - June Noble
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Gary Borthwick
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - David Colligan
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Neil W A McGowan
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Philip Starkey Lewis
- Medical Research Council (MRC) Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Alasdair R Fraser
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Joanne C Mountford
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Roderick N Carter
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Nicholas M Morton
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Marc L Turner
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - John D M Campbell
- Advanced Therapeutics, Scottish National Blood Transfusion Service, Edinburgh EH14 4BE, UK. .,Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
196
|
Luo Z, Lin J, Sun Y, Wang C, Chen J. Bone Marrow Stromal Cell-Derived Exosomes Promote Muscle Healing Following Contusion Through Macrophage Polarization. Stem Cells Dev 2021; 30:135-148. [PMID: 33323007 DOI: 10.1089/scd.2020.0167] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle contusion is among the most common injuries in traumatology and clinics of sports medicine. The injured muscle is vulnerable to re-injury owing to fibrosis formation. Given that the bone marrow stromal cell-derived exosomes (BMSC-Exos) displayed promising therapeutic effect for various tissues, we used BMSC-Exos to treat skeletal muscle contusion and investigated its effects on muscle healing. In this study, the in vivo model of skeletal muscle contusion was established by subjecting the tibialis anterior of young male mice to hit injury, and the in vitro inflammation model was established by lipopolysaccharide treatment on macrophages. Macrophage depletion model was built by intraperitoneal injection with clodronate-containing liposomes. Exosomes were isolated and purified from the supernatant of BMSCs using gradient centrifugation. Nanoparticle tracking analysis, transmission electron microscope, and western blot were used to identify the exosomes. HE stain, Masson stain, immunofluorescence, and biomechanical testing were carried out on the muscle tissue. In addition, enzyme-linked immunosorbent assay (ELISA) assays, real-time qPCR, flow cytometry, and PKH67 fluorescence trace were conducted in vitro. Intramuscular injection of BMSC-Exos to mice after muscle contusion alleviated inflammation level, reduced fibrosis size, promoted muscle regeneration, and improved biomechanical property. After macrophages depletion, the effects of BMSC-Exos were inhibited. In vitro, PKH-67 fluorescence was internalized into macrophages. BMSC-Exos promoted M2 macrophages polarization both in vivo and in vitro. At the same time, BMSC-Exos reduced the production of inflammatory cytokines under the inflammatory microenvironment and upregulated anti-inflammatory factors expression. In conclusion, BMSC-Exos attenuated muscle contusion injury and promoted muscle healing in mice by modifying the polarization status of macrophages and suppressing the inflammatory reaction.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinrong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Chenghui Wang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiwu Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
197
|
Akyuz E, Eroglu E. Envisioning the crosstalk between environmental enrichment and epilepsy: A novel perspective. Epilepsy Behav 2021; 115:107660. [PMID: 33328107 DOI: 10.1016/j.yebeh.2020.107660] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/16/2020] [Accepted: 11/21/2020] [Indexed: 12/31/2022]
Abstract
Epilepsies are a diverse group of neurological disorders characterized by an unprovoked seizure and a brain that has an enduring predisposition to seizures. The lack of disease-modifying treatment strategies against the same has led to the exploration of novel treatment strategies that could halt epileptic seizures. In this regard, environmental enrichment (EE) has gained increased attention in recent days. EE modulates the effects of interactions between the genes and the environment on the structure and function of the brain. EE therapy can improve seizure-related symptoms in neurological diseases such as epilepsy. EE therapy can have a significant effect on cognitive disorders such as learning and memory impairments associated with seizures. EE therapy in epileptic hippocampus tissue can improve seizure-related symptoms by inducing enhanced neurogenesis and neuroprotective mechanisms. In this context, the efficiency of EE is regulated in the epilepsy by the brain-derived neurotrophic factor (BDNF)/extracellular signal-regulated kinase (ERK) signaling pathway regulated by extracellular signaling. Herein, we provide experimental evidence supporting the beneficial effects of EE in epileptic seizures and its underlying mechanism.
Collapse
Affiliation(s)
- Enes Akyuz
- Yozgat Bozok University, Medical School, Department of Biophysics, 66100 Yozgat, Turkey.
| | - Ece Eroglu
- Yozgat Bozok University, Medical School, 66100 Yozgat, Turkey.
| |
Collapse
|
198
|
Shafiee S, Heidarpour M, Sabbagh S, Amini E, Saffari H, Dolati S, Meamar R. Stem cell transplantation therapy for diabetic foot ulcer: a narrative review. ASIAN BIOMED 2021; 15:3-18. [PMID: 37551298 PMCID: PMC10388749 DOI: 10.2478/abm-2021-0002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Diabetes mellitus is a chronic metabolic disease associated with high cardiovascular risk. A vascular complication of diabetes is foot ulcers. Diabetic foot ulcers are prevalent and substantially reduce the quality of life of patients who have them. Currently, diabetic foot ulcer is a major problem for wound care specialists, and its treatment requires considerable health care resources. So far, various therapeutic modalities have been proposed to treat diabetic foot ulcers and one of them is stem cell-based therapy. Stem cell-based therapy has shown great promise for the treatment of diabetic foot ulcers. This strategy has been shown to be safe and effective in both preclinical and clinical trials. In this review, we provide an overview of the stem cell types and possible beneficial effects of stem cell transplantation therapy for diabetic foot ulcers, and an overview of the current status of stem cell research in both preclinical and clinical trial stages of treatment strategies for diabetic foot ulcers.
