1
|
Al-Sherief S, El-Hadidy A, Hamed S, El-Hawwary A, Mazroa S. Bone marrow mesenchymal stem cells (BM-MSCs) modulate MMP9 expression and promote articular cartilage regeneration in knee joint of a model of arthritis induced in adult rat: histological and immunohistochemical study. J Mol Histol 2024; 56:38. [PMID: 39661261 DOI: 10.1007/s10735-024-10284-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 10/31/2024] [Indexed: 12/12/2024]
Abstract
Arthritis is characterized by the progressive degeneration of articular cartilage, and the avascular nature of cartilage limits its capacity for self-repair. Stem cells are considered a promising treatment option due to their multipotent differentiation potential. The aim of this work was to investigate the structural changes in the hyaline articular cartilage of the knee joint in a model of arthritis induced by complete Freund's adjuvant, and to assess intra-articular injection of bone marrow mesenchymal stem cells (BM-MSCs) through both histological and immunohistochemical study. Adult male albino rats were divided into four groups: group 0 (donor group), group I (control group), group II (arthritis group) and group III (BM-MSCs treated arthritis group). Samples were collected 2, 6 and 10 weeks after the onset of the experiment. Sections were stained with; hematoxylin and eosin, Safranin O fast green stain, Masson's trichrome stain and anti-MMP9 antibody. In Group II (arthritis group), the articular cartilage showed signs of degeneration, including chondrocyte extensive proliferation, fibrillations, fissuring, and denudation, with fibrous tissue covering the exposed surface. There was a significant decrease in cartilage thickness, collagen content, and proteoglycan levels. The integrated density of MMP9 in the cartilage was significantly increased compared to Group I (control group). In contrast, Group III (BM-MSCs-treated arthritis group) exhibited a continuous cartilage surface with no cracks or fissures. There was a significant increase in cartilage thickness, collagen content, and proteoglycan levels, while the integrated density of MMP9 was significantly decreased compared to Group II (arthritis group).
Collapse
Affiliation(s)
- Sara Al-Sherief
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Azza El-Hadidy
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Shereen Hamed
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amany El-Hawwary
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Shireen Mazroa
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
2
|
Salem M, Ateya A, Shouman Z, Salama B, Hamed B, Batiha G, Ataya F, Alexiou A, Papadakis M, Abass M. Amelioration of full-thickness cutaneous wound healing using stem cell exosome and zinc oxide nanoparticles in rats. Heliyon 2024; 10:e38994. [PMID: 39568845 PMCID: PMC11577189 DOI: 10.1016/j.heliyon.2024.e38994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 11/22/2024] Open
Abstract
Background Wound healing is a complex procedure that requires the coordination of several factors, so this study aimed to assess the zinc oxide nanoparticles' regenerated effect and stem cell exosomes on full-thickness wounds in rats. Methods Seventy-two Wistar male rats were subjected to a full-thickness skin defect (20 mm2) on the dorsal surface of each rat between two shoulder joints. The rats were randomized into four groups (18/group) according to wound treatments. The wounds were irrigated with normal saline (Control group), or the wound's edges were subcutaneously injected daily with 0.3 ml of exosome (Exo-group), or 1 ml of zinc oxide nanoparticles (ZnO2-NPs group), or 0.3 ml of exosome in combined with 1 ml of zinc oxide nanoparticles (Exo/ZnO2-NPs group). On the 7th, 14th, and 21st days post-wounding, the weight of the rats, the wound healing breaking strength, the wound size, and the contraction percent were evaluated. Six rats in each group were euthanized at each time point for histopathological, immunohistochemical examination of collagen, the levels of alpha-smooth muscle actin (α-SMA), and epidermal growth factor receptor (EGFR). additionally, the gene expression analysis of the relative renal nuclear factor erythroid 2-related factor2 (Nrf2 mRNA), Transforming growth factor beta-1 (TGFβ1), fibroblast growth factor-7 (FGF7), Transforming growth factor beta-1 (TGFβ1), Lysyl oxidase (LOX), and Vascular endothelial growth factor (VEGF) were applied. Results The Exo-group exhibited a significant decrease in wound size and a significant increase in wound contraction compared with other groups. Histopathologically evaluation during the three intervals revealed that the Exo-group had the highest collagen deposition area with a significant reduction of the granulation tissue. Moreover, upregulated gene expression profiles of the growth factors genes at all time points post-wounding. Discussion The exosomes-treated group revealed superior wound healing and contraction, with minimal inflammatory signs, higher angiogenesis, and myofibroblasts, and associated with higher growth factor expression genes compared to the other groups. Conclusions Exosome-based therapy demonstrates potential as a treatment method to promote and accelerate wound healing by modulating angiogenesis, re-epithelialization, collagen deposition, and gene expression profiles.
Collapse
Affiliation(s)
- Mohamed Salem
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
- Department of Veterinary Clinical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ahmed Ateya
- Department of Development of Animal Wealth, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Zeinab Shouman
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Basma Salama
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Basma Hamed
- Mansoura experimental research center (MERC), Faculty of Medicine, Mansoura, 35516, Egypt
| | - Gaber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| | - Farid Ataya
- Department of Biochemistry, College of Science, King Saud University, PO Box 2455, Riyadh, 11451, Saudi Arabia
| | - Athanasios Alexiou
- Department of Research & Development, Funogen, Athens, 11741, Greece
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany
| | - Marwa Abass
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
3
|
Ghaemi A, Abnous K, Taghdisi SM, Vakili-Azghandi M, Ramezani M, Alibolandi M. Robust aptamer-targeted CRISPR/Cas9 delivery using mesenchymal stem cell membrane -liposome hybrid: BIRC5 gene knockout against melanoma. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 62:102778. [PMID: 39127174 DOI: 10.1016/j.nano.2024.102778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/10/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
In this study, a platform was fabricated by combining a cationic lipid, 1,2-Dioleoyl-3-trimethylammonium-propane (DOTAP) with mesenchymal stem cell membrane (MSCM) to produce a positively charged hybrid vesicle. The prepared hybrid vesicle was used to condense BIRC5 CRISPR/Cas9 plasmid for survivin (BIRC5) gene editing. The Sgc8-c aptamer (against protein tyrosine kinase 7) was then attached to the surface of the prepared NPs through electrostatic interactions. In this regard, melanoma cancer cells (B16F0 cell line) overexpressing PTK7 receptor could be targeted. Investigations were conducted on this system to evaluate its transfection efficiency, cellular toxicity, and therapeutic performance in preclinical stage using B16F0 tumor bearing C57BL/6 J mice. The results verified the superiority of the Hybrid/ BIRC5 compared to Liposome/ BIRC5 in terms of cellular toxicity and transfection efficiency. The cells exposure to Hybrid/BIRC5 significantly enhanced cytotoxicity. Moreover, Apt-Hybrid/BIRC5 showed higher anti-proliferation activity toward PTK7-positive B16F0 cancer cells than that of the PKT7-negative CHO cell line. The active tumor targeting nanoparticles increased the cytotoxicity through down-regulation of BIRC5 expression as confirmed by Western blot analysis. In preclinical stage, Apt-Hybrid/BIRC5 showed remarkable tumor growth suppression toward B16F0 tumorized mice. Thus, our study suggested that genome editing for BIRC5 through the CRISPR/Cas9 system could provide a potentially safe approach for melanoma cancer therapy and has great potential for clinical translation.
Collapse
Affiliation(s)
- Asma Ghaemi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoumeh Vakili-Azghandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Li P, Alenazi KKK, Dally J, Woods EL, Waddington RJ, Moseley R. Role of oxidative stress in impaired type II diabetic bone repair: scope for antioxidant therapy intervention? FRONTIERS IN DENTAL MEDICINE 2024; 5:1464009. [PMID: 39917650 PMCID: PMC11797775 DOI: 10.3389/fdmed.2024.1464009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/02/2024] [Indexed: 02/09/2025] Open
Abstract
Impaired bone healing is a significant complication observed in individuals with type 2 diabetes mellitus (T2DM), leading to prolonged recovery, increased risk of complications, impaired quality of life, and increased risk of patient morbidity. Oxidative stress, resulting from an imbalance between the generation of reactive oxygen species (ROS) and cellular/tissue antioxidant defence mechanisms, has been identified as a critical contributor to the pathogenesis of impaired bone healing in T2DM. Antioxidants have shown promise in mitigating oxidative stress and promoting bone repair, particularly non-enzymic antioxidant entities. This comprehensive narrative review aims to explore the underlying mechanisms and intricate relationship between oxidative stress, impaired bone healing and T2DM, with a specific focus on the current preclinical and clinical evidence advocating the potential of antioxidant therapeutic interventions in improving bone healing outcomes in individuals with T2DM. From the ever-emerging evidence available, it is apparent that exogenously supplemented antioxidants, especially non-enzymic antioxidants, can ameliorate the detrimental effects of oxidative stress, inflammation, and impaired cellular function on bone healing processes during uncontrolled hyperglycaemia; and therefore, hold considerable promise as novel efficacious therapeutic entities. However, despite such conclusions, several important gaps in our knowledge remain to be addressed, including studies involving more sophisticated enzymic antioxidant-based delivery systems, further mechanistic studies into how these antioxidants exert their desirable reparative effects; and more extensive clinical trial studies into the optimisation of antioxidant therapy dosing, frequency, duration and their subsequent biodistribution and bioavailability. By enhancing our understanding of such crucial issues, we can fully exploit the oxidative stress-neutralising properties of these antioxidants to develop effective antioxidant interventions to mitigate impaired bone healing and reduce the associated complications in such T2DM patient populations.
