1
|
Vogt A, Faher A, Kucharczak J, Birch M, McCaskie A, Khan W. The Effects of Gender on Mesenchymal Stromal Cell (MSC) Proliferation and Differentiation In Vitro: A Systematic Review. Int J Mol Sci 2024; 25:13585. [PMID: 39769346 PMCID: PMC11677156 DOI: 10.3390/ijms252413585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Mesenchymal stromal cells (MSCs) have the potential for novel treatments of several musculoskeletal conditions due to their ability to differentiate into several cell lineages including chondrocytes, adipocytes and osteocytes. Researchers are exploring whether this could be utilized for novel therapies for joint afflictions. The role of gender in the ability of MSCs to differentiate and proliferate into different cells has not been clearly defined. This systematic review aims to report the current literature on studies, characterized by high quality and in-depth analysis even though quantitatively limited, that have looked at the role of gender in the differentiation and proliferation of MSCs. Sixteen studies were identified during the literature search, reporting 533 patients, of which 202 were male and 331 were female. MSC proliferