1
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
2
|
Humpfle L, Hachem NE, Simon P, Weinhold B, Galuska SP, Middendorff R. Knockout of the polysialyltransferases ST8SiaII and ST8SiaIV leads to a dilatation of rete testis during postnatal development. Front Physiol 2023; 14:1240296. [PMID: 37520830 PMCID: PMC10382229 DOI: 10.3389/fphys.2023.1240296] [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: 06/14/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Polysialic acid (polySia) is a carbohydrate polymer that modulates several cellular processes, such as migration, proliferation and differentiation processes. In the brain, its essential impact during postnatal development is well known. However, in most other polySia positive organs, only its localization has been described so far. For instance, in the murine epididymis, smooth muscle cells of the epididymal duct are polysialylated during the first 2 weeks of postnatal development. To understand the role of polySia during the development of the epididymis, the consequences of its loss were investigated in postnatal polySia knockout mice. As expected, no polysialylation was visible in the absence of the polysialyltransferases ST8SiaII and ST8SiaIV. Interestingly, cGMP-dependent protein kinase I (PGK1), which is essentially involved in smooth muscle cell relaxation, was not detectable in peritubular smooth muscle cells when tissue sections of polySia knockout mice were analyzed by immunohistochemistry. In contrast to this signaling molecule, the structural proteins smooth muscle actin (SMA) and calponin were expressed. As shown before, in the duct system of the testis, even the expression of these structural proteins was impaired due to the loss of polySia. We now found that the rete testis, connecting the duct system of the testis and epididymis, was extensively dilated. The obtained data suggest that less differentiated smooth muscle cells of the testis and epididymis result in disturbed contractility and thus, fluid transport within the duct system visible in the enlarged rete testis.
Collapse
Affiliation(s)
- Luisa Humpfle
- Institute of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Giessen, Germany
| | - Nadim E. Hachem
- Institute of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Giessen, Germany
| | - Peter Simon
- Institute of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Giessen, Germany
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Giessen, Germany
| | - Birgit Weinhold
- Institute of Cellular Chemistry, Hannover Medical School, Hannover, Germany
| | | | - Ralf Middendorff
- Institute of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
3
|
Kamaraj M, Giri PS, Mahapatra S, Pati F, Rath SN. Bioengineering strategies for 3D bioprinting of tubular construct using tissue-specific decellularized extracellular matrix. Int J Biol Macromol 2022; 223:1405-1419. [PMID: 36375675 DOI: 10.1016/j.ijbiomac.2022.11.064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
The goal of the current study is to develop an extracellular matrix bioink that could mimic the biochemical components present in natural blood vessels. Here, we have used an innovative approach to recycle the discarded varicose vein for isolation of endothelial cells and decellularization of the same sample to formulate the decellularized extracellular matrix (dECM) bioink. The shift towards dECM bioink observed as varicose vein dECM provides the tissue-specific biochemical factors that will enhance the regeneration capability. Interestingly, the encapsulated umbilical cord mesenchymal stem cells expressed the markers of vascular smooth muscle cells because of the cues present in the vein dECM. Further, in vitro immunological investigation of dECM revealed a predominant M2 polarization which could further aid in tissue remodeling. A novel approach was used to fabricate vascular construct using 3D bioprinting without secondary support. The outcomes suggest that this could be a potential approach for patient- and tissue-specific blood vessel regeneration.
Collapse
Affiliation(s)
- Meenakshi Kamaraj
- Regenerative Medicine and Stem cell (RMS) Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Pravin Shankar Giri
- Regenerative Medicine and Stem cell (RMS) Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Sandeep Mahapatra
- Vascular & Endovascular Surgery, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Falguni Pati
- BioFabTE Lab, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Subha Narayan Rath
- Regenerative Medicine and Stem cell (RMS) Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India.
| |
Collapse
|
4
|
Lechner J, Medina RJ, Lois N, Stitt AW. Advances in cell therapies using stem cells/progenitors as a novel approach for neurovascular repair of the diabetic retina. Stem Cell Res Ther 2022; 13:388. [PMID: 35907890 PMCID: PMC9338609 DOI: 10.1186/s13287-022-03073-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/20/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Diabetic retinopathy, a major complication of diabetes mellitus, is a leading cause of sigh-loss in working age adults. Progressive loss of integrity of the retinal neurovascular unit is a central element in the disease pathogenesis. Retinal ischemia and inflammatory processes drive interrelated pathologies such as blood retinal barrier disruption, fluid accumulation, gliosis, neuronal loss and/or aberrant neovascularisation. Current treatment options are somewhat limited to late-stages of the disease where there is already significant damage to the retinal architecture arising from degenerative, edematous and proliferative pathology. New preventive and interventional treatments to target early vasodegenerative and neurodegenerative stages of the disease are needed to ensure avoidance of sight-loss. MAIN BODY Historically, diabetic retinopathy has been considered a primarily microvascular disease of the retina and clinically it is classified based on the presence and severity of vascular lesions. It is now known that neurodegeneration plays a significant role during the pathogenesis. Loss of neurons has been documented at early stages in pre-clinical models as well as in individuals with diabetes and, in some, even prior to the onset of clinically overt diabetic retinopathy. Recent studies suggest that some patients have a primarily neurodegenerative phenotype. Retinal pigment epithelial cells and the choroid are also affected during the disease pathogenesis and these tissues may also need to be addressed by new regenerative treatments. Most stem cell research for diabetic retinopathy to date has focused on addressing vasculopathy. Pre-clinical and clinical studies aiming to restore damaged vasculature using vasoactive progenitors including mesenchymal stromal/stem cells, adipose stem cells, CD34+ cells, endothelial colony forming cells and induced pluripotent stem cell derived endothelial cells are discussed in this review. Stem cells that could replace dying neurons such as retinal progenitor cells, pluripotent stem cell derived photoreceptors and ganglion cells as well as Müller stem cells are also discussed. Finally, challenges of stem cell therapies relevant to diabetic retinopathy are considered. CONCLUSION Stem cell therapies hold great potential to replace dying cells during early and even late stages of diabetic retinopathy. However, due to the presence of different phenotypes, selecting the most suitable stem cell product for individual patients will be crucial for successful treatment.
Collapse
Affiliation(s)
- Judith Lechner
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK.
| | - Reinhold J Medina
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Noemi Lois
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Alan W Stitt
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
5
|
Wen Y, Kong Y, Cao G, Xu Y, Zhang C, Zhang J, Xiao P, Wang Y. Di-n-butyl phthalate regulates vascular smooth muscle cells phenotypic switching by MiR-139-5p-MYOCD pathways. Toxicology 2022; 477:153279. [PMID: 35926758 DOI: 10.1016/j.tox.2022.153279] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/20/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022]
Abstract
Di-n-butyl phthalate (DBP) is ubiquitous in environment and has been detected in almost all human bodies. Few data could be found about the effects of DBP on cardiovascular system, though its reproductive toxicities have been studied extensively. This study aimed to explore effects of DBP on phenotypic switching of vascular smooth muscle cells (VSMCs), an essential step during the formation of atherosclerosis (AS). A7r5 cells were employed and exposed to various levels of DBP (10-9, 10-8, 10-7, 10-6, and 10-5 M) or DMSO as control. CCK-8 assay was used to detect the effects of DBP on cell viability. Expressions of mRNA/miRNAs and proteins were measured by qRT-PCR and western blotting, respectively. Bioinformatic analysis and dual-luciferase reporter assay were used to analyze the combination between miR-139-5p and Myocardin (MYOCD). Results revealed that DBP at 10-7 M prompted phenotypic switching from contractile to synthetic of VSMCs by inhibiting contractile VSMCs marker genes via suppressing the expression of MYOCD. Moreover, miR-139c-5p directly targeted MYOCD 3'UTR and modulated MYOCD expression. Besides, DBP inhibited the expression of MYOCD and VSMCs marker genes by upregulating miR-139-5p. Collectively, these data suggested that DBP could promote the phenotypic switching from contractile to synthetic of VSMCs in A7r5 cells through miR-139-5p-MYOCD.