Collapse
Affiliation(s)
- Pui Li
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Kuraym Khalid Kuraym Alenazi
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Jordanna Dally
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Emma Louise Woods
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Rachel Jane Waddington
- Biomaterials Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Ryan Moseley
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
5
|
Li CJ, Park JH, Jin GS, Mandakhbayar N, Yeo D, Lee JH, Lee JH, Kim HS, Kim HW. Strontium/Silicon/Calcium-Releasing Hierarchically Structured 3D-Printed Scaffolds Accelerate Osteochondral Defect Repair. Adv Healthc Mater 2024; 13:e2400154. [PMID: 38647029 DOI: 10.1002/adhm.202400154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/24/2024] [Indexed: 04/25/2024]
Abstract
Articular cartilage defects are a global challenge, causing substantial disability. Repairing large defects is problematic, often exceeding cartilage's self-healing capacity and damaging bone structures. To tackle this problem, a scaffold-mediated therapeutic ion delivery system is developed. These scaffolds are constructed from poly(ε-caprolactone) and strontium (Sr)-doped bioactive nanoglasses (SrBGn), creating a unique hierarchical structure featuring macropores from 3D printing, micropores, and nanotopologies due to SrBGn integration. The SrBGn-embedded scaffolds (SrBGn-µCh) release Sr, silicon (Si), and calcium (Ca) ions, which improve chondrocyte activation, adhesion, proliferation, and maturation-related gene expression. This multiple ion delivery significantly affects metabolic activity and maturation of chondrocytes. Importantly, Sr ions may play a role in chondrocyte regulation through the Notch signaling pathway. Notably, the scaffold's structure and topological cues expedite the recruitment, adhesion, spreading, and proliferation of chondrocytes and bone marrow-derived mesenchymal stem cells. Si and Ca ions accelerate osteogenic differentiation and blood vessel formation, while Sr ions enhance the polarization of M2 macrophages. The findings show that SrBGn-µCh scaffolds accelerate osteochondral defect repair by delivering multiple ions and providing structural/topological cues, ultimately supporting host cell functions and defect healing. This scaffold holds great promise for osteochondral repair applications.
Collapse
Affiliation(s)
- Cheng Ji Li
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeong-Hui Park
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Gang Shi Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Donghyeon Yeo
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Dankook Physician Scientist Research Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Dankook Physician Scientist Research Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Dankook Physician Scientist Research Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Dankook Physician Scientist Research Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
6
|
Wu H, Ye J, Zhang M, Zhang L, Lin S, Li Q, Liu Y, Han Y, Huang C, Wu Y, Cheng Y, Cai S, Ke L, Liu G, Li W, Chu C. A SU6668 pure nanoparticle-based eyedrops: toward its high drug Accumulation and Long-time treatment for corneal neovascularization. J Nanobiotechnology 2024; 22:290. [PMID: 38802884 PMCID: PMC11129376 DOI: 10.1186/s12951-024-02510-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Corneal neovascularization (CNV) is one of the common blinding factors worldwide, leading to reduced vision or even blindness. However, current treatments such as surgical intervention and anti-VEGF agent therapy still have some shortcomings or evoke some adverse effects. Recently, SU6668, an inhibitor targeting angiogenic tyrosine kinases, has demonstrated growth inhibition of neovascularization. But the hydrophobicity and low ocular bioavailability limit its application in cornea. Hereby, we proposed the preparation of SU6668 pure nanoparticles (NanoSU6668; size ~135 nm) using a super-stable pure-nanomedicine formulation technology (SPFT), which possessed uniform particle size and excellent aqueous dispersion at 1 mg/mL. Furthermore, mesenchymal stem cell membrane vesicle (MSCm) was coated on the surface of NanoSU6668, and then conjugated with TAT cell penetrating peptide, preparing multifunctional TAT-MSCm@NanoSU6668 (T-MNS). The T-MNS at a concentration of 200 µg/mL was treated for CNV via eye drops, and accumulated in blood vessels with a high targeting performance, resulting in elimination of blood vessels and recovery of cornea transparency after 4 days of treatment. Meanwhile, drug safety test confirmed that T-MNS did not cause any damage to cornea, retina and other eye tissues. In conclusion, the T-MNS eye drop had the potential to treat CNV effectively and safely in a low dosing frequency, which broke new ground for CNV theranostics.
Collapse
Affiliation(s)
- Han Wu
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Jinfa Ye
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Minjie Zhang
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Department of Rheumatology and Clinical Immunology, School of Medicine, the First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, XM, 361000, China
- Municipal Clinical Research Center for Immune Diseases, Xiamen, XM, 361000, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, XM, 361000, China
| | - Lingyu Zhang
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Sijie Lin
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Qingjian Li
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Yanbo Liu
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Yun Han
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Caihong Huang
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Yiming Wu
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Yuhang Cheng
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Shundong Cai
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Lang Ke
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361002, China.
- Shen Zhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| | - Wei Li
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China.
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China.
| | - Chengchao Chu
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China.
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China.
- Shen Zhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| |
Collapse
|
7
|
Bastami F, Safavi SM, Seifi S, Nadjmi N, Khojasteh A. Addition of Bone-Marrow Mesenchymal Stem Cells to 3D-Printed Alginate/Gelatin Hydrogel Containing Freeze-Dried Bone Nanoparticles Accelerates Regeneration of Critical Size Bone Defects. Macromol Biosci 2024; 24:e2300065. [PMID: 37846197 DOI: 10.1002/mabi.202300065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 08/20/2023] [Indexed: 10/18/2023]
Abstract
A 3D-printed biodegradable hydrogel, consisting of alginate, gelatin, and freeze-dried bone allograft nanoparticles (npFDBA), is developed as a scaffold for enhancing cell adhesion, proliferation, and osteogenic differentiation when combined with rat bone marrow mesenchymal stem cells (rBMSCs). This composite hydrogel is intended for the regeneration of critical-sized bone defects using a rat calvaria defect model. The behavior of rBMSCs seeded onto the scaffold is evaluated through scanning electron microscope, MTT assays, and quantitative real-time PCR. In a randomized study, thirty rats are assigned to five treatment groups: 1) rBMSCs-loaded hydrogel, 2) rBMSCs-loaded FDBA microparticles, 3) hydrogel alone, 4) FDBA alone, and 5) an empty defect serving as a negative control. After 8 weeks, bone regeneration is assessed using H&E, Masson's trichrome staining, and immunohistochemistry. The 3D-printed hydrogel displays excellent adhesion, proliferation, and differentiation of rBMSCs. The rBMSCs-loaded hydrogel exhibits comparable new bone regeneration to the rBMSCs-loaded FDBA group, outperforming other groups with statistical significance (P-value < 0.05). These findings are corroborated by Masson's trichrome staining and osteocalcin expression. The rBMSCs-loaded 3D-printed hydrogel demonstrates promising potential for significantly enhancing bone regeneration, surpassing the conventional clinical approach (FDBA).
Collapse
Affiliation(s)
- Farshid Bastami
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh-Mina Safavi
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sina Seifi
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Nadjmi
- Department of Cranio-Maxillofacial Surgery, University Hospital, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Arash Khojasteh
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Cranio-Maxillofacial Surgery, University Hospital, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Demianova IV, Akoulina EA, Zharkova II, Voinova VV, Chesnokova DV, Hossain AM, Makhina TK, Bonartseva GA, Kulikouskaya VI, Nikalaichuk VV, Mukhortova YR, Pryadko AS, Surmeneva MA, Surmenev RA, Shaitan KV, Bonartsev AP. Growth of Mesenchymal Stem Cells on Oriented Microstructured Films and Electrospun Scaffolds. MOSCOW UNIVERSITY BIOLOGICAL SCIENCES BULLETIN 2023; 78:S34-S39. [DOI: 10.3103/s0096392523700189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/15/2023] [Accepted: 09/25/2023] [Indexed: 01/11/2025]
|
9
|
Yang H, Chen J, Li J. Isolation, culture, and delivery considerations for the use of mesenchymal stem cells in potential therapies for acute liver failure. Front Immunol 2023; 14:1243220. [PMID: 37744328 PMCID: PMC10513107 DOI: 10.3389/fimmu.2023.1243220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Acute liver failure (ALF) is a high-mortality syndrome for which liver transplantation is considered the only effective treatment option. A shortage of donor organs, high costs and surgical complications associated with immune rejection constrain the therapeutic effects of liver transplantation. Recently, mesenchymal stem cell (MSC) therapy was recognized as an alternative strategy for liver transplantation. Bone marrow mesenchymal stem cells (BMSCs) have been used in clinical trials of several liver diseases due to their ease of acquisition, strong proliferation ability, multipotent differentiation, homing to the lesion site, low immunogenicity and anti-inflammatory and antifibrotic effects. In this review, we comprehensively summarized the harvest and culture expansion strategies for BMSCs, the development of animal models of ALF of different aetiologies, the critical mechanisms of BMSC therapy for ALF and the challenge of clinical application.
Collapse
Affiliation(s)
| | | | - Jun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
10
|
Medvecky L, Giretova M, Stulajterova R, Sopcak T, Jevinova P, Luptakova L. Novel Biocement/Honey Composites for Bone Regenerative Medicine. J Funct Biomater 2023; 14:457. [PMID: 37754871 PMCID: PMC10649667 DOI: 10.3390/jfb14090457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/28/2023] Open
Abstract
New biocements based on a powdered mixture of calcium phosphate/monetite (TTCPM) modified with the addition of honey were prepared by mixing the powder and honey liquid components at a non-cytotoxic concentration of honey (up to 10% (w/v)). The setting process of the cements was not affected by the addition of honey, and the setting time of ~4 min corresponded to the fast setting calcium phosphate cements (CPCs). The cement powder mixture was completely transformed into calcium-deficient nanohydroxyapatite after 24 h of hardening in a simulated body fluid, and the columnar growth of long, needle-like nanohydroxyapatite particles around the original calcium phosphate particles was observed in the honey cements. The compressive strength of the honey cements was reduced with the content of honey in the cement. Comparable antibacterial activities were found for the cements with honey solutions on Escherichia coli, but very low antibacterial activities were found for Staphylococcus aureus for all the cements. The enhanced antioxidant inhibitory activity of the composite extracts was verified. In vitro cytotoxicity testing verified the non-cytotoxic nature of the honey cement extracts, and the addition of honey promoted alkaline phosphatase activity, calcium deposit production, and the upregulation of osteogenic genes (osteopontin, osteocalcin, and osteonectin) by mesenchymal stem cells, demonstrating the positive synergistic effect of honey and CPCs on the bioactivity of cements that could be promising therapeutic candidates for the repair of bone defects.