Collapse
Affiliation(s)
- Sahar Shafiee
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan81746-73461, Iran
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Maryam Heidarpour
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan81737-35131, Iran
| | - Sima Sabbagh
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan81746-73461, Iran
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Elham Amini
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan81746-73461, Iran
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Hanieh Saffari
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Sara Dolati
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Rokhsareh Meamar
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan81746-73461, Iran
| |
Collapse
|
199
|
Kim IG, Cho H, Shin J, Cho JH, Cho SW, Chung EJ. Regeneration of irradiation-damaged esophagus by local delivery of mesenchymal stem-cell spheroids encapsulated in a hyaluronic-acid-based hydrogel. Biomater Sci 2021; 9:2197-2208. [PMID: 33506817 DOI: 10.1039/d0bm01655a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Radiation therapy (RT) is a typical treatment for head and neck cancers. Generally, prolonged irradiation of the esophagus causes esophageal fibrosis due to increased reactive oxygen species and proinflammatory cytokines. This study was designed to determine whether catechol-functionalized hyaluronic acid (HA-CA) hydrogel-encapsulated human mesenchymal stem-cell spheroids (MSC-SPs) could ameliorate damage to the esophagus in a mouse model of radiation-induced esophageal fibrosis. MSC-SPs were cultured in concave microwells 600 μm in diameter at a cell density of 1 × 106 cells per mL. Most cells formed spheroids with a 100-300 μm size distribution in concave microwells. MSC-SPs were well maintained in the HA gel, and live-dead staining confirmed that most cells survived. The HA gel containing the MSC-SPs was then injected into the damaged esophageal layer. Inflammatory signs or adverse tissue reactions were not observed after esophageal injection of HA-gel-encapsulated MSC-SPs. Based on Masson's trichrome staining at 4 and 12 weeks postinjection, the inner esophageal layer (IEL) was significantly thinner in the MSC-SP + HA gel group compared to those in the other experimental groups. While the saline and HA gel treatments made the esophageal muscles loose and thick, the MSC-SP + HA gel group showed bundles of tightly packed esophageal muscles, as assayed by desmin immunostaining. qPCR analysis showed that epithelial genes tended to increase over time in the MSC-SP + HA gel group, and the expression of most fibrosis-related genes decreased. This study proposes the potential of using HA-CA-hydrogel-encapsulated MSC-SPs as a promising therapy against radiation-induced esophageal fibrosis.
Collapse
Affiliation(s)
- In Gul Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
200
|
Kukumberg M, Phermthai T, Wichitwiengrat S, Wang X, Arjunan S, Chong SY, Fong CY, Wang JW, Rufaihah AJ, Mattar CNZ. Hypoxia-induced amniotic fluid stem cell secretome augments cardiomyocyte proliferation and enhances cardioprotective effects under hypoxic-ischemic conditions. Sci Rep 2021; 11:163. [PMID: 33420256 PMCID: PMC7794288 DOI: 10.1038/s41598-020-80326-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
Secretome derived from human amniotic fluid stem cells (AFSC-S) is rich in soluble bioactive factors (SBF) and offers untapped therapeutic potential for regenerative medicine while avoiding putative cell-related complications. Characterization and optimal generation of AFSC-S remains challenging. We hypothesized that modulation of oxygen conditions during AFSC-S generation enriches SBF and confers enhanced regenerative and cardioprotective effects on cardiovascular cells. We collected secretome at 6-hourly intervals up to 30 h following incubation of AFSC in normoxic (21%O2, nAFSC-S) and hypoxic (1%O2, hAFSC-S) conditions. Proliferation of human adult cardiomyocytes (hCM) and umbilical cord endothelial cells (HUVEC) incubated with nAFSC-S or hAFSC-S were examined following culture in normoxia or hypoxia. Lower AFSC counts and richer protein content in AFSC-S were observed in hypoxia. Characterization of AFSC-S by multiplex immunoassay showed higher concentrations of pro-angiogenic and anti-inflammatory SBF. hCM demonstrated highest proliferation with 30h-hAFSC-S in hypoxic culture. The cardioprotective potential of concentrated 30h-hAFSC-S treatment was demonstrated in a myocardial ischemia-reperfusion injury mouse model by infarct size and cell apoptosis reduction and cell proliferation increase when compared to saline treatment controls. Thus, we project that hypoxic-generated AFSC-S, with higher pro-angiogenic and anti-inflammatory SBF, can be harnessed and refined for tailored regenerative applications in ischemic cardiovascular disease.
Collapse
Affiliation(s)
- Marek Kukumberg
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tatsanee Phermthai
- Stem Cell Research and Development for Medical Therapy Unit, Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suparat Wichitwiengrat
- Stem Cell Research and Development for Medical Therapy Unit, Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Xiaoyuan Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
| | - Subramanian Arjunan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suet Yen Chong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
| | - Chui-Yee Fong
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Abdul Jalil Rufaihah
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Citra Nurfarah Zaini Mattar
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Obstetrics and Gynaecology, National University Health System, Singapore, Singapore.
| |
Collapse
|