Collapse
Affiliation(s)
- Yun Wen
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Yi Kong
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Guofa Cao
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Yuan Xu
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Chengxiang Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Jingshu Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Pingxi Xiao
- The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yubang Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
6
|
Mayoral I, Bevilacqua E, Gómez G, Hmadcha A, González-Loscertales I, Reina E, Sotelo J, Domínguez A, Pérez-Alcántara P, Smani Y, González-Puertas P, Mendez A, Uribe S, Smani T, Ordoñez A, Valverde I. Tissue engineered in-vitro vascular patch fabrication using hybrid 3D printing and electrospinning. Mater Today Bio 2022; 14:100252. [PMID: 35509864 PMCID: PMC9059085 DOI: 10.1016/j.mtbio.2022.100252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 11/05/2022] Open
Abstract
Three-dimensional (3D) engineered cardiovascular tissues have shown great promise to replace damaged structures. Specifically, tissue engineering vascular grafts (TEVG) have the potential to replace biological and synthetic grafts. We aimed to design an in-vitro patient-specific patch based on a hybrid 3D print combined with vascular smooth muscle cells (VSMC) differentiation. Based on the medical images of a 2 months-old girl with aortic arch hypoplasia and using computational modelling, we evaluated the most hemodynamically efficient aortic patch surgical repair. Using the designed 3D patch geometry, the scaffold was printed using a hybrid fused deposition modelling (FDM) and electrospinning techniques. The scaffold was seeded with multipotent mesenchymal stem cells (MSC) for later maturation to derived VSMC (dVSMC). The graft showed adequate resistance to physiological aortic pressure (burst pressure 101 ± 15 mmHg) and a porosity gradient ranging from 80 to 10 μm allowing cells to infiltrate through the entire thickness of the patch. The bio-scaffolds showed good cell viability at days 4 and 12 and adequate functional vasoactive response to endothelin-1. In summary, we have shown that our method of generating patient-specific patch shows adequate hemodynamic profile, mechanical properties, dVSMC infiltration, viability and functionality. This innovative 3D biotechnology has the potential for broad application in regenerative medicine and potentially in heart disease prevention.
Collapse
Key Words
- 3D printing
- Electrospinning
- Endothelin Receptor A, ETA
- Endothelin Receptor B, ETB
- Mesenchymal stem cells
- Reverse Transcription, Rt
- Three-dimensional, 3D
- Tissue engineering
- Vascular graft
- anti-alpha-smooth muscle actin, α-SMA
- anti-cluster of differentiation 31, CD31
- anti-fibroblast specific protein 1, FSP1
- anti-smooth muscle protein 22, SM-22
- bone morphogenetic protein, BMP4
- computation fluid dynamic, CFD
- computed tomography, CT
- derived VSMC, dVSMC
- endothelin-1, ET-1
- extracellular matrix, ECM
- fused deposition modelling, FDM
- mesenchymal stem cells, MSC
- platelet-derived growth factor composed by two beta chains, PDGF-BB
- room temperature, RT
- tissue engineering vascular grafts, TEVG
- transforming growth factor beta 1, TGFβ-1
- vascular smooth muscle cells, VSMC
- wall shear stress, WSS
- western blotting, WB
Collapse
Affiliation(s)
- Isabel Mayoral
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville- IBiS, University of Seville /HUVR/CSIC, Seville, Spain
| | - Elisa Bevilacqua
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville- IBiS, University of Seville /HUVR/CSIC, Seville, Spain
| | - Gorka Gómez
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville- IBiS, University of Seville /HUVR/CSIC, Seville, Spain
| | - Abdelkrim Hmadcha
- Advanced Therapies and Regenerative Medicine Research Group.General Hospital, Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain
| | - Ignacio González-Loscertales
- Department Mechanical, Thermal and Fluids Engineering, School of Engineering, University of Málaga, Málaga, Spain
| | - Esther Reina
- Department of Mechanical and Manufacturing Engineering, University of Seville, Seville, Spain
| | - Julio Sotelo
- School of Biomedical Engineering, Universidad de Valparaíso, Valparaíso, Chile
- Millennium Institute for Intelligent Healthcare Engineering, iHEALTH, Millennium Nucleus in Cardiovascular Magnetic Resonance, Cardio MR, and Biomedical Imaging Center, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Pedro Pérez-Alcántara
- Department of Mechanical and Manufacturing Engineering, University of Seville, Seville, Spain
| | - Younes Smani
- Department of Molecular Biology and Biochemical Engineering, Andalusian Center of Developmental Biology, CSIC, University of Pablo de Olavide, Seville, Spain
| | | | - Ana Mendez
- Pediatric Cardiology Unit, Hospital Virgen Del Rocio, Seville, Spain
| | - Sergio Uribe
- Millennium Institute for Intelligent Healthcare Engineering, iHEALTH, Millennium Nucleus in Cardiovascular Magnetic Resonance, Cardio MR, and Biomedical Imaging Center, Pontificia Universidad Católica de Chile, Santiago, Chile
- Radiology Department, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tarik Smani
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville- IBiS, University of Seville /HUVR/CSIC, Seville, Spain
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, Seville, Spain
| | - Antonio Ordoñez
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville- IBiS, University of Seville /HUVR/CSIC, Seville, Spain
| | - Israel Valverde
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville- IBiS, University of Seville /HUVR/CSIC, Seville, Spain
- Pediatric Cardiology Unit, Hospital Virgen Del Rocio, Seville, Spain
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Department of Pharmacology, Pediatric and Radiology, School of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
7
|
Ma S, Wu J, Hu H, Mu Y, Zhang L, Zhao Y, Bian X, Jing W, Wei P, Zhao B, Deng J, Liu Z. Novel fusion peptides deliver exosomes to modify injectable thermo-sensitive hydrogels for bone regeneration. Mater Today Bio 2022; 13:100195. [PMID: 35024598 PMCID: PMC8724941 DOI: 10.1016/j.mtbio.2021.100195] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/20/2022] Open
Abstract
Injectable thermo-sensitive hydrogels composed of small intestinal submucosa (SIS) with exosomes derived from bone marrow mesenchymal stem cells (BMSCs) are desired for bone regeneration. However, poor mechanical properties limit the clinical application of SIS hydrogels. Herein, the mechanical properties of SIS hydrogels incorporated with 3-(3,4-dihydroxyphenyl) propionic acid (CA) are assessed. The results show that the mechanical properties of SIS hydrogels are improved. In addition, the retention and stability of exosomes over time at the defect site are also challenges. Fusion peptides are designed by connecting collagen-binding domines (CBDs) of collagen type I/III with exosomal capture peptides CP05 (CRHSQMTVTSRL) directly or via rigid linkers (EAAAK). In vitro experiments demonstrate that fusion peptides are contribute to promoting the positive effect of exosomes on osteogenic differentiation of BMSCs. Meanwhile, the results of hydrogels combining exosomes and fusion peptides in the treatment of rat skull defect models reveal that fusion peptides could enhance the retention and stability of exosomes, thereby strengthen the therapeutic effect for skull defects. Therefore, SIS hydrogels with CA modified by fusion peptides and exosomes appear to be a promising strategy in bone regenerative medicine.