Collapse
Affiliation(s)
- Lubomir Medvecky
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (L.M.); (M.G.); (T.S.)
| | - Maria Giretova
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (L.M.); (M.G.); (T.S.)
| | - Radoslava Stulajterova
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (L.M.); (M.G.); (T.S.)
| | - Tibor Sopcak
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (L.M.); (M.G.); (T.S.)
| | - Pavlina Jevinova
- Department of Food Hygiene, Technology and Safety, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Lenka Luptakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
| |
Collapse
|
11
|
Yusop N, Moseley R, Waddington RJ. Hyperglycemia exerts disruptive effects on the secretion of TGF-β 1 and its matrix ligands, decorin and biglycan, by mesenchymal sub-populations and macrophages during bone repair. FRONTIERS IN DENTAL MEDICINE 2023; 4:1200122. [PMID: 39916897 PMCID: PMC11797960 DOI: 10.3389/fdmed.2023.1200122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/05/2023] [Indexed: 02/09/2025] Open
Abstract
Introduction Bone has a high capacity for repair, but for patients with uncontrolled type 2 diabetes mellitus (T2DM), the associated hyperglycemia can significantly delay osteogenic processes. These patients respond poorly to fracture repair and bone grafts, leading to lengthy care plans due to arising complications. Mesenchymal stromal cells (MSCs) and M2 macrophages are both major sources of transforming growth factor-β1 (TGF-β1), a recognized mediator for osteogenesis and whose bioavailability and activities are further regulated by matrix small leucine-rich proteoglycans (SLRPs), decorin and biglycan. The aim of this study was to investigate how in vivo and in vitro hyperglycemic (HGly) environments can influence the levels of TGF-β1, decorin, and biglycan during bone repair, with additional consideration for how long-term glucose exposure and cell aging can also influence this process. Results Following bone healing within a T2DM in vivo model, histological and immunolabeling analyses of bone tissue sections confirmed delayed healing, which was associated with significantly elevated TGF-β1 levels within the bone matrices of young diabetic rats, compared with their normoglycemic (Norm) and aged counterparts. Studies continued to assess in vitro the effects of normal (5.5 mM) and high (25 mM) glucose exposure on the osteogenic differentiation of compact bone-derived mesenchymal stromal cells (CB-MSCs) at population doubling (PD)15, characterized to contain populations of lineage-committed osteoblasts, and at PD150, where transit-amplifying cells predominate. Short-term glucose exposure increased TGF-β1 and decorin secretion by committed osteoblasts but had a lesser effect on transit-amplifying cells. In contrast, the long-term exposure of CB-MSCs to high glucose was associated with decreased TGF-β1 and increased decorin secretion. Similar assessments on macrophage populations indicated high glucose inhibited TGF-β1 secretion, preventing M2 formation. Discussion Collectively, these findings highlight how hyperglycemia associated with T2DM can perturb TGF-β1 and decorin secretion by MSCs and macrophages, thereby potentially influencing TGF-β1 bioavailability and signaling during bone repair.
Collapse
Affiliation(s)
- Norhayati Yusop
- School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | | | | |
Collapse
|
12
|
Rayia DMA, Izzularab BM, Harras S, Ghafar MTA, Azzam AR, Harras H, Younis RL, Soliman S, Saad AE. Stem cell biotherapy: A new remedy for Trichinella spiralis-induced inflammatory myopathy. Parasitol Int 2023; 96:102773. [PMID: 37330041 DOI: 10.1016/j.parint.2023.102773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Trichinella spiralis (T. spiralis)-induced myopathy is an inflammatory myopathy that is difficult to treat unless the parasite is combated in its early intestinal phase before it reaches the muscles. This study aimed to evaluate the effect of local mesenchymal stem cell (MSC) therapy on T. spiralis-induced inflammatory myopathy in rats. Rats were divided into four groups: Group 1 (non-infected non-treated group); Group 2 (infected non-treated group); Group 3 (infected albendazole (ABZ)-treated group); and Group 4 (infected MSC-treated group). Their muscle status was assessed physiologically with the righting reflex and electromyography (EMG), parasitologically with the total muscle larval count, histopathologically with hematoxylin and eosin and Mallory's trichrome stains, as well as immunohistochemically for myogenin as a marker of muscle regeneration. Additionally, serum muscle enzymes creatine kinase (CK) and lactate dehydrogenase (LDH), as well as muscle matrix metalloproteinases MMP1 and MMP9, were assayed. Finally, the immunological response was assessed by measuring the levels of the muscle inflammatory cytokines tumor necrosis factor-alpha (TNF-α), interferon-gamma (INF-γ), and interleukin-4 (IL-4). Our findings revealed that MSC therapy markedly improved muscle EMG and righting reflex, as well as the histopathological appearance of the muscles, reduced inflammatory cellular infiltrates, and increased myogenin immunostaining. It also reduced serum CK and LDH levels, as well as muscle INF-γ, TNF-α, IL-4, MMP1, and MMP9 levels. However, it had no effect on the total muscle larval count. Accordingly, due to its anti-inflammatory properties and muscle-regenerative effect, MSC therapy could be a promising new remedy for T. spiralis-induced myopathy.
Collapse
Affiliation(s)
- Dina Moustafa Abou Rayia
- Medical Parasitology Department, Faulty of Medicine, Tanta University, Egypt; Medical Parasitology Subunit, Microbiology and Immunology Department, Faculty of Medicine, Mutah University, Jordan.
| | - Batoul M Izzularab
- Biochemistry Division, Chemistry Department, Faculty of Science, Damanhour University, Egypt
| | - Samar Harras
- Zoology Department, Faculty of Science, Tanta University, Egypt
| | | | - Asmaa Ramadan Azzam
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Egypt
| | - Heba Harras
- Histopathology Department, Faculty of Medicine, Tanta University, Egypt
| | | | - Shaimaa Soliman
- Biostatistics and Public Health Department, Faculty of Medicine, Menoufia University, Egypt
| | - Abeer Ezzat Saad
- Medical Parasitology Department, Faulty of Medicine, Tanta University, Egypt; Medical Parasitology Subunit, Pathology Department, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
13
|
Stulajterova R, Giretova M, Medvecky L, Sopcak T, Luptakova L, Girman V. The Influence of Nanosilica on Properties of Cement Based on Tetracalcium Phosphate/Monetite Mixture with Addition of Magnesium Pyrophoshate. MATERIALS (BASEL, SWITZERLAND) 2022; 15:8212. [PMID: 36431697 PMCID: PMC9692293 DOI: 10.3390/ma15228212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/04/2022] [Accepted: 11/14/2022] [Indexed: 06/16/2023]
Abstract
The effect of nanosilica on the microstructure setting process of tetracalcium phosphate/nanomonetite calcium phosphate cement mixture (CPC) with the addition of 5 wt% of magnesium pyrophosphate (assigned as CT5MP) and osteogenic differentiation of mesenchymal stem cells cultured in cement extracts were studied. A more compact microstructure was observed in CT5MP cement with 0.5 wt% addition of nanosilica (CT5MP1Si) due to the synergistic effect of Mg2P2O7 particles, which strengthened the cement matrix and nanosilica, which supported gradual growth and recrystallization of HAP particles to form compact agglomerates. The addition of 0.5 wt% of nanosilica to CT5MP cement caused an increase in CS from 18 to 24 MPa while the setting time increased almost twofold. It was verified that adding nanosilica to CPC cement, even in a low amount (0.5 and 1 wt% of nanosilica), positively affected the injectability of cement pastes and differentiation of cells with upregulation of osteogenic markers in cells cultured in cement extracts. Results revealed appropriate properties of these types of cement for filling bone defects.
Collapse
Affiliation(s)
- Radoslava Stulajterova
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia
| | - Maria Giretova
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia
| | - Lubomir Medvecky
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia
| | - Tibor Sopcak
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia
| | - Lenka Luptakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia
| | - Vladimir Girman
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia
| |
Collapse
|
14
|
Nassar SZ, Abdelmonsif DA, Ali RG, Badae NM. Sodium hydrosulfide and bone marrow derived mesenchymal stem cells combined therapy for bleomycin induced pulmonary fibrosis in rats: Implication of micro RNA-21 and Lnc GAS5. Life Sci 2022; 309:120988. [PMID: 36155181 DOI: 10.1016/j.lfs.2022.120988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022]
Abstract
AIMS Pulmonary fibrosis (PF) is considered as an end stage for many lung diseases. Mesenchymal stem cells (MSC) as regenerative therapy have become a remarkably valuable therapeutic strategy in different diseases. Hydrogen sulfide has been recently introduced into the medical field for its antifibrotic properties in addition to enhancement of MSC stemness and function. The aim of the present study was to investigate the ability of BM-MSC in combination with NaHS to attenuate Bleomycin induced pulmonary fibrosis was studied in rats. A special emphasis was given to miR-21 and GAS5 as important players in the development of PF. MAIN METHODS PF was induced in 32 Wistar male rats by single endotracheal injection of bleomycin, those were randomly divided into four groups (8 rats each): (untreated PF group) - (PF + MSC) treated group- (PF + NaHS treated group) - PF + combined (NAHS + MSC) treated group. KEY FINDINGS Induction of PF was associated with increased miR-21 and decreased lncRNA-GAS5 expression. Treatment with either NaHS or BM-MSC leads to an inhibitory effect on pulmonary fibrosis as evidenced by improvement of histopathological studies, pulmonary function tests, reduction of inflammatory and fibrotic markers like Hydroxyproline, TNF α, TGF-β and caspase -3 together with downregulation miR-21 and increase lncRNA-GAS5 expression. SIGNIFICANCE The current work revealed the inhibitory effect of combined NaHS and BM-MSC on pulmonary fibrosis with concomitant modulation of miR-21 and lncRNA-GAS5 expression.
Collapse
Affiliation(s)
- Seham Z Nassar
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Doaa A Abdelmonsif
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt; Molecular Biology and Nanomedicine Labs, Centre of Excellence for Regenerative Medicine Research & Applications, University of Alexandria, Alexandria, Egypt
| | - Rania Gaber Ali
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Noha Mohamed Badae
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
15
|
Therapeutic Potential of Mesenchymal Stem Cells versus Omega n − 3 Polyunsaturated Fatty Acids on Gentamicin-Induced Cardiac Degeneration. Pharmaceutics 2022; 14:pharmaceutics14071322. [PMID: 35890218 PMCID: PMC9319609 DOI: 10.3390/pharmaceutics14071322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/28/2022] [Accepted: 06/17/2022] [Indexed: 01/27/2023] Open
Abstract
This study compared the cardioprotective action of mesenchymal stem cells (MSCs) and PUFAs in a rat model of gentamicin (GM)-induced cardiac degeneration. Male Wistar albino rats were randomized into four groups of eight rats each: group I (control group), group II (gentamicin-treated rats receiving gentamicin intraperitoneally (IP) at dose of 100 mg/kg/day for 10 consecutive days), group III (gentamicin and PUFA group receiving gentamicin IP at dose of 100 mg/kg/day for 10 consecutive days followed by PUFAs at a dose of 100 mg/kg/day for 4 weeks), and group IV (gentamicin and MSC group receiving gentamicin IP at dose of 100 mg/kg/day followed by a single dose of MSCs (1 × 106)/rat IP). Cardiac histopathology was evaluated via light and electron microscopy. Immunohistochemical detection of proliferating cell nuclear antigen (PCNA), caspase-3 (apoptosis), Bcl2, and Bax expression was performed. Moreover, cardiac malonaldehyde (MDA) content, catalase activity, and oxidative stress parameters were biochemically evaluated. Light and electron microscopy showed that both MSCs and PUFAs had ameliorative effects. Their actions were mediated by upregulating PCNA expression, downregulating caspase-3 expression, mitigating cardiac MDA content, catalase activity, and oxidative stress parameters. MSCs and PUFAs had ameliorative effects against gentamicin-induced cardiac degeneration, with MSCs showing higher efficacy compared to PUFAs.
Collapse
|
16
|
Highly Porous Type II Collagen-Containing Scaffolds for Enhanced Cartilage Repair with Reduced Hypertrophic Cartilage Formation. Bioengineering (Basel) 2022; 9:bioengineering9060232. [PMID: 35735475 PMCID: PMC9220058 DOI: 10.3390/bioengineering9060232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 12/15/2022] Open
Abstract
The ability to regenerate damaged cartilage capable of long-term performance in an active joint remains an unmet clinical challenge in regenerative medicine. Biomimetic scaffold biomaterials have shown some potential to direct effective cartilage-like formation and repair, albeit with limited clinical translation. In this context, type II collagen (CII)-containing scaffolds have been recently developed by our research group and have demonstrated significant chondrogenic capacity using murine cells. However, the ability of these CII-containing scaffolds to support improved longer-lasting cartilage repair with reduced calcified cartilage formation still needs to be assessed in order to elucidate their potential therapeutic benefit to patients. To this end, CII-containing scaffolds in presence or absence of hyaluronic acid (HyA) within a type I collagen (CI) network were manufactured and cultured with human mesenchymal stem cells (MSCs) in vitro under chondrogenic conditions for 28 days. Consistent with our previous study in rat cells, the results revealed enhanced cartilage-like formation in the biomimetic scaffolds. In addition, while the variable chondrogenic abilities of human MSCs isolated from different donors were highlighted, protein expression analysis illustrated consistent responses in terms of the deposition of key cartilage extracellular matrix (ECM) components. Specifically, CI/II-HyA scaffolds directed the greatest cell-mediated synthesis and accumulation in the matrices of type II collagen (a principal cartilage ECM component), and reduced deposition of type X collagen (a key protein associated with hypertrophic cartilage formation). Taken together, these results provide further evidence of the capability of these CI/II-HyA scaffolds to direct enhanced and longer-lasting cartilage repair in patients with reduced hypertrophic cartilage formation.
Collapse
|
17
|
Stulajterova R, Medvecky L, Giretova M, Sopcak T, Luptakova L, Bures R, Szekiova E. Characterization of Tetracalcium Phosphate/Monetite Biocement Modified by Magnesium Pyrophosphate. MATERIALS (BASEL, SWITZERLAND) 2022; 15:2586. [PMID: 35407918 PMCID: PMC9000233 DOI: 10.3390/ma15072586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 11/24/2022]
Abstract
Magnesium pyrophosphate modified tetracalcium phosphate/monetite cement mixtures (MgTTCPM) were prepared by simple mechanical homogenization of compounds in a ball mill. The MgP2O7 was chosen due to the suitable setting properties of the final cements, in contrast to cements with the addition of amorphous (Ca, Mg) CO3 or newberite, which significantly extended the setting time even in small amounts (corresponding ~to 1 wt% of Mg in final cements). The results showed the gradual dissolution of the same amount of Mg2P2O7 phase, regardless of its content in the cement mixtures, and the refinement of formed HAP nanoparticles, which were joined into weakly and mutually bound spherical agglomerates. The compressive strength of composite cements was reduced to 14 MPa and the setting time was 5-10 min depending on the composition. Cytotoxicity of cements or their extracts was not detected and increased proliferative activity of mesenchymal stem cells with upregulation of osteopontin and osteonectin genes was verified in cells cultured for 7 and 15 days in cement extracts. The above facts, including insignificant changes in the pH of simulated body fluid solution and mechanical strength close to cancellous bone, indicate that MgTTCPM cement mixtures could be suitable biomaterials for use in the treatment of bone defects.
Collapse
Affiliation(s)
- Radoslava Stulajterova
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (R.S.); (M.G.); (T.S.); (R.B.)
| | - Lubomir Medvecky
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (R.S.); (M.G.); (T.S.); (R.B.)
| | - Maria Giretova
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (R.S.); (M.G.); (T.S.); (R.B.)
| | - Tibor Sopcak
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (R.S.); (M.G.); (T.S.); (R.B.)
| | - Lenka Luptakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Radovan Bures
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (R.S.); (M.G.); (T.S.); (R.B.)
| | - Eva Szekiova
- Institute of Neurobiology of Biomedical Research Center of SAS, Soltesovej 4–6, 040 01 Kosice, Slovakia;
| |
Collapse
|
18
|
Brown SV, Dewitt S, Clayton A, Waddington RJ. Identifying the Efficacy of Extracellular Vesicles in Osteogenic Differentiation: An EV-Lution in Regenerative Medicine. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.849724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have long been the focus for regenerative medicine and the restoration of damaged or aging cells throughout the body. However, the efficacy of MSCs in cell-based therapy still remains unpredictable and carries with it enumerable risks. It is estimated that only 3-10% of MSCs survive transplantation, and there remains undefined and highly variable heterogeneous biological potency within these administered cell populations. The mode of action points to secreted factors produced by MSCs rather than the reliance on engraftment. Hence harnessing such secreted elements as a replacement for live-cell therapies is attractive. Extracellular vesicles (EVs) are heterogenous lipid bounded structures, secreted by cells. They comprise a complex repertoire of molecules including RNA, proteins and other factors that facilitate cell-to-cell communication. Described as protected signaling centers, EVs can modify the cellular activity of recipient cells and are emerging as a credible alternative to cell-based therapies. EV therapeutics demonstrate beneficial roles for wound healing by preventing apoptosis, moderating immune responses, and stimulating angiogenesis, in addition to promoting cell proliferation and differentiation required for tissue matrix synthesis. Significantly, EVs maintain their signaling function following transplantation, circumventing the issues related to cell-based therapies. However, EV research is still in its infancy in terms of their utility as medicinal agents, with many questions still surrounding mechanistic understanding, optimal sourcing, and isolation of EVs for regenerative medicine. This review will consider the efficacy of using cell-derived EVs compared to traditional cell-based therapies for bone repair and regeneration. We discuss the factors to consider in developing productive lines of inquiry and establishment of standardized protocols so that EVs can be harnessed from optimal secretome production, to deliver reproducible and effective therapies.
Collapse
|
19
|
Liu X, Zhan Y, Xu W, Liu L, Liu X, Da J, Zhang K, Zhang X, Wang J, Liu Z, Jin H, Zhang B, Li Y. Characterization of transcriptional landscape in bone marrow-derived mesenchymal stromal cells treated with aspirin by RNA-seq. PeerJ 2022; 10:e12819. [PMID: 35127290 PMCID: PMC8793730 DOI: 10.7717/peerj.12819] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/30/2021] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Aspirin is a common antipyretic, analgesic, and anti-inflammatory drug, which has been reported to extend life in animal models and application in the treatment of aging-related diseases. However, it remains unclear about the effects of aspirin on bone marrow-derived mesenchymal stromal cells (BM-MSCs). Here, we aimed to analyze the influence of aspirin on senescence and young BM-MSCs. METHODS BM-MSCs were serially passaged to construct a replicative senescence model. SA-β-gal staining, PCR, western blot, and RNA-sequencing were performed on BM-MSCs with or without aspirin treatment, to examine aspirin's impact on bone marrow-derived mesenchymal stem cells. RESULTS SA-β-gal staining, PCR, and western blot revealed that aspirin could alleviate the cellular expression of senescence-related indicators of BM-MSCs, including a decrease of SA-β-gal-positive cells and staining intensity, and downregulation of p16, p21, and p53 expression after aspirin treatment. RNA-sequencing results shown in the biological processes related to aging, aspirin could influence cellular immune response and lipid metabolism. CONCLUSION The efficacy of aspirin for retarding senescence of BM-MSCs was demonstrated. Our study indicated that the mechanisms of this delay might involve influencing immune response and lipid metabolism.
Collapse
Affiliation(s)
- Xinpeng Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuanbo Zhan
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China,The Second Affiliated Hospital of Harbin Medical University, Department of Periodontology and Oral Mucosa, Harbin, China
| | - Wenxia Xu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lixue Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyao Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Junlong Da
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kai Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinjian Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianqun Wang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ziqi Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Han Jin
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bin Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China,Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Ying Li
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
20
|
Ikram R, Shamsuddin SAA, Mohamed Jan B, Abdul Qadir M, Kenanakis G, Stylianakis MM, Anastasiadis SH. Impact of Graphene Derivatives as Artificial Extracellular Matrices on Mesenchymal Stem Cells. Molecules 2022; 27:379. [PMID: 35056690 PMCID: PMC8781794 DOI: 10.3390/molecules27020379] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 02/04/2023] Open
Abstract
Thanks to stem cells' capability to differentiate into multiple cell types, damaged human tissues and organs can be rapidly well-repaired. Therefore, their applicability in the emerging field of regenerative medicine can be further expanded, serving as a promising multifunctional tool for tissue engineering, treatments for various diseases, and other biomedical applications as well. However, the differentiation and survival of the stem cells into specific lineages is crucial to be exclusively controlled. In this frame, growth factors and chemical agents are utilized to stimulate and adjust proliferation and differentiation of the stem cells, although challenges related with degradation, side effects, and high cost should be overcome. Owing to their unique physicochemical and biological properties, graphene-based nanomaterials have been widely used as scaffolds to manipulate stem cell growth and differentiation potential. Herein, we provide the most recent research progress in mesenchymal stem cells (MSCs) growth, differentiation and function utilizing graphene derivatives as extracellular scaffolds. The interaction of graphene derivatives in human and rat MSCs has been also evaluated. Graphene-based nanomaterials are biocompatible, exhibiting a great potential applicability in stem-cell-mediated regenerative medicine as they may promote the behaviour control of the stem cells. Finally, the challenges, prospects and future trends in the field are discussed.
Collapse
Affiliation(s)
- Rabia Ikram
- Department of Chemical Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | | | - Badrul Mohamed Jan
- Department of Chemical Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | | | - George Kenanakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, GR-70013 Heraklion, Greece; (G.K.); (S.H.A.)
| | - Minas M. Stylianakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, GR-70013 Heraklion, Greece; (G.K.); (S.H.A.)