Collapse
Affiliation(s)
- Shiqing Ma
- Department of Stomotology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Jinzhe Wu
- School and Hospital of Stomatology, Tianjin Medical University, 12 Observatory Road, Tianjin, 300070, China
| | - Han Hu
- School and Hospital of Stomatology, Tianjin Medical University, 12 Observatory Road, Tianjin, 300070, China
| | - Yuzhu Mu
- School and Hospital of Stomatology, Tianjin Medical University, 12 Observatory Road, Tianjin, 300070, China
| | - Lei Zhang
- School and Hospital of Stomatology, Tianjin Medical University, 12 Observatory Road, Tianjin, 300070, China
| | - Yifan Zhao
- School and Hospital of Stomatology, Tianjin Medical University, 12 Observatory Road, Tianjin, 300070, China
| | - Xiaowei Bian
- School and Hospital of Stomatology, Tianjin Medical University, 12 Observatory Road, Tianjin, 300070, China
| | - Wei Jing
- Beijing Biosis Healing Biological Technology Co., Ltd., No. 6 Plant West, Valley No. 1 Bio-medicine Industry Park, Beijing, 102600, China
- Foshan (Southern China) Institute for New Materials, Foshan, 528220, China
| | - Pengfei Wei
- Beijing Biosis Healing Biological Technology Co., Ltd., No. 6 Plant West, Valley No. 1 Bio-medicine Industry Park, Beijing, 102600, China
| | - Bo Zhao
- Beijing Biosis Healing Biological Technology Co., Ltd., No. 6 Plant West, Valley No. 1 Bio-medicine Industry Park, Beijing, 102600, China
| | - Jiayin Deng
- School and Hospital of Stomatology, Tianjin Medical University, 12 Observatory Road, Tianjin, 300070, China
| | - Zihao Liu
- School and Hospital of Stomatology, Tianjin Medical University, 12 Observatory Road, Tianjin, 300070, China
| |
Collapse
|
8
|
Walters B, Turner PA, Rolauffs B, Hart ML, Stegemann JP. Controlled Growth Factor Delivery and Cyclic Stretch Induces a Smooth Muscle Cell-like Phenotype in Adipose-Derived Stem Cells. Cells 2021; 10:cells10113123. [PMID: 34831345 PMCID: PMC8624888 DOI: 10.3390/cells10113123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are an abundant and easily accessible multipotent stem cell source with potential application in smooth muscle regeneration strategies. In 3D collagen hydrogels, we investigated whether sustained release of growth factors (GF) PDGF-AB and TGF-β1 from GF-loaded microspheres could induce a smooth muscle cell (SMC) phenotype in ASCs, and if the addition of uniaxial cyclic stretch could enhance the differentiation level. This study demonstrated that the combination of cyclic stretch and GF release over time from loaded microspheres potentiated the differentiation of ASCs, as quantified by protein expression of early to late SMC differentiation markers (SMA, TGLN and smooth muscle MHC). The delivery of GFs via microspheres produced large ASCs with a spindle-shaped, elongated SMC-like morphology. Cyclic strain produced the largest, longest, and most spindle-shaped cells regardless of the presence or absence of growth factors or the growth factor delivery method. Protein expression and cell morphology data confirmed that the sustained release of GFs from GF-loaded microspheres can be used to promote the differentiation of ASCs into SMCs and that the addition of uniaxial cyclic stretch significantly enhances the differentiation level, as quantified by intermediate and late SMC markers and a SMC-like elongated cell morphology.
Collapse
Affiliation(s)
- Brandan Walters
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Paul A. Turner
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Bernd Rolauffs
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
| | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| |
Collapse
|
9
|
Yu H, Hua Y, He Y, Wang Y, Hu X, Chen S, Liu J, Yang J, Li H. Sustained Release of MiR-217 Inhibitor by Nanoparticles Facilitates MSC-Mediated Attenuation of Neointimal Hyperplasia After Vascular Injury. Front Cardiovasc Med 2021; 8:739107. [PMID: 34708092 PMCID: PMC8542691 DOI: 10.3389/fcvm.2021.739107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/13/2021] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been proven capable of differentiating into endothelial cells (ECs) and increasing vascular density in mouse ischemia models. However, the therapeutic potential of MSCs in neointimal hyperplasia after vascular injury is still not fully understood. In this study, we proposed that sustained release of miR-217 inhibitor encapsulated by nanoparticles in MSCs can enhance the therapeutic effects of MSCs on alleviating neointimal hyperplasia in a standard mouse wire injury model. We intravenously administered MSCs to mice with injured arteries and examined neointimal proliferation, endothelial differentiation and senescence. We demonstrated that MSCs localized to the luminal surface of the injured artery within 24 h after injection and subsequently differentiated into endothelial cells, inhibited neointimal proliferation and migration of vascular smooth muscle cells. Transfection of MSCs with poly lactic-co-glycolic acid nanoparticles (PLGA-NP) encapsulating an miR-217 agomir abolished endothelial differentiation as well as the therapeutic effect of MSCs. On the contrary, silencing of endogenous miR-217 improved the therapeutic efficacy of MSCs. Our study provides a new strategy of augmenting the therapeutic potency of MSCs in treatment of vascular injury.
Collapse
Affiliation(s)
- Hong Yu
- Department of Otorhinolaryngology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yutao Hua
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yecheng He
- Department of Clinical Medicine, Suzhou Vocational Health College, Suzhou, China
| | - Yin Wang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjian Hu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Si Chen
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Junwei Liu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Yang
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Huadong Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Qi Y, Liang X, Guan H, Sun J, Yao W. RhoGDI1-Cdc42 Signaling Is Required for PDGF-BB-Induced Phenotypic Transformation of Vascular Smooth Muscle Cells and Neointima Formation. Biomedicines 2021; 9:biomedicines9091169. [PMID: 34572355 PMCID: PMC8470270 DOI: 10.3390/biomedicines9091169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 11/24/2022] Open
Abstract
RhoGTPase is involved in PDGF-BB-mediated VSMC phenotypic modulation. RhoGDIs are key factors in regulating RhoGTPase activation. In the present study, we investigated the regulatory effect of RhoGDI1 on the activation of RhoGTPase in VSMC transformation and neointima formation. Western blot and co-immunoprecipitation assays showed that the PDGF receptor inhibition by crenolanib promoted RhoGDI1 polyubiquitination and degradation. Inhibition of RhoGDI1 degradation via MG132 reversed the decrease in VSMC phenotypic transformation. In addition, RhoGDI1 knockdown significantly inhibited VSMC phenotypic transformation and neointima formation in vitro and in vivo. These results suggest that PDGF-BB promotes RhoGDI1 stability via the PDGF receptor and induces the VSMC synthetic phenotype. The co-immunoprecipitation assay showed that PDGF-BB enhanced the interaction of RhoGDI1 with Cdc42 and promoted the activation of Cdc42; these enhancements were blocked by crenolanib and RhoGDI1 knockdown. Moreover, RhoGDI1 knockdown and crenolanib pretreatment prevented the localization of Cdc42 to the plasma membrane (PM) to activate and improve the accumulation of Cdc42 on endoplasmic reticulum (ER). Furthermore, Cdc42 inhibition or suppression significantly reduced VSMC phenotypic transformation and neointima formation in vitro and in vivo. This study revealed the novel mechanism by which RhoGDI1 stability promotes the RhoGDI1-Cdc42 interaction and Cdc42 activation, thereby affecting VSMC phenotypic transformation and neointima formation.
Collapse
Affiliation(s)
| | | | | | | | - Wenjuan Yao
- Correspondence: ; Tel.: +86-513-8505-1728; Fax: +86-513-8505-1858
| |
Collapse
|
11
|
Yamanaka H, Mahara A, Morimoto N, Yamaoka T. REDV-modified decellularized microvascular grafts for arterial and venous reconstruction. J Biomed Mater Res A 2021; 110:547-558. [PMID: 34486215 DOI: 10.1002/jbm.a.37305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/02/2021] [Accepted: 08/26/2021] [Indexed: 11/09/2022]
Abstract
Recently, a decellularized microvascular graft (inner diameter: 0.6 mm) modified with the integrin α4β1 ligand, REDV, was developed to provide an alternative to autologous-vein grafting in reconstructive microsurgery, showing good early-stage patency under arterial flow in rats. This consecutive study evaluated its potential utility not only as an arterial substitute, but also as a venous substitute, using a rat-tail replantation model. Graft remodeling depending on hemodynamic status was also investigated. ACI rat tail arteries were decellularized via ultra-high-hydrostatic pressure treatment and modified with REDV to induce antithrombogenic interfaces and promote endothelialization after implantation. Grafts were implanted into the tail artery and vein to re-establish blood circulation in amputated Lewis rat tails (n = 12). The primary endpoint was the survival of replants. Secondary endpoints were graft patency, remodeling, and regeneration for 6 months. In all but three cases with technical errors or postoperative self-mutilation, tails survived without any evidence of ischemia or congestion. Six-month Kaplan-Meier patency was 100% for tail-artery implanted grafts and 62% for tail-vein implanted grafts. At 6 months, the neo-tunica media (thickness: 95.0 μm in tail-artery implanted grafts, 9.3 μm in tail-vein implanted grafts) was regenerated inside the neo-intima. In conclusion, the microvascular grafts functioned well both as arterial and venous paths of replanted-rat tails, with different remodeling under arterial and venous conditions.