- Department of Nursing, Faculty of Health Sciences, Hellenic Mediterranean University, GR-71410 Heraklion, Greece
| | - Spiros H. Anastasiadis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, GR-70013 Heraklion, Greece; (G.K.); (S.H.A.)
| |
Collapse
|
21
|
Wang P, Zhu P, Yu C, Wu J. The Proliferation and Stemness of Peripheral Blood-Derived Mesenchymal Stromal Cells Were Enhanced by Hypoxia. Front Endocrinol (Lausanne) 2022; 13:873662. [PMID: 35634504 PMCID: PMC9134856 DOI: 10.3389/fendo.2022.873662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/05/2022] [Indexed: 01/08/2023] Open
Abstract
This study aimed to address the dilemma of low peripheral blood-derived mesenchymal stromal cell (PBMSC) activity and reduced phenotype in bone or cartilage tissue engineering. Rat PBMSCs (rPBMSCs) were obtained by density gradient centrifugation, and stromal cell characteristics were confirmed by flow cytometry (FCM) and multi-differentiation potential induction experiments. Cell growth curve, viability experiments, and clone formation experiments were performed by [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] (MTS) and cell counting, and the cell cycle was confirmed by cell FCM. The proliferation signal pathway and stemness-related proteins were detected by molecular methods including Western blot and real-time polymerase chain reaction. CD73, CD90, and CD105 were highly expressed, and CD14, CD19, CD34, CD45, and HLA-DR were barely expressed in rPBMSCs. rPBMSCs possessed the potential to differentiate into chondrocytes, adipocytes, and osteoblasts under their respective induction conditions. Cell growth curve and viability experiments were performed under hypoxic conditions: 19% O2, 5% O2, and 1% O2. Specifically, 5% O2 accelerated the proliferation and expression of the stemness of PBMSCs. Cycle experiments proved that hypoxia promoted the cell transition from the G1 phase to the S phase. Molecular experiments confirmed that 5% O2 hypoxia significantly elevated the expressions of hypoxia-inducible factor 1α and β-catenin and simultaneously the expressions of cycle-related genes including CyclinE/CDK2 and stemness-related genes including Nanog and SOX2. The appropriate concentration of hypoxia (i.e., 5% O2) enhanced the proliferation and stemness of rPBMSCs and increased the multidirectional differentiation potential of stromal cells. The proposed culture method could improve the viability and maintain the phenotype of rPBMSCs in cartilage or bone tissue engineering.
Collapse
Affiliation(s)
- Pengzhen Wang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
- *Correspondence: Pengzhen Wang,
| | - Pingping Zhu
- Department of Neurology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Chaosheng Yu
- Department of Otorhinolaryngology, Guangzhou Red Cross Medicine, Jinan University, Guangzhou, China
| | - Jian Wu
- Department of Otorhinolaryngology, Guangzhou Red Cross Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
22
|
Pimenta DB, Varela VA, Datoguia TS, Caraciolo VB, Lopes GH, Pereira WO. The Bone Marrow Microenvironment Mechanisms in Acute Myeloid Leukemia. Front Cell Dev Biol 2021; 9:764698. [PMID: 34869355 PMCID: PMC8639599 DOI: 10.3389/fcell.2021.764698] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
Bone marrow (BM) is a highly complex tissue that provides important regulatory signals to orchestrate hematopoiesis. Resident and transient cells occupy and interact with some well characterized niches to produce molecular and cellular mechanisms that interfere with differentiation, migration, survival, and proliferation in this microenvironment. The acute myeloid leukemia (AML), the most common and severe hematological neoplasm in adults, arises and develop in the BM. The osteoblastic, vascular, and reticular niches provide surface co-receptors, soluble factors, cytokines, and chemokines that mediate important functions on hematopoietic cells and leukemic blasts. There are some evidences of how AML modify the architecture and function of these three BM niches, but it has been still unclear how essential those modifications are to maintain AML development. Basic studies and clinical trials have been suggesting that disturbing specific cells and molecules into the BM niches might be able to impair leukemia competencies. Either through niche-specific molecule inhibition alone or in combination with more traditional drugs, the bone marrow microenvironment is currently considered the potential target for new strategies to treat AML patients. This review describes the cellular and molecular constitution of the BM niches under healthy and AML conditions, presenting this anatomical compartment by a new perspective: as a prospective target for current and next generation therapies.
Collapse
Affiliation(s)
- Débora Bifano Pimenta
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Vanessa Araujo Varela
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Tarcila Santos Datoguia
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Victória Bulcão Caraciolo
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Gabriel Herculano Lopes
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Welbert Oliveira Pereira
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
23
|
Purwaningrum M, Jamilah NS, Purbantoro SD, Sawangmake C, Nantavisai S. Comparative characteristic study from bone marrow-derived mesenchymal stem cells. J Vet Sci 2021; 22:e74. [PMID: 34697921 PMCID: PMC8636658 DOI: 10.4142/jvs.2021.22.e74] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 11/29/2022] Open
Abstract
Tissue engineering has been extensively investigated and proffered to be a potential platform for novel tissue regeneration. The utilization of mesenchymal stem cells (MSCs) from various sources has been widely explored and compared. In this regard, MSCs derived from bone marrow have been proposed and described as a promising cell resource due to their high yield of isolated cells with colony-forming potential, self-renewal capacity, MSC surface marker expression, and multi-lineage differentiation capacities in vitro. However, there is evidence for bone marrow MSCs (BM-MSCs) both in vitro and in vivo from different species presenting identical and distinct potential stemness characteristics. In this review, the fundamental knowledge of the growth kinetics and stemness properties of BM-MSCs in different animal species and humans are compared and summarized. Finally, to provide a full perspective, this review will procure results of current information studies focusing on the use of BM-MSCs in clinical practice.
Collapse
Affiliation(s)
- Medania Purwaningrum
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.,Department of Biochemistry, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Nabila Syarifah Jamilah
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Steven Dwi Purbantoro
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chenphop Sawangmake
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.,Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.,Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sirirat Nantavisai
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.,Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
24
|
Medvecky L, Giretova M, Stulajterova R, Luptakova L, Sopcak T, Girman V. Osteogenic potential and properties of injectable silk fibroin/tetracalcium phosphate/monetite composite powder biocement systems. J Biomed Mater Res B Appl Biomater 2021; 110:668-678. [PMID: 34569694 DOI: 10.1002/jbm.b.34945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/31/2021] [Accepted: 09/09/2021] [Indexed: 11/08/2022]
Abstract
The powdered cement tetracalcium phosphate/monetite/silk fibroin composite (CFIB) was prepared by simple mechanical milling of tetracalcium phosphate/monetite powder mixture with fibrous soluble silk fibroin (SF). The powder composite cement mixtures contained 5 and 10 wt % of SF and 2% NaH2 PO4 solution with 0.1% genipin was used as a liquid component. The setting time of CFIB cement increased with addition of SF from 5 to 25 min in fully injectable cement with 10 wt % of SF. The compressive strength of hardened composites was reduced to 14 MPa which is close to strength of cancellous bone. The 8% of SF from origin amount in CFIB composites was only desorbed from cements after 7 days soaking in simulated body fluid (SBF). It was found almost full transformation of calcium phosphate components in composite to rod-like nanohydroxyapatite after hardening of CFIB cements in SBF. The SF in hardened cements was present in fine globular form after dissolution, actively affected the fluidity of pastes, morphology of hydroxyapatite particles, and microstructure. The excellent cell proliferation and a high over expression of osteogenic gene markers in MSCs were confirmed after the long-time cultivation in CFIB10 cement extract. Injectable CFIB10 cements have appropriate properties for utilization in bone defect treatments with possible positive effect on healing process.
Collapse
Affiliation(s)
- Lubomir Medvecky
- Department of Functional and Hybrid Materials, Institute of Materials Research of SAS, Kosice, Slovakia
| | - Maria Giretova
- Department of Functional and Hybrid Materials, Institute of Materials Research of SAS, Kosice, Slovakia
| | - Radoslava Stulajterova
- Department of Functional and Hybrid Materials, Institute of Materials Research of SAS, Kosice, Slovakia
| | - Lenka Luptakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Kosice, Slovakia
| | - Tibor Sopcak
- Department of Functional and Hybrid Materials, Institute of Materials Research of SAS, Kosice, Slovakia
| | - Vladimir Girman
- Department of Functional and Hybrid Materials, Institute of Materials Research of SAS, Kosice, Slovakia
| |
Collapse
|
25
|
Karabıyık Acar Ö, Bedir S, Kayitmazer AB, Kose GT. Chondro-inductive hyaluronic acid/chitosan coacervate-based scaffolds for cartilage tissue engineering. Int J Biol Macromol 2021; 188:300-312. [PMID: 34358603 DOI: 10.1016/j.ijbiomac.2021.07.176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022]
Abstract
Injuries related to articular cartilage are among the most challenging musculoskeletal problems because of poor repair capacity of this tissue. The lack of efficient treatments for chondral defects has stimulated research on cartilage tissue engineering applications combining porous biocompatible scaffolds with stem cells in the presence of external stimuli. This work presents the role of rat bone marrow mesenchymal stem cell (BMSC) encapsulated-novel three-dimensional (3D) coacervate scaffolds prepared through complex coacervation between different chitosan salts (CHI) and sodium hyaluronate (HA). The 3D architecture of BMSC encapsulated scaffolds (HA/CHI) was shown by scanning electron microscopy (SEM) to have an interconnected structure to allow cell-cell and cell-matrix interactions. Chondrogenic induction of encapsulated BMSCs within HA/CHI coacervates demonstrated remarkable cellular viability in addition to the elevated expression levels of chondrogenic markers such as sex determining region Y-box 9 protein (SOX9), aggrecan (ACAN), cartilage oligomeric matrix protein (COMP) and collagen type II (COL2A1) by immunofluorescence staining, qPCR and ELISA test. Collectively, HA/CHI coacervates are promising candidates for future use of these scaffolds in cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Özge Karabıyık Acar
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.
| | - Seden Bedir
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | | | - Gamze Torun Kose
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
26
|
Flanagan M, Pathak I, Gan Q, Winter L, Emnet R, Akel S, Montaño AM. Umbilical mesenchymal stem cell-derived extracellular vesicles as enzyme delivery vehicle to treat Morquio A fibroblasts. Stem Cell Res Ther 2021; 12:276. [PMID: 33957983 PMCID: PMC8101245 DOI: 10.1186/s13287-021-02355-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mucopolysaccharidosis IVA (Morquio A syndrome) is a lysosomal storage disease caused by the deficiency of enzyme N-acetylgalactosamine-6-sulfate sulfatase (GALNS), which results in the accumulation of the glycosaminoglycans (GAGs), keratan sulfate, and chondroitin-6-sulfate in the lysosomes of all tissues causing systemic dysfunction. Current treatments include enzyme replacement therapy (ERT) which can treat only certain aspects of the disease such as endurance-related biological endpoints. A key challenge in ERT is ineffective enzyme uptake in avascular tissues, which makes the treatment of the corneal, cartilage, and heart valvular tissue difficult. The aim of this study was to culture human umbilical mesenchymal stem cells (UMSC), demonstrate presence of GALNS enzyme activity within the extracellular vesicles (EVs) derived from these UMSC, and study how these secreted EVs are taken up by GALNS-deficient cells and used by the deficient cell's lysosomes. METHODS We obtained and cultured UMSC from the umbilical cord tissue from anonymous donors from the Saint Louis Cord Blood Bank. We characterized UMSC cell surface markers to confirm phenotype by cell sorting analyses. In addition, we confirmed that UMSC secrete GALNS enzyme creating conditioned media for co-culture experiments with GALNS deficient cells. Lastly, we isolated EVs derived from UMSC by ultracentrifugation to confirm source of GALNS enzyme. RESULTS Co-culture and confocal microscopy experiments indicated that the lysosomal content from UMSC migrated to deficient cells as evidenced by the peak signal intensity occurring at 15 min. EVs released by UMSC were characterized indicating that the EVs contained the active GALNS enzyme. Uptake of GALNS within EVs by deficient fibroblasts was not affected by mannose-6-phosphate (M6P) inhibition, suggesting that EV uptake by these fibroblasts is gradual and might be mediated by a different means than the M6P receptor. CONCLUSIONS UMSC can deliver EVs containing functional GALNS enzyme to deficient cells. This enzyme delivery method, which was unaffected by M6P inhibition, can function as a novel technique for reducing GAG accumulation in cells in avascular tissues, thereby providing a potential treatment option for Morquio A syndrome.
Collapse
Affiliation(s)
- Michael Flanagan
- Department of Pediatrics, School of Medicine, Saint Louis University, 1100 South Grand Blvd., Room 313, St. Louis, MO, 63104, USA
| | - Isha Pathak
- School of Medicine, Saint Louis University, Saint Louis, Missouri, USA
| | - Qi Gan
- Department of Pediatrics, School of Medicine, Saint Louis University, 1100 South Grand Blvd., Room 313, St. Louis, MO, 63104, USA
| | - Linda Winter
- Department of Pediatrics, School of Medicine, Saint Louis University, 1100 South Grand Blvd., Room 313, St. Louis, MO, 63104, USA
| | - Ryan Emnet
- St. Louis Cord Blood Bank, SSM Cardinal Glennon Children's Medical Center, St Louis, MO, USA
| | - Salem Akel
- St. Louis Cord Blood Bank, SSM Cardinal Glennon Children's Medical Center, St Louis, MO, USA
| | - Adriana M Montaño
- Department of Pediatrics, School of Medicine, Saint Louis University, 1100 South Grand Blvd., Room 313, St. Louis, MO, 63104, USA.
- Department of Biochemistry and Molecular Biology, School of Medicine, Saint Louis University, Saint Louis, Missouri, USA.
| |
Collapse
|
27
|
Medvecky L, Giretova M, Stulajterova R, Luptakova L, Sopcak T. Tetracalcium Phosphate/Monetite/Calcium Sulfate Hemihdrate Biocement Powder Mixtures Prepared by the One-Step Synthesis for Preparation of Nanocrystalline Hydroxyapatite Biocement-Properties and In Vitro Evaluation. MATERIALS 2021; 14:ma14092137. [PMID: 33922310 PMCID: PMC8122770 DOI: 10.3390/ma14092137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/28/2022]
Abstract
A modified one-step process was used to prepare tetracalcium phosphate/monetite/calcium sulfate hemihydrate powder cement mixtures (CAS). The procedure allowed the formation of monetite and calcium sulfate hemihydrate (CSH) in the form of nanoparticles. It was hypothesized that the presence of nanoCSH in small amounts enhances the in vitro bioactivity of CAS cement in relation to osteogenic gene markers in mesenchymal stem cells (MSCs). The CAS powder mixtures with 15 and 5 wt.% CSH were prepared by milling powder tetracalcium phosphate in an ethanolic solution of both orthophosphoric and sulfuric acids. The CAS cements had short setting times (around 5 min). The fast setting of the cement samples after the addition of the liquid component (water solution of NaH2PO4) was due to the partial formation of calcium sulfate dihydrate and hydroxyapatite before soaking in SBF with a small change in the original phase composition in cement powder samples after milling. Nanocrystalline hydroxyapatite biocement was produced by soaking of cement samples after setting in simulated body fluid (SBF). The fast release of calcium ions from CAS5 cement, as well as a small rise in the pH of SBF during soaking, were demonstrated. After soaking in SBF for 7 days, the final product of the cement transformation was nanocrystalline hydroxyapatite. The compressive strength of the cement samples (up to 30 MPa) after soaking in simulated body fluid (SBF) was comparable to that of bone. Real time polymerase chain reaction (RT-PCR) analysis revealed statistically significant higher gene expressions of alkaline phosphatase (ALP), osteonectin (ON) and osteopontin (OP) in cells cultured for 14 days in CAS5 extract compared to CSH-free cement. The addition of a small amount of nanoCSH (5 wt.%) to the tetracalcium phosphate (TTCP)/monetite cement mixture significantly promoted the over expression of osteogenic markers in MSCs. The prepared CAS powder mixture with its enhanced bioactivity can be used for bone defect treatment and has good potential for bone healing.
Collapse
Affiliation(s)
- Lubomir Medvecky
- Department of Functional and Hybrid Materials, Institute of Materials Research of SAS, Watsonova 47, 04 001 Kosice, Slovakia; (M.G.); (R.S.); (T.S.)
- Correspondence:
| | - Maria Giretova
- Department of Functional and Hybrid Materials, Institute of Materials Research of SAS, Watsonova 47, 04 001 Kosice, Slovakia; (M.G.); (R.S.); (T.S.)
| | - Radoslava Stulajterova
- Department of Functional and Hybrid Materials, Institute of Materials Research of SAS, Watsonova 47, 04 001 Kosice, Slovakia; (M.G.); (R.S.); (T.S.)
| | - Lenka Luptakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Tibor Sopcak
- Department of Functional and Hybrid Materials, Institute of Materials Research of SAS, Watsonova 47, 04 001 Kosice, Slovakia; (M.G.); (R.S.); (T.S.)
| |
Collapse
|
28
|
El-Habashy SE, Eltaher HM, Gaballah A, Zaki EI, Mehanna RA, El-Kamel AH. Hybrid bioactive hydroxyapatite/polycaprolactone nanoparticles for enhanced osteogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 119:111599. [PMID: 33321643 DOI: 10.1016/j.msec.2020.111599] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/20/2020] [Accepted: 09/23/2020] [Indexed: 02/05/2023]
Abstract
Hydroxyapatite nanoparticles (HApN) are largely employed as osteogenic inorganic material. Inorganic/polymeric hybrid nanostructures can provide versatile bioactivity for superior osteogenicity, particularly as nanoparticles. Herein, we present hybrid biomaterial-based hydroxyapatite/polycaprolactone nanoparticles (HAp/PCL NPs) realized using simple preparation techniques to augment HApN osteogenicity. Using wet chemical precipitation, we optimized HApN crystalline properties utilizing a 23-factorial design. Optimized HApN exhibited typical Ca/P elemental ratio with high reaction yield. Surface area analysis revealed their mesoporous nature and high surface area. Hybrid HAp/PCL NPs prepared using direct emulsification-solvent evaporation maintained HApN crystallinity with no observed chemical interactions. To the best of our knowledge, we are the first to elaborate the biocompatibility and osteogenicity of nanoparticulate hybrid HAp/PCL. Hybrid HAp/PCL NPs outperformed HApN regarding mesenchymal cell proliferation and osteodifferentiation with reduction of possible cytotoxicity. Unlike HApN, hybrid HAp/PCL NPs presented moderate expression of early osteogenic markers, Runx-2 and osteopontin and significantly elevated expression of the late osteogenic marker, bone sialoprotein after 10-day culture. Our results indicate that hybrid bioactive HAp/PCL NPs could offer a more prominent osteogenic potential than plain HApN for bone regenerative applications as a standalone nanoplatform or as part of complex engineered systems.
Collapse
Affiliation(s)
- Salma E El-Habashy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Hoda M Eltaher
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ahmed Gaballah
- Microbiology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Eiman I Zaki
- Histology and Cell Biology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Radwa A Mehanna
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications CERRMA, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amal H El-Kamel
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
29
|
Lo CH, Shay G, McGuire JJ, Li T, Shain KH, Choi JY, Fuerst R, Roush WR, Knapinska AM, Fields GB, Lynch CC. Host-Derived Matrix Metalloproteinase-13 Activity Promotes Multiple Myeloma-Induced Osteolysis and Reduces Overall Survival. Cancer Res 2021; 81:2415-2428. [PMID: 33526510 DOI: 10.1158/0008-5472.can-20-2705] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/02/2020] [Accepted: 01/25/2021] [Indexed: 11/16/2022]
Abstract
Multiple myeloma promotes systemic skeletal bone disease that greatly contributes to patient morbidity. Resorption of type I collagen-rich bone matrix by activated osteoclasts results in the release of sequestered growth factors that can drive progression of the disease. Matrix metalloproteinase-13 (MMP13) is a collagenase expressed predominantly in the skeleton by mesenchymal stromal cells (MSC) and MSC-derived osteoblasts. Histochemical analysis of human multiple myeloma specimens also demonstrated that MMP13 largely localizes to the stromal compartment compared with CD138+ myeloma cells. In this study, we further identified that multiple myeloma induces MMP13 expression in bone stromal cells. Because of its ability to degrade type I collagen, we examined whether bone stromal-derived MMP13 contributed to myeloma progression. Multiple myeloma cells were inoculated into wild-type or MMP13-null mice. In independent in vivo studies, MMP13-null mice demonstrated significantly higher overall survival rates and lower levels of bone destruction compared with wild-type controls. Unexpectedly, no differences in type I collagen processing between the groups were observed. Ex vivo stromal coculture assays showed reduced formation and activity in MMP13-null osteoclasts. Analysis of soluble factors from wild-type and MMP13-null MSCs revealed decreased bioavailability of various osteoclastogenic factors including CXCL7. CXCL7 was identified as a novel MMP13 substrate and regulator of osteoclastogenesis. Underscoring the importance of host MMP13 catalytic activity in multiple myeloma progression, we demonstrate the in vivo efficacy of a novel and highly selective MMP13 inhibitor that provides a translational opportunity for the treatment of this incurable disease. SIGNIFICANCE: Genetic and pharmacologic approaches show that bone stromal-derived MMP13 catalytic activity is critical for osteoclastogenesis, bone destruction, and disease progression. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/9/2415/F1.large.jpg.