Collapse
Affiliation(s)
- Hiroki Yamanaka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan.,Department of Plastic and Reconstructive Surgery, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Naoki Morimoto
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| |
Collapse
|
12
|
Schuster M, Tewary G, Bao X, Subedi P, Hauck SM, Olsen AK, Eide DM, Trott KR, Götz S, Atkinson MJ, Rosemann M. In vitro cellular and proteome assays identify Wnt pathway and CDKN2A-regulated senescence affected in mesenchymal stem cells from mice after a chronic LD gamma irradiation in utero. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2021; 60:397-410. [PMID: 34287697 PMCID: PMC8310520 DOI: 10.1007/s00411-021-00925-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
Reliable data on the effects of chronic prenatal exposure to low dose (LD) of ionizing radiation in humans are missing. There are concerns about adverse long-term effects that may persist throughout postnatal life of the offspring. Due to their slow cell cycle kinetics and life-long residence time in the organism, mesenchymal stem cells (MSCs) are more susceptible to low level genotoxic stress caused by extrinsic multiple LD events. The aim of this study was to investigate the effect of chronic, prenatal LD gamma irradiation to the biology of MSCs later in life. C3H mice were exposed in utero to chronic prenatal irradiation of 10 mGy/day over a period of 3 weeks. Two years later, MSCs were isolated from the bone marrow and analyzed in vitro for their radiosensitivity, for cellular senescence and for DNA double-strand break recognition after a second acute gamma-irradiation. In addition to these cellular assays, changes in protein expression were measured using HPLC-MS/MS and dysregulated molecular signaling pathways identified using bioinformatics. We observed radiation-induced proteomic changes in MSCs from the offspring of in utero irradiated mice (leading to ~ 9.4% of all detected proteins being either up- or downregulated) as compared to non-irradiated controls. The proteomic changes map to regulation pathways involved in the extracellular matrix, the response to oxidative stress, and the Wnt signaling pathway. In addition, chronic prenatal LD irradiation lead to an increased rate of in vitro radiation-induced senescence later in life and to an increased number of residual DNA double-strand breaks after 4 Gy irradiation, indicating a remarkable interaction of in vivo radiation in combination with a second acute dose of in vitro radiation. This study provides the first insight into a molecular mechanism of persistent MSC damage response by ionizing radiation exposure during prenatal time and will help to predict therapeutic safety and efficacy with respect to a clinical application of stem cells.
Collapse
Affiliation(s)
- Martina Schuster
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Gargi Tewary
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Xuanwen Bao
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Prabal Subedi
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), 80939, Munich, Germany
| | - Ann Karin Olsen
- Department of Molecular Biology/Domain for Infection Control and Environmental Health, Norwegian Institute of Public Health, Lovisenberggt. 8, 0456, Oslo, Norway
| | - Dag Markus Eide
- Department of Molecular Biology/Domain for Infection Control and Environmental Health, Norwegian Institute of Public Health, Lovisenberggt. 8, 0456, Oslo, Norway
| | - Klaus Rüdiger Trott
- Chair of Radiation Biology, Technical University Munich (TUM), 80333, Munich, Germany
| | - Sebastian Götz
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
- Chair of Radiation Biology, Technical University Munich (TUM), 80333, Munich, Germany
| | - Michael Rosemann
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health GmbH (HMGU), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany.
- Medical Graduate School, Technical University Munich (TUM), 80333, Munich, Germany.
| |
Collapse
|
13
|
Romano E, Rosa I, Fioretto BS, Matucci-Cerinic M, Manetti M. New Insights into Profibrotic Myofibroblast Formation in Systemic Sclerosis: When the Vascular Wall Becomes the Enemy. Life (Basel) 2021; 11:610. [PMID: 34202703 PMCID: PMC8307837 DOI: 10.3390/life11070610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
In systemic sclerosis (SSc), abnormalities in microvessel morphology occur early and evolve into a distinctive vasculopathy that relentlessly advances in parallel with the development of tissue fibrosis orchestrated by myofibroblasts in nearly all affected organs. Our knowledge of the cellular and molecular mechanisms underlying such a unique relationship between SSc-related vasculopathy and fibrosis has profoundly changed over the last few years. Indeed, increasing evidence has suggested that endothelial-to-mesenchymal transition (EndoMT), a process in which profibrotic myofibroblasts originate from endothelial cells, may take center stage in SSc pathogenesis. While in arterioles and small arteries EndoMT may lead to the accumulation of myofibroblasts within the vessel wall and development of fibroproliferative vascular lesions, in capillary vessels it may instead result in vascular destruction and formation of myofibroblasts that migrate into the perivascular space with consequent tissue fibrosis and microvessel rarefaction, which are hallmarks of SSc. Besides endothelial cells, other vascular wall-resident cells, such as pericytes and vascular smooth muscle cells, may acquire a myofibroblast-like synthetic phenotype contributing to both SSc-related vascular dysfunction and fibrosis. A deeper understanding of the mechanisms underlying the differentiation of myofibroblasts inside the vessel wall provides the rationale for novel targeted therapeutic strategies for the treatment of SSc.
Collapse
Affiliation(s)
- Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, 50134 Florence, Italy; (E.R.); (B.S.F.); (M.M.-C.)
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy;
| | - Bianca Saveria Fioretto
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, 50134 Florence, Italy; (E.R.); (B.S.F.); (M.M.-C.)
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, 50134 Florence, Italy; (E.R.); (B.S.F.); (M.M.-C.)
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy;
| |
Collapse
|
14
|
Hachem NE, Humpfle L, Simon P, Kaese M, Weinhold B, Günther J, Galuska SP, Middendorff R. The Loss of Polysialic Acid Impairs the Contractile Phenotype of Peritubular Smooth Muscle Cells in the Postnatal Testis. Cells 2021; 10:1347. [PMID: 34072405 PMCID: PMC8230264 DOI: 10.3390/cells10061347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
In the testis, the germinal epithelium of seminiferous tubules is surrounded by contractile peritubular cells, which are involved in sperm transport. Interestingly, in postnatal testis, polysialic acid (polySia), which is also an essential player for the development of the brain, was observed around the tubules. Western blotting revealed a massive decrease of polySia from postnatal day 1 towards puberty, together with a fundamental reduction of the net-like intertubular polySia. Using polysialyltransferase knockout mice, we investigated the consequences of the loss of polySia in the postnatal testis. Compared to postnatal wild-type animals, polySia knockouts showed slightly reduced smooth muscle actin (SMA) immunostaining of peritubular smooth muscle cells (SMCs), while calponin, marking more differentiated SMCs, dramatically decreased. In contrast, testicular SMA and calponin immunostaining remained unchanged in vascular SMCs in all genotypes. In addition, the cGMP-dependent protein kinase PKG I, a key enzyme of SMC relaxation, was nearly undetectable in the peritubular SMCs. Cell proliferation in the peritubular layer increased significantly in the knockouts, as shown by proliferating cell nuclear anti (PCNA) staining. Taken together, in postnatal testis, the absence of polySia resulted in an impaired differentiation of peritubular, but not vascular, SMCs to a more synthetic phenotype. Thus, polySia might influence the maintenance of a differentiated phenotype of non-vascular SMCs.
Collapse
Affiliation(s)
- Nadim E. Hachem
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany; (N.E.H.); (L.H.)
| | - Luisa Humpfle
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany; (N.E.H.); (L.H.)
| | - Peter Simon
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Friedrichstr. 24, 35392 Giessen, Germany; (P.S.); (M.K.)
| | - Miriam Kaese
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Friedrichstr. 24, 35392 Giessen, Germany; (P.S.); (M.K.)
| | - Birgit Weinhold
- Institute of Clinical Biochemistry, OE 4340, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany;
| | - Juliane Günther
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany;
| | - Sebastian P. Galuska
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Friedrichstr. 24, 35392 Giessen, Germany; (P.S.); (M.K.)
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany;
| | - Ralf Middendorff
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany; (N.E.H.); (L.H.)
| |
Collapse
|
15
|
Eradication of specific donor-dependent variations of mesenchymal stem cells in immunomodulation to enhance therapeutic values. Cell Death Dis 2021; 12:357. [PMID: 33824286 PMCID: PMC8024246 DOI: 10.1038/s41419-021-03644-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cells (MSCs) are one of the most widely clinically trialed stem cells, due to their abilities to differentiate into multiple cell lineages, to secrete regenerative/rejuvenative factors, and to modulate immune functions, among others. In this study, we analyzed human umbilical-cord-derived MSCs from 32 donors and revealed donor-dependent variations in two non-correlated properties, (1) cell proliferation, and (2) immune modulatory functions in vitro and in vivo, which might explain inconsistent clinical efficacies of MSCs. Through unbiased transcriptomic analyses, we discovered that IFN-γ and NF-κB signaling were positively associated with immune modulatory function of MSCs. Activation of these two pathways via IFN-γ and TNF-α treatment eradicated donor-dependent variations. Additional transcriptomic analyses revealed that treatment with these two factors, while having abolished donor-dependent variations in immune modulatory function, did not overall make different donor-derived MSCs the same at whole transcriptomic levels, demonstrating that the cells were still different in many other biological perspectives, and may not perform equally for therapeutic purposes other than immune modulation. Pre-selection or pre-treatment to eradicate MSC variations in a disease-treatment-specific manner would therefore be necessary to ensure clinical efficacies. Together this study provided novel insights into the quality control perspective of using different-donor-derived MSCs to treat inflammation-related clinical conditions and/or autoimmune diseases.
Collapse
|
16
|
Travnickova M, Kasalkova NS, Sedlar A, Molitor M, Musilkova J, Slepicka P, Svorcik V, Bacakova L. Differentiation of adipose tissue-derived stem cells towards vascular smooth muscle cells on modified poly(L-lactide) foils. Biomed Mater 2021; 16:025016. [PMID: 33599213 DOI: 10.1088/1748-605x/abaf97] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The aim of our research was to study the behaviour of adipose tissue-derived stem cells (ADSCs) and vascular smooth muscle cells (VSMCs) on variously modified poly(L-lactide) (PLLA) foils, namely on pristine PLLA, plasma-treated PLLA, PLLA grafted with polyethylene glycol (PEG), PLLA grafted with dextran (Dex), and the tissue culture polystyrene (PS) control. On these materials, the ADSCs were biochemically differentiated towards VSMCs by a medium supplemented with TGFβ1, BMP4 and ascorbic acid (i.e. differentiation medium). ADSCs cultured in a non-differentiation medium were used as a negative control. Mature VSMCs cultured in both types of medium were used as a positive control. The impact of the variously modified PLLA foils and/or differences in the composition of the medium were studied with reference to cell adhesion, growth and differentiation. We observed similar adhesion and growth of ADSCs on all PLLA samples when they were cultured in the non-differentiation medium. The differentiation medium supported the expression of specific early, mid-term and/or late markers of differentiation (i.e. type I collagen, αSMA, calponin, smoothelin, and smooth muscle myosin heavy chain) in ADSCs on all tested samples. Moreover, ADSCs cultured in the differentiation medium revealed significant differences in cell growth among the samples that were similar to the differences observed in the cultures of VSMCs. The round morphology of the VSMCs indicated worse adhesion to pristine PLLA, and this sample was also characterized by the lowest cell proliferation. Culturing VSMCs in the differentiation medium inhibited their metabolic activity and reduced the cell numbers. Both cell types formed the most stable monolayer on plasma-treated PLLA and on the PS control. The behaviour of ADSCs and VSMCs on the tested PLLA foils differed according to the specific cell type and culture conditions. The suitable biocompatibility of both cell types on the tested PLLA foils seems to be favourable for vascular tissue engineering purposes.
Collapse
Affiliation(s)
- Martina Travnickova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4, Czech Republic.,Second Faculty of Medicine, Charles University, V Uvalu 84, 150 06, Prague 5, Czech Republic
| | - Nikola Slepickova Kasalkova
- Department of Solid State Engineering, University of Chemistry and Technology, Technicka 5, 166 28, Prague 6, Czech Republic
| | - Antonin Sedlar
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Martin Molitor
- Department of Plastic Surgery, Na Bulovce Hospital and First Faculty of Medicine, Charles University, Budinova 67/2, 180 81, Prague 8, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Petr Slepicka
- Department of Solid State Engineering, University of Chemistry and Technology, Technicka 5, 166 28, Prague 6, Czech Republic
| | - Vaclav Svorcik
- Department of Solid State Engineering, University of Chemistry and Technology, Technicka 5, 166 28, Prague 6, Czech Republic
| | - Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4, Czech Republic
| |
Collapse
|
17
|
Extracellular Vesicles: Novel Roles in Neurological Disorders. Stem Cells Int 2021; 2021:6640836. [PMID: 33679989 PMCID: PMC7904361 DOI: 10.1155/2021/6640836] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/13/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
Exosomes are small extracellular vesicles (EVs) secreted by almost all cells, which have been recognized as a novel platform for intercellular communication in the central nervous system (CNS). Exosomes are capable of transferring proteins, nucleic acids, lipids, and metabolites between neurons and glial cells, contributing to CNS development and maintenance of homeostasis. Evidence shows that exosomes originating from CNS cells act as suppressors or promoters in the initiation and progression of neurological disorders. Moreover, these exosomes have been shown to transfer molecules associated with diseases through the blood-brain barrier (BBB) and thus can be detected in blood. This unique feature enables exosomes to act as potential diagnostic biomarkers for neurological disorders. In addition, a substantial number of researches have indicated that exosomes derived from mesenchymal stem cells (MSCs) have repair effects on neurological disorders. Herein, we briefly introduce the roles of exosomes under physiological and pathological conditions. In particular, novel roles of exosomes as potential diagnostic biomarkers and therapeutic tools for neurological disorders are highlighted.
Collapse
|
18
|
Extracellular Vesicles from Adipose Tissue Stem Cells in Diabetes and Associated Cardiovascular Disease; Pathobiological Impact and Therapeutic Potential. Int J Mol Sci 2020; 21:ijms21249598. [PMID: 33339409 PMCID: PMC7766415 DOI: 10.3390/ijms21249598] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) are pluripotent mesenchymal stem cells found in relatively high percentages in the adipose tissue and able to self-renew and differentiate into many different types of cells. “Extracellular vesicles (EVs), small membrane vesicular structures released during cell activation, senescence, or apoptosis, act as mediators for long distance communication between cells, transferring their specific bioactive molecules into host target cells”. There is a general consensus on how to define and isolate ADSCs, however, multiple separation and characterization protocols are being used in the present which complicate the results’ integration in a single theory on ADSCs’ and their derived factors’ way of action. Metabolic syndrome and type 2 diabetes mellitus (T2DM) are mainly caused by abnormal adipose tissue size, distribution and metabolism and so ADSCs and their secretory factors such as EVs are currently investigated as therapeutics in these diseases. Moreover, due to their relatively easy isolation and propagation in culture and their differentiation ability, ADSCs are being employed in preclinical studies of implantable devices or prosthetics. This review aims to provide a comprehensive summary of the current knowledge on EVs secreted from ADSCs both as diagnostic biomarkers and therapeutics in diabetes and associated cardiovascular disease, the molecular mechanisms involved, as well as on the use of ADSC differentiation potential in cardiovascular tissue repair and prostheses.