Collapse
Affiliation(s)
- Chen Hao Lo
- Department of Tumor Biology, University of South Florida, Tampa, Florida.,Cancer Biology Ph.D. Program, Department of Cell Biology Microbiology and Molecular Biology, University of South Florida, Tampa, Florida
| | - Gemma Shay
- Department of Tumor Biology, University of South Florida, Tampa, Florida
| | - Jeremy J McGuire
- Department of Tumor Biology, University of South Florida, Tampa, Florida
| | - Tao Li
- Department of Tumor Biology, University of South Florida, Tampa, Florida
| | - Kenneth H Shain
- Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Jun Yong Choi
- Department of Chemistry and Biochemistry Queens College, Queens, New York.,Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York, New York, New York
| | - Rita Fuerst
- Department of Organic Chemistry, Graz University of Technology, Graz, Austria
| | - William R Roush
- Department of Chemistry, Scripps Research Institute, Jupiter, Florida
| | | | | | - Conor C Lynch
- Department of Tumor Biology, University of South Florida, Tampa, Florida.
| |
Collapse
|
30
|
Noueihed B, Rivera JC, Dabouz R, Abram P, Omri S, Lahaie I, Chemtob S. Mesenchymal Stromal Cells Promote Retinal Vascular Repair by Modulating Sema3E and IL-17A in a Model of Ischemic Retinopathy. Front Cell Dev Biol 2021; 9:630645. [PMID: 33553187 PMCID: PMC7859341 DOI: 10.3389/fcell.2021.630645] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic retinopathies (IRs), such as retinopathy of prematurity and diabetic retinopathy, are characterized by an initial phase of microvascular degeneration that results in retinal ischemia, followed by exaggerated pathologic neovascularization (NV). Mesenchymal stromal cells (MSCs) have potent pro-angiogenic and anti-inflammatory properties associated with tissue repair and regeneration, and in this regard exert protection to neurons in ischemic and degenerative conditions; however, the exact mechanisms underlying these functions remain largely unknown. Class III Semaphorins (A–G) are particularly implicated in regulating neural blood supply (as well as neurogenesis) by suppressing angiogenesis and affecting myeloid cell function; this is the case for distinct neuropillin-activating Sema3A as well as PlexinD1-activating Sema3E; but during IR the former Sema3A increases while Sema3E decreases. We investigated whether retinal vascular repair actions of MSCs are exerted by normalizing Semaphorin and downstream cytokines in IR. Intravitreal administration of MSCs or their secretome (MSCs-conditioned media [MSCs-CM]) significantly curtailed vasoobliteration as well as aberrant preretinal NV in a model of oxygen-induced retinopathy (OIR). The vascular repair effects of MSCs-CM in the ischemic retina were associated with restored levels of Sema3E. Vascular benefits of MSCs-CM were reversed by anti-Sema3E; while intravitreal injection of anti-angiogenic recombinant Sema3E (rSema3E) in OIR-subjected mice reproduced effects of MSCs-CM by inhibiting as expected preretinal NV but also by decreasing vasoobliteration. To explain these opposing vascular effects of Sema3E we found in OIR high retinal levels, respectively, of the pro- and anti-angiogenic IL-17A and Sema3A-regulating IL-1β; IL-17A positively affected expression of IL-1β. rSema3E decreased concentrations of these myeloid cell-derived pro-inflammatory cytokines in vitro and in vivo. Importantly, IL-17A suppression by MSCs-CM was abrogated by anti-Sema3E neutralizing antibody. Collectively, our findings provide novel evidence by which MSCs inhibit aberrant NV and diminish vasoobliteration (promoting revascularization) in retinopathy by restoring (at least in part) neuronal Sema3E levels that reduce pathological levels of IL-17A (and in turn other proinflammatory factors) in myeloid cells. The ability of MSCs to generate a microenvironment permissive for vascular regeneration by controlling the production of neuronal factors involved in immunomodulatory activities is a promising opportunity for stem cell therapy in ocular degenerative diseases.
Collapse
Affiliation(s)
- Baraa Noueihed
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - José Carlos Rivera
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Rabah Dabouz
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Pénélope Abram
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Samy Omri
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Isabelle Lahaie
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Sylvain Chemtob
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| |
Collapse
|
31
|
|
32
|
Johnstone BH, Miller HM, Beck MR, Gu D, Thirumala S, LaFontaine M, Brandacher G, Woods EJ. Identification and characterization of a large source of primary mesenchymal stem cells tightly adhered to bone surfaces of human vertebral body marrow cavities. Cytotherapy 2020; 22:617-628. [PMID: 32873509 PMCID: PMC8919862 DOI: 10.1016/j.jcyt.2020.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/12/2020] [Accepted: 07/05/2020] [Indexed: 12/13/2022]
Abstract
Background: Therapeutic allogeneic mesenchymal stromal cells (MSCs) are currently in clinical trials to evaluate their effectiveness in treating many different disease indications. Eventual commercialization for broad distribution will require further improvements in manufacturing processes to economically manufacture MSCs at scales sufficient to satisfy projected demands. A key contributor to the present high cost of goods sold for MSC manufacturing is the need to create master cell banks from multiple donors, which leads to variability in large-scale manufacturing runs. Therefore, the availability of large single donor depots of primary MSCs would greatly benefit the cell therapy market by reducing costs associated with manufacturing. Methods: We have discovered that an abundant population of cells possessing all the hallmarks of MSCs is tightly associated with the vertebral body (VB) bone matrix and only liberated by proteolytic digestion. Here we demonstrate that these vertebral bone-adherent (vBA) MSCs possess all the International Society of Cell and Gene Therapy-defined characteristics (e.g., plastic adherence, surface marker expression and trilineage differentiation) of MSCs, and we have therefore termed them vBA-MSCs to distinguish this population from loosely associated MSCs recovered through aspiration or rinsing of the bone marrow compartment. Results: Pilot banking and expansion were performed with vBA-MSCs obtained from 3 deceased donors, and it was demonstrated that bank sizes averaging 2.9 × 108 ± 1.35 × 108 vBA-MSCs at passage 1 were obtainable from only 5 g of digested VB bone fragments. Each bank of cells demonstrated robust proliferation through a total of 9 passages, without significant reduction in population doubling times. The theoretical total cell yield from the entire amount of bone fragments (approximately 300 g) from each donor with limited expansion through 4 passages is 100 trillion (1 × 1014) vBA-MSCs, equating to over 105 doses at 10 × 106 cells/kg for an average 70-kg recipient. Discussion: Thus, we have established a novel and plentiful source of MSCs that will benefit the cell therapy market by overcoming manufacturing and regulatory inefficiencies due to donor-to-donor variability.
Collapse
Affiliation(s)
- Brian H Johnstone
- Ossium Health, Inc, Indianapolis, Indiana, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA.
| | - Hannah M Miller
- Ossium Health, Inc, Indianapolis, Indiana, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA
| | - Madelyn R Beck
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dongsheng Gu
- Ossium Health, Inc, Indianapolis, Indiana, USA; Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sreedhar Thirumala
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael LaFontaine
- Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Erik J Woods
- Ossium Health, Inc, Indianapolis, Indiana, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
33
|
Avery SJ, Ayre WN, Sloan AJ, Waddington RJ. Interrogating the Osteogenic Potential of Implant SurfacesIn Vitro: A Review of Current Assays. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:217-229. [DOI: 10.1089/ten.teb.2019.0312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Steven James Avery
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Wayne Nishio Ayre
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Alastair James Sloan
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Rachel Jane Waddington
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
34
|
Aziz NS, Yusop N, Ahmad A. Importance of Stem Cell Migration and Angiogenesis Study for Regenerative Cell-based Therapy: A Review. Curr Stem Cell Res Ther 2020; 15:284-299. [DOI: 10.2174/1574888x15666200127145923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/01/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022]
Abstract
Stem cells play an essential role in maintaining homeostasis, as well as participating in new
tissue regeneration. Over the past 20 years, a great deal of effort has been made to investigate the behaviour
of stem cells to enable their potential use in regenerative medicine. However, a variety of biological
characteristics are known to exist among the different types of stem cells due to variations in
the methodological approach, formulation of cell culture medium, isolation protocol and cellular
niches, as well as species variation. In recent years, cell-based therapy has emerged as one of the advanced
techniques applied in both medical and clinical settings. Cell therapies aim to treat and repair
the injury sites and replace the loss of tissues by stimulating the repair and regeneration process. In
order to enable the use of stem cells in regenerative therapies, further characterisation of cell behaviour,
in terms of their proliferation and differentiation capacity, mainly during the quiescent and inductive
state is regarded as highly necessary. The central focus of regenerative medicine revolves around
the use of human cells, including adult stem cells and induced pluripotent stem cells for cell-based
therapy. The purpose of this review was to examine the existing body of literature on stem cell research
conducted on cellular angiogenesis and migration, to investigate the validity of different strategies and
variations of the cell type used. The information gathered within this review may then be shared with
fellow researchers to assist in future research work, engaging in stem cell homing for cell-based therapy
to enhance wound healing and tissue regeneration process.
Collapse
Affiliation(s)
- Nur S. Aziz
- Postgraduate Unit, School of Dentistry, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Norhayati Yusop
- Basic Sciences and Oral Biology Unit, School of Dentistry, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Azlina Ahmad
- Basic Sciences and Oral Biology Unit, School of Dentistry, Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
35
|
Mahmoud NS, Ahmed HH, Mohamed MR, Amr KS, Aglan HA, Ali MAM, Tantawy MA. Role of nanoparticles in osteogenic differentiation of bone marrow mesenchymal stem cells. Cytotechnology 2020; 72:1-22. [PMID: 31722051 PMCID: PMC7002803 DOI: 10.1007/s10616-019-00353-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/02/2019] [Indexed: 01/11/2023] Open
Abstract
The present study aimed to investigate the osteoinductive potentiality of some selected nanostructures; Hydroxyapatite (HA-NPs), Gold (Au-NPs), Chitosan (C-NPs), Gold/hydroxyapatite (Au/HA-NPs) and Chitosan/hydroxyapatite (CH-NPs) on bone marrow- derived mesenchymal stem cells (BM-MSCs). These nanostructures were characterized using transmission electron microscope and Zetasizer. MSCs were isolated from bone marrow of rat femur bones and their identity was documented by morphology, flow cytometry and multi-potency capacity. The influence of the selected nanostructures on the viability, osteogenic differentiation and subsequent matrix mineralization of BM-MSCs was determined by MTT assay, molecular genetic analysis and alizarin red S staining, respectively. MTT analysis revealed insignificant toxicity of the tested nanostructures on BM-MSCs at concentrations ranged from 2 to 25 µg/ml over 48 h and 72 h incubation period. Notably, the tested nanostructures potentiate the osteogenic differentiation of BM-MSCs as evidenced by a prominent over-expression of runt-related transcription factor 2 (Runx-2) and bone morphogenetic protein 2 (BMP-2) genes after 7 days incubation. Moreover, the tested nanostructures induced matrix mineralization of BM-MSCs after 21 days as manifested by the formation of calcium nodules stained with alizarin red S. Conclusively, these data provide a compelling evidence for the functionality of the studied nanostructures as osteoinductive materials motivating the differentiation of BM-MSCs into osteoblasts with the most prominent effect observed with Au-NPs and Au/HA-NPs, followed by CH-NPs.