Collapse
|
19
|
Liang W, Chen X, Dong Y, Zhou P, Xu F. Recent advances in biomaterials as instructive scaffolds for stem cells in tissue repair and regeneration. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1848832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| | - Xuerong Chen
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Yongqiang Dong
- Department of Orthopaedics, Xinchang People’s Hospital, Shaoxing, P. R. China
| | - Ping Zhou
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Fangming Xu
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| |
Collapse
|
20
|
The self-organized differentiation from MSCs into SMCs with manipulated micro/Nano two-scale arrays on TiO2 surfaces for biomimetic construction of vascular endothelial substratum. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111179. [DOI: 10.1016/j.msec.2020.111179] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 01/26/2023]
|
21
|
Antonyshyn JA, D'''''Costa KA, Santerre JP. Advancing tissue-engineered vascular grafts via their endothelialization and mechanical conditioning. THE JOURNAL OF CARDIOVASCULAR SURGERY 2020; 61:555-576. [PMID: 32909708 DOI: 10.23736/s0021-9509.20.11582-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tissue engineering has garnered significant attention for its potential to address the predominant modes of failure of small diameter vascular prostheses, namely mid-graft thrombosis and anastomotic intimal hyperplasia. In this review, we described two main features underpinning the promise of tissue-engineered vascular grafts: the incorporation of an antithrombogenic endothelium, and the generation of a structurally and biomechanically mimetic extracellular matrix. From the early attempts at the in-vitro endothelialization of vascular prostheses in the 1970s through to the ongoing clinical trials of fully tissue-engineered vascular grafts, the historical advancements and unresolved challenges that characterize the current state-of-the-art are summarized in a manner that establishes a guide for the development of an effective vascular prosthesis for small diameter arterial reconstruction. The importance of endothelial cell purity and their arterial specification for the prevention of both diffuse neointimal hyperplasia and the accelerated development of atherosclerotic lesions is delineated. Additionally, the need for an extracellular matrix that recapitulates both the composition and structure of native elastic arteries to facilitate the protracted stability and patency of an engineered vasoactive conduit is described. Finally, the capacity of alternative sources of cells and mechanical conditioning to overcome these technical barriers to the clinical translation of an effective small diameter vascular prosthesis is discussed. In conclusion, this review provides an overview of the historical development of tissue-engineered vascular grafts, highlighting specific areas warranting further research, and commentating on the outlook of a clinically feasible and therapeutically efficacious vascular prosthesis for small diameter arterial reconstruction.
Collapse
Affiliation(s)
- Jeremy A Antonyshyn
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Katya A D'''''Costa
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - J Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada - .,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada.,Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
22
|
Yao Z, Liu H, Yang M, Bai Y, Zhang B, Wang C, Yan Z, Niu G, Zou Y, Li Y. Bone marrow mesenchymal stem cell-derived endothelial cells increase capillary density and accelerate angiogenesis in mouse hindlimb ischemia model. Stem Cell Res Ther 2020; 11:221. [PMID: 32513272 PMCID: PMC7278145 DOI: 10.1186/s13287-020-01710-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/20/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) can improve limb perfusion and increase vessel density in a murine model of hindlimb ischemia. But low engraftment rate of those cells limited their therapeutic effect. Endothelial cells (ECs) play an important role in neovascularization. And MSCs can differentiate into ECs in vitro. The aim of this study is to investigate if EC differentiation of MSCs in vitro before transplantation is effective in improving therapeutic outcomes in the treatment of ischemic disease in a murine ischemia animal model. Methods MSCs were isolated from the bone marrow of EGFP-transgenic mice by density gradient centrifugation. The identity of the MSCs was determined by their cluster of differentiation (CD) marker profile by flow cytometry. Inducing medium containing a few cytokines was applied to induce the MSCs to differentiate into ECs. Endothelial differentiation was quantitatively evaluated using flow cytometry, quantitative real-time PCR (qRT-PCR), immunofluorescence, Matrigel tube formation assay, and Dil-labeled acetylated low-density lipoprotein uptake assay. Mouse hindlimb ischemia model was made by excision of the femoral artery. Uninduced EGFP+ MSCs, induced EGFP+ MSCs, and PBS were intramuscularly injected into the gastrocnemius following ischemia no later than 24 h after operation. Restoration of blood flow and muscle function was evaluated by laser Doppler perfusion imaging. Immunofluorescence was conducted to evaluate the engraftment of transplanted MSCs. Histological analysis was performed to evaluate blood vessel formation. Results Induced EGFP+ MSCs expressed endothelial markers and exhibited tube formation capacity. Mice in the induced EGFP+ MSCs group had a better blood perfusion recovery, enhanced vessel densities, higher engraftment, and improved function of the ischemic limb than those in the uninduced EGFP+ MSCs or PBS groups. Conclusions This study reveals that after short-term pre-treatment in the EC-inducing medium, induced MSCs acquire stronger vessel formation capability and enhanced angiogenic therapeutic effect in the murine hindlimb ischemia model.
Collapse
Affiliation(s)
- Ziping Yao
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Huihui Liu
- Department of Hematology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Min Yang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Yun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Bihui Zhang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Chengen Wang
- Department of Minimally Invasive Tumor Therapies Center, National Center of Gerontology, Beijing Hospital, Beijing, China
| | - Ziguang Yan
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Guochen Niu
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Yinghua Zou
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China.
| | - Yuan Li
- Department of Hematology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China.
| |
Collapse
|
23
|
Xiao D, Zhang J, Zhang C, Barbieri D, Yuan H, Moroni L, Feng G. The role of calcium phosphate surface structure in osteogenesis and the mechanisms involved. Acta Biomater 2020; 106:22-33. [PMID: 31926336 DOI: 10.1016/j.actbio.2019.12.034] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/11/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Calcium phosphate (CaP) ceramics have been widely used for bone regeneration because of their ability to induce osteogenesis. Surface properties, including chemical composition and surface structure, are known to play a crucial role in osteoconduction and osteoinduction. This review systematically analyzes the effects of surface properties, in particular the surface structure, of CaP scaffolds on cell behavior and new bone formation. We also summarize the possible signaling pathways involved in the osteogenic differentiation of bone-related cells when cultured on surfaces with various structures in vitro. The significant immune response initiated by surface structure involved in osteogenic differentiation of cells is also discussed in this review. Taken together, the new biological principle for advanced biomaterials is not only to directly stimulate osteogenic differentiation of bone-related cells but also to modulate the immune response in vivo. Although the reaction mechanism responsible for bone formation induced by CaP surface structure is not clear yet, the insights on surface structure-mediated osteogenic differentiation and osteoimmunomodulation could aid the optimization of CaP-based biomaterials for bone regeneration. STATEMENT OF SIGNIFICANCE: CaP ceramics have similar inorganic composition with natural bone, which have been widely used for bone tissue scaffolds. CaP themselves are not osteoinductive; however, osteoinductive properties could be introduced to CaP materials by surface engineering. This paper systematically summarizes the effects of surface properties, especially surface structure, of CaP scaffolds on bone formation. Additionally, increasing evidence has proved that the bone healing process is not only affected by the osteogenic differentiation of bone-related cells, but also relevant to the the cooperation of immune system. Thus, we further review the possible signaling pathways involved in the osteogenic differentiation and immune response of cells cultured on scaffold surface. These insights into surface structure-mediated osteogenic differentiation and osteoimmunomodulated-based strategy could aid the optimization of CaP-based biomaterials.
Collapse
|
24
|
Mallis P, Papapanagiotou A, Katsimpoulas M, Kostakis A, Siasos G, Kassi E, Stavropoulos-Giokas C, Michalopoulos E. Efficient differentiation of vascular smooth muscle cells from Wharton's Jelly mesenchymal stromal cells using human platelet lysate: A potential cell source for small blood vessel engineering. World J Stem Cells 2020; 12:203-221. [PMID: 32266052 PMCID: PMC7118289 DOI: 10.4252/wjsc.v12.i3.203] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/17/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The development of fully functional small diameter vascular grafts requires both a properly defined vessel conduit and tissue-specific cellular populations. Mesenchymal stromal cells (MSCs) derived from the Wharton's Jelly (WJ) tissue can be used as a source for obtaining vascular smooth muscle cells (VSMCs), while the human umbilical arteries (hUAs) can serve as a scaffold for blood vessel engineering. AIM To develop VSMCs from WJ-MSCs utilizing umbilical cord blood platelet lysate. METHODS WJ-MSCs were isolated and expanded until passage (P) 4. WJ-MSCs were properly defined according to the criteria of the International Society for Cell and Gene Therapy. Then, these cells were differentiated into VSMCs with the use of platelet lysate from umbilical cord blood in combination with ascorbic acid, followed by evaluation at the gene and protein levels. Specifically, gene expression profile analysis of VSMCs for ACTA2, MYH11, TGLN, MYOCD, SOX9, NANOG homeobox, OCT4 and GAPDH, was performed. In addition, immunofluorescence against ACTA2 and MYH11 in combination with DAPI staining was also performed in VSMCs. HUAs were decellularized and served as scaffolds for possible repopulation by VSMCs. Histological and biochemical analyses were performed in repopulated hUAs. RESULTS WJ-MSCs exhibited fibroblastic morphology, successfully differentiating into "osteocytes", "adipocytes" and "chondrocytes", and were characterized by positive expression (> 90%) of CD90, CD73 and CD105. In addition, WJ-MSCs were successfully differentiated into VSMCs with the proposed differentiation protocol. VSMCs successfully expressed ACTA2, MYH11, MYOCD, TGLN and SOX9. Immunofluorescence results indicated the expression of ACTA2 and MYH11 in VSMCs. In order to determine the functionality of VSMCs, hUAs were isolated and decellularized. Based on histological analysis, decellularized hUAs were free of any cellular or nuclear materials, while their extracellular matrix retained intact. Then, repopulation of decellularized hUAs with VSMCs was performed for 3 wk. Decellularized hUAs were repopulated efficiently by the VSMCs. Biochemical analysis revealed the increase of total hydroyproline and sGAG contents in repopulated hUAs with VSMCs. Specifically, total hydroxyproline and sGAG content after the 1st, 2nd and 3rd wk was 71 ± 10, 74 ± 9 and 86 ± 8 μg hydroxyproline/mg of dry tissue weight and 2 ± 1, 3 ± 1 and 3 ± 1 μg sGAG/mg of dry tissue weight, respectively. Statistically significant differences were observed between all study groups (P < 0.05). CONCLUSION VSMCs were successfully obtained from WJ-MSCs with the proposed differentiation protocol. Furthermore, hUAs were efficiently repopulated by VSMCs. Differentiated VSMCs from WJ-MSCs could provide an alternative source of cells for vascular tissue engineering.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece.
| | - Aggeliki Papapanagiotou
- Department of Biological Chemistry, Medical School, National and Kapodistrian Univesity of Athens, Athens 15772, Greece
| | - Michalis Katsimpoulas
- Center of Experimental Surgery, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Alkiviadis Kostakis
- Center of Experimental Surgery, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Gerasimos Siasos
- Department of Biological Chemistry, Medical School, National and Kapodistrian Univesity of Athens, Athens 15772, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian Univesity of Athens, Athens 15772, Greece
| | | | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| |
Collapse
|
25
|
Development of double porous poly (ε - caprolactone)/chitosan polymer as tissue engineering scaffold. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 107:110257. [DOI: 10.1016/j.msec.2019.110257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/24/2022]
|
26
|
Zhang X, Simmons CA, Paul Santerre J. Paracrine signalling from monocytes enables desirable extracellular matrix accumulation and temporally appropriate phenotype of vascular smooth muscle cell-like cells derived from adipose stromal cells. Acta Biomater 2020; 103:129-141. [PMID: 31821896 DOI: 10.1016/j.actbio.2019.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/12/2022]
Abstract
In vascular tissue engineering, the ability to obtain a robust and safe vascular tissue cell source (e.g. vascular smooth muscle cells (VSMCs)) and to promote vascular tissue-specific extracellular matrix (ECM) protein production is critically important. Mature blood vessel-derived VSMCs are not practical for in vitro vascular tissue regeneration. The authors have conceived a strategy to differentiate adipose derived stromal cells (ASCs) into VSMC-like cells (ASC-VSMCs) that were similar to mature umbilical artery VSMCs at the transcriptional, protein and contraction function levels. Monocytes/macrophages are known as important regulators of the inflammation and regeneration processes within different tissue types of the body. However, our understanding of the potential interactions between specific tissue-like cells differentiated from stem/stromal cells (e.g. ASC-VSMCs) and monocytes/macrophages (cued by specific biomaterial scaffolds) is still limited. In this study, indirect and direct ASC-VSMC-monocyte co-cultures were constructed within a porous polyurethane scaffold (D-PHI) previously shown to have an immunomodulatory character. The effects of monocytes/macrophages on the cellularity (cell number detected with DNA quantification assay), ECM (glycosaminoglycan (GAG), collagen, and elastin) accumulation as well as the maintenance of contractile VSMC markers (calponin and smoothelin) of the ASC-VSMCs after a month of co-culture were investigated. It was found that monocyte paracrine signalling in D-PHI positively affected the cellularity and ECM accumulation of ASC-VSMCs in co-culture. Cause-effect relationships were also identified between the release of pro-inflammatory/anti-inflammatory factors (i.e. IL6, TGF-β1) in co-culture and the expression of contractile proteins (calponin and smoothelin) by ASC-VSMCs. This study demonstrated the importance of combining an immune cell strategy with stromal cell derived VSMCs (i.e. ASC-VSMCs) to achieve a practical vascular tissue engineering outcome. STATEMENT OF SIGNIFICANCE: Adipose stromal cell derived-vascular smooth muscle cells (ASC-VSMCs) are a promising cell source for vascular tissue engineering. Monocytes/monocyte derived macrophages can be harnessed as an immune-assisted strategy to promote vascular tissue regeneration. This study demonstrated that the co-culture of human ASC-VSMCs with monocytes significantly enhanced the cellularity and extracellular matrix (ECM) accumulation within anionic polyurethane (D-PHI) scaffolds, partially mediated by monocyte paracrine signalling mechanisms. In addition, specific VSMC contractile markers (calponin and smoothelin) were still present in ASC-VSMCs when the cells were exposed to monocytes for a month in vitro. This study corroborated the potential selection of ASC-VSMCs for in vitro engineering of vascular tissue in an immunomodulatory biomaterial scaffold (e.g. D-PHI) based co-culture system containing monocytes.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th floor, room 1435, Toronto, Ontario M5G 1M1, Canada
| | - Craig A Simmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada; Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th floor, room 1435, Toronto, Ontario M5G 1M1, Canada
| | - J Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th floor, room 1435, Toronto, Ontario M5G 1M1, Canada.
| |
Collapse
|
27
|
Ni H, Zhao Y, Ji Y, Shen J, Xiang M, Xie Y. Adipose-derived stem cells contribute to cardiovascular remodeling. Aging (Albany NY) 2019; 11:11756-11769. [PMID: 31800397 PMCID: PMC6932876 DOI: 10.18632/aging.102491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/17/2019] [Indexed: 02/06/2023]
Abstract
Obesity is an independent risk factor for cardiovascular disease. Adipose tissue was initially thought to be involved in metabolism through paracrine. Recent researches discovered mesenchymal stem cells inside adipose tissue which could differentiate into vascular lineages in vitro and in vivo, participating vascular remodeling. However, there were few researches focusing on distinct characteristics and functions of adipose-derived stem cells (ADSCs) from different regions. This is the first comprehensive review demonstrating the variances of ADSCs from the perspective of their origins.
Collapse
Affiliation(s)
- Hui Ni
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiming Zhao
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongli Ji
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
28
|
Lau S, Klingenberg M, Mrugalla A, Helms F, Sedding D, Haverich A, Wilhelmi M, Böer U. Biochemical Myogenic Differentiation of Adipogenic Stem Cells Is Donor Dependent and Requires Sound Characterization. Tissue Eng Part A 2019; 25:936-948. [PMID: 30648499 DOI: 10.1089/ten.tea.2018.0172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPACT STATEMENT We here showed that even under optimized conditions for biochemical differentiation of adipose-derived stem cells (with respect to a pronounced marker protein expression for a reasonable period of time) it was not possible to obtain functional smooth muscle cells from all donors. Moreover, an underestimated role may play the effect of the scaffold material on smooth muscle cell functionality. Both aspects are crucial for the successful tissue engineering of the vascular medial layer combining autologous cells with a suitable scaffold material and thus should be thoroughly addressed in each individualized therapeutic approach.
Collapse
Affiliation(s)
- Skadi Lau
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Melanie Klingenberg
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Anna Mrugalla
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany
| | - Florian Helms
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany
| | - Daniel Sedding
- 3Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Mathias Wilhelmi
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Ulrike Böer
- 1Lower Saxony Centre of Biotechnology Implant Research and Development (NIFE), Hannover Medical School, Hannover, Germany.,2Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
29
|
He C, Wang M, Yan Z, Zhang S, Liu H. Isolation and culture of vascular smooth muscle cells from rat placenta. J Cell Physiol 2019; 234:7675-7682. [PMID: 30478916 DOI: 10.1002/jcp.27721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 10/16/2018] [Indexed: 12/17/2023]
Abstract
We developed a new separation method for isolating placental vascular smooth muscle cells (PVSMCs) from a rat in this study. Our method used the magnetic force between a magnet and ferrous ferric oxide (Fe3 O 4 ) to make the separation and extraction processes easier and more efficient. From the first to sixth generation, the cells isolated using this protocol were identified as smooth muscle cells (SMCs) by their immunoreactivity to the SMC markers and by the "hill and valley" morphology. PVSMCs were exposed to angiotensin II (1 μmol/L) and resulted in sharply increased intracellular Ca 2+ concentration. Furthermore, activation of protein kinase C (PKC) increased concomitantly with a decrease in calponin expression. These results indicate that the isolated cells had biological activity. Our method of isolating PVSMCs from rat leads to isolation of cultured cells with activity and high purity. The approach will be useful in research studies on placental vascular diseases.