Collapse
Affiliation(s)
- Nadia S. Mahmoud
- Hormones Department, Medical Research Division, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.), Dokki, Giza, P.O. 12622 Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Hanaa H. Ahmed
- Hormones Department, Medical Research Division, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.), Dokki, Giza, P.O. 12622 Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Mohamed R. Mohamed
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Khalda S. Amr
- Medical Molecular Genetics Department, Human Genetics and Genome Researches Division, National Research Centre, Dokki, Giza, Egypt
| | - Hadeer A. Aglan
- Hormones Department, Medical Research Division, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.), Dokki, Giza, P.O. 12622 Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Mohamed A. M. Ali
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohamed A. Tantawy
- Hormones Department, Medical Research Division, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.), Dokki, Giza, P.O. 12622 Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
36
|
Chu DT, Phuong TNT, Tien NLB, Tran DK, Thanh VV, Quang TL, Truong DT, Pham VH, Ngoc VTN, Chu-Dinh T, Kushekhar K. An Update on the Progress of Isolation, Culture, Storage, and Clinical Application of Human Bone Marrow Mesenchymal Stem/Stromal Cells. Int J Mol Sci 2020; 21:E708. [PMID: 31973182 PMCID: PMC7037097 DOI: 10.3390/ijms21030708] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Bone marrow mesenchymal stem/stromal cells (BMSCs), which are known as multipotent cells, are widely used in the treatment of various diseases via their self-renewable, differentiation, and immunomodulatory properties. In-vitro and in-vivo studies have supported the understanding mechanisms, safety, and efficacy of BMSCs therapy in clinical applications. The number of clinical trials in phase I/II is accelerating; however, they are limited in the size of subjects, regulations, and standards for the preparation and transportation and administration of BMSCs, leading to inconsistency in the input and outcome of the therapy. Based on the International Society for Cellular Therapy guidelines, the characterization, isolation, cultivation, differentiation, and applications can be optimized and standardized, which are compliant with good manufacturing practice requirements to produce clinical-grade preparation of BMSCs. This review highlights and updates on the progress of production, as well as provides further challenges in the studies of BMSCs, for the approval of BMSCs widely in clinical application.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam;
| | - Thuy Nguyen Thi Phuong
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju 61186, Korea
| | - Nguyen Le Bao Tien
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam; (N.L.B.T.); (V.V.T.)
| | - Dang Khoa Tran
- Department of Anatomy, University of Medicine Pham Ngoc Thach, Ho Chi Minh City 700000, Vietnam;
| | - Vo Van Thanh
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam; (N.L.B.T.); (V.V.T.)
- Department of Surgery, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Thuy Luu Quang
- Center for Anesthesia and Surgical Intensive Care, Viet Duc Hospital, Hanoi 100000, Vietnam;
| | | | - Van Huy Pham
- AI Lab, Faculty of Information Technology, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Vo Truong Nhu Ngoc
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam;
| | - Thien Chu-Dinh
- Institute for Research and Development, Duy Tan University, Danang 550000, Vietnam
| | - Kushi Kushekhar
- Institute of Cancer Research, Oslo University Hospital, 0310 Oslo, Norway;
| |
Collapse
|
37
|
Al-Qarakhli AMA, Yusop N, Waddington RJ, Moseley R. Effects of high glucose conditions on the expansion and differentiation capabilities of mesenchymal stromal cells derived from rat endosteal niche. BMC Mol Cell Biol 2019; 20:51. [PMID: 31752674 PMCID: PMC6873668 DOI: 10.1186/s12860-019-0235-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022] Open
Abstract
Background Mesenchymal stromal cells in the endosteal niche lining compact bone (CB-MSCs) represent a heterogeneous population, all of which contribute to bone repair and remodelling. Hyperglycaemia associated with type 2 diabetes mellitus (T2DM) can delay and impair the bone healing process. Therefore, this study investigated the influences of high (25 mM) glucose conditions on CB-MSC populations isolated from male Wistar rats, versus normal (5.5 mM) glucose conditions; in terms of proliferation (population doublings, PDs), senescence characteristics, stem cell marker expression, colony forming efficiencies (CFEs); and osteogenic/adipogenic differentiation, following extended culture in vitro. Results CB-MSCs under both normoglycaemic and hyperglycaemic conditions demonstrated similar morphologies and rapid exponential growth to >300PDs, although high glucose conditions promoted more rapid and persistent proliferation beyond ~50PDs, with few indications of senescence. Limited senescence was confirmed by minimal SA-β-galactosidase staining, low senescence marker (p53, p21waf1, p16INK4a) expression and positive telomere maintenance marker (rTERT, TR) expression. However, telomere lengths varied throughout culture expansion, with hyperglycaemia significantly reducing telomere lengths at PD50 and PD200. Furthermore, CB-MSCs expanded in normal and high glucose conditions remained non-transformed, exhibiting similar MSC (CD73/CD90/CD105), multipotency (CD146) and embryonic (Slug, Snail) markers throughout extended culture, but negligible hematopoietic (CD34/CD45) or pluripotency (Nanog, Oct4) markers. Hyperglycaemia significantly increased CFEs at PD50 and PD100, which decreased at PD200. CB-MSC osteogenic differentiation was also inhibited by hyperglycaemia at PD15, PD100 and PD200, but not at PD50. Hyperglycaemia inhibited CB-MSC adipogenic differentiation to a lesser extent at PD15 and PD50, with reduced adipogenesis overall at PD100 and PD200. Conclusion This study demonstrates the limited negative impact of hyperglycaemia on the proliferative and stem cell characteristics of heterogeneous CB-MSC populations, although minor sub-population(s) appear more susceptible to these conditions leading to impaired osteogenic/adipogenic differentiation capabilities. Such findings potentially highlight the impact of hyperglycaemia on CB-MSC bone repair capabilities in situ.
Collapse
Affiliation(s)
- Ahmed Makki A Al-Qarakhli
- School of Dentistry, Cardiff Institute of Tissue Engineering and Repair (CITER), College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK.,College of Dentistry, University of Anbar, Anbar, Iraq
| | - Norhayati Yusop
- School of Dentistry, Cardiff Institute of Tissue Engineering and Repair (CITER), College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK.,School of Dental Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Rachel J Waddington
- School of Dentistry, Cardiff Institute of Tissue Engineering and Repair (CITER), College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK
| | - Ryan Moseley
- School of Dentistry, Cardiff Institute of Tissue Engineering and Repair (CITER), College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK.
| |
Collapse
|
38
|
Improved Isolation of Mesenchymal Stem Cells Based on Interactions between N-Acetylglucosamine-Bearing Polymers and Cell-Surface Vimentin. Stem Cells Int 2019; 2019:4341286. [PMID: 31814834 PMCID: PMC6878802 DOI: 10.1155/2019/4341286] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/13/2019] [Accepted: 09/17/2019] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) in bone marrow and adipose tissues are expected to be effective tools for regenerative medicine to treat various diseases. To obtain MSCs that possess both high differentiation and tissue regenerative potential, it is necessary to establish an isolation system that does not require long-term culture. It has previously been reported that the cytoskeletal protein vimentin, expressed on the surfaces of multiple cell types, possesses N-acetylglucosamine- (GlcNAc-) binding activity. Therefore, we tried to exploit this interaction to efficiently isolate MSCs from rat bone marrow cells using GlcNAc-bearing polymer-coated dishes. Cells isolated by this method were identified as MSCs because they were CD34-, CD45-, and CD11b/c-negative and CD90-, CD29-, CD44-, CD54-, CD73-, and CD105-positive. Osteoblast, adipocyte, and chondrocyte differentiation was observed in these cells. In total, yields of rat MSCs were threefold to fourfold higher using GlcNAc-bearing polymer-coated dishes than yields using conventional tissue-culture dishes. Interestingly, MSCs isolated with GlcNAc-bearing polymer-coated dishes strongly expressed CD106, whereas those isolated with conventional tissue-culture dishes had low CD106 expression. Moreover, senescence-associated β-galactosidase activity in MSCs from GlcNAc-bearing polymer-coated dishes was lower than that in MSCs from tissue-culture dishes. These results establish an improved isolation method for high-quality MSCs.
Collapse
|
39
|
Mesenchymal Progenitors Derived from Different Locations in Long Bones Display Diverse Characteristics. Stem Cells Int 2019; 2019:5037578. [PMID: 31089329 PMCID: PMC6476036 DOI: 10.1155/2019/5037578] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 11/20/2022] Open
Abstract
Mesenchymal progenitors within bone marrow have multiple differentiation potential and play an essential role in the maintenance of adult skeleton homeostasis. Mesenchymal progenitors located in bone regions other than the bone marrow also display bone-forming properties. However, owing to the differences in each distinct microenvironment, the mesenchymal characteristics of skeletal progenitor cells within different regions of long bones may show some differences. In order to clearly elucidate these differences, we performed a comparative study on mesenchymal progenitors from different regions of long bones. Here, we isolated mesenchymal progenitors from the periosteum, endosteum, and bone marrow of rat long bones. The three groups exhibited similar cellular morphologies and expressed the typical surface markers associated with mesenchymal stem cells. Interestingly, after cell proliferation assays and bidirectional differentiation analysis, periosteal mesenchymal progenitors showed a higher proliferative ability and adipogenic differentiation potential. In contrast, endosteal mesenchymal progenitors were more prone to osteogenic differentiation. Using in vitro osteoclast culture systems, conditioned media from different mesenchymal progenitor cultures were used to induce osteoclastic differentiation. Osteoclast formation was found to be significantly promoted by the secretion of RANKL and IL-6 by endosteal progenitors. Overall, our results provide strong evidence for the importance of selecting the appropriate source of skeletal progenitors for applications in future skeleton regeneration therapies.
Collapse
|