Collapse
Affiliation(s)
- Chunyu He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Capital Medical University, Beijing, China
| | - Meili Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Capital Medical University, Beijing, China
| | - Zi Yan
- Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Capital Medical University, Beijing, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Directional Topography Influences Adipose Mesenchymal Stromal Cell Plasticity: Prospects for Tissue Engineering and Fibrosis. Stem Cells Int 2019; 2019:5387850. [PMID: 31191675 PMCID: PMC6525798 DOI: 10.1155/2019/5387850] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/24/2018] [Accepted: 02/11/2019] [Indexed: 01/17/2023] Open
Abstract
Introduction Progenitor cells cultured on biomaterials with optimal physical-topographical properties respond with alignment and differentiation. Stromal cells from connective tissue can adversely differentiate to profibrotic myofibroblasts or favorably to smooth muscle cells (SMC). We hypothesized that myogenic differentiation of adipose tissue-derived stromal cells (ASC) depends on gradient directional topographic features. Methods Polydimethylsiloxane (PDMS) samples with nanometer and micrometer directional topography gradients (wavelength (w) = 464-10, 990 nm; amplitude (a) = 49-3, 425 nm) were fabricated. ASC were cultured on patterned PDMS and stimulated with TGF-β1 to induce myogenic differentiation. Cellular alignment and adhesion were assessed by immunofluorescence microscopy after 24 h. After seven days, myogenic differentiation was examined by immunofluorescence microscopy, gene expression, and immunoblotting. Results Cell alignment occurred on topographies larger than w = 1758 nm/a = 630 nm. The number and total area of focal adhesions per cell were reduced on topographies from w = 562 nm/a = 96 nm to w = 3919 nm/a = 1430 nm. Focal adhesion alignment was increased on topographies larger than w = 731 nm/a = 146 nm. Less myogenic differentiation of ASC occurred on topographies smaller than w = 784 nm/a = 209 nm. Conclusion ASC adherence, alignment, and differentiation are directed by topographical cues. Our evidence highlights a minimal topographic environment required to facilitate the development of aligned and differentiated cell layers from ASC. These data suggest that nanotopography may be a novel tool for inhibiting fibrosis.
Collapse
|
31
|
ZHAO XIAOE, YANG ZHENSHAN, GAO ZHEN, GE JUNBANG, WEI QIANG, MA BAOHUA. 6-Bromoindirubin-3’-oxime promotes osteogenic differentiation of canine BMSCs through inhibition of GSK3β activity and activation of the Wnt/β-catenin signaling pathway. ACTA ACUST UNITED AC 2019; 91:e20180459. [PMID: 30916158 DOI: 10.1590/0001-3765201920180459] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/09/2018] [Indexed: 12/16/2022]
|
32
|
Zhu M, Wu Y, Li W, Dong X, Chang H, Wang K, Wu P, Zhang J, Fan G, Wang L, Liu J, Wang H, Kong D. Biodegradable and elastomeric vascular grafts enable vascular remodeling. Biomaterials 2018; 183:306-318. [DOI: 10.1016/j.biomaterials.2018.08.063] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 02/07/2023]
|
33
|
Zhang X, Simmons CA, Santerre JP. Alterations of MEK1/2-ERK1/2, IFNγ and Smad2/3 associated Signalling pathways during cryopreservation of ASCs affect their differentiation towards VSMC-like cells. Stem Cell Res 2018; 32:115-125. [DOI: 10.1016/j.scr.2018.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/06/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
|
34
|
Substrate elasticity regulates adipose-derived stromal cell differentiation towards osteogenesis and adipogenesis through β-catenin transduction. Acta Biomater 2018; 79:83-95. [PMID: 30134207 DOI: 10.1016/j.actbio.2018.08.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 08/03/2018] [Accepted: 08/17/2018] [Indexed: 02/05/2023]
Abstract
It is generally recognised that mesenchymal stem cells (MSCs) can differentiate into multiple lineages through guidance from the biophysical properties of the substrates. However, the precise biophysical mechanism that enables MSCs to respond to substrate properties remains unclear. In the current study, polydimethylsiloxane (PDMS) substrates with different stiffnesses were fabricated and the way in which the elastic modulus of the substrate regulated differentiation towards osteogenesis and adipogenesis in adipose-derived stromal cells (ASCs) was explored. Initially, a cell morphology change by SEM was observed between the stiff and soft substrates. The cytoskeleton stains including microfilament by F-actin and microtubule by α- and β-tubulin further showed a larger cell spreading area on the stiff substrate. Then the expression of vinculin, in charge for the linkage of adhesion molecules to the actin cytoskeleton, was enhanced on the stiff substrate. This change in focal adhesion plaque further triggered intracellular β-catenin signaling and promoted its nuclear translocation especially on the stiff substrate. The influence of β-catenin signaling on direct differentiation to osteogenic lineages was through direct binding between its downstream protein, Lef-1, and the osteogenic transcriptional factors, Runx2 and Osx, while on differentiation to adipogenic lineages was through modulating the expression of PPARγ. The imbalance of stiffness-induced β-catenin signaling finally induced a stronger osteogenesis and a weaker adipogenesis on the stiff substrate relative to those on the soft substrate. This study indicates the importance of stiffness on ASC differentiation and could help to increase understanding of the mechanism underlying molecular signal transduction from mechanosensing, mechanotransducing to stem cell differentiation. STATEMENT OF SIGNIFICANCE Mesenchymal stem cells can differentiate into multiple lineages, such as adipogenesis, myogenesis, neurogenesis, angiogenesis and osteogenesis, through influence of biophysical properties of the extracellular matrix. However, the precise bio-mechanism that triggers stem cell differentiation in response to matrix biophysical properties remains unclear. In the current study, we provide a series of experiments involving the characterization of cell morphology, microfilament, microtubule and adhesion capacity of adipose-derived stromal cells (ASCs) in response to substrate stiffness, and further elucidation of cytoplasmic β-catenin-dependent signal transduction, nuclear translocation and resultant promoter activation of transcriptional factors for osteogenesis and adipogenesis. This study provides an explanation on deeper understanding of bio-mechanism underlying substrate stiffness-triggered β-catenin signal transduction from active mechanosensing, mechanotransducing to stem cell differentiation.
Collapse
|
35
|
Developmental Pathways Pervade Stem Cell Responses to Evolving Extracellular Matrices of 3D Bioprinted Microenvironments. Stem Cells Int 2018; 2018:4809673. [PMID: 29765414 PMCID: PMC5896227 DOI: 10.1155/2018/4809673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/10/2017] [Accepted: 12/10/2017] [Indexed: 12/17/2022] Open
Abstract
Developmental studies and 3D in vitro model systems show that the production and engagement of extracellular matrix (ECM) often precede stem cell differentiation. Yet, unclear is how the ECM triggers signaling events in sequence to accommodate multistep process characteristic of differentiation. Here, we employ transcriptome profiling and advanced imaging to delineate the specificity of ECM engagement to particular differentiation pathways and to determine whether specificity in this context is a function of long-term ECM remodeling. To this end, human mesenchymal stem cells (hMSCs) were cultured in 3D bioprinted prisms created from ECM proteins and associated controls. We found that exogenous ECM provided in 3D microenvironments at early time points impacts on the composition of microenvironments at later time points and that each evolving 3D microenvironment is uniquely poised to promote stem cell differentiation. Moreover, 2D cultures undergo minimal ECM remodeling and are ill-equipped to stimulate pathways associated with development.
Collapse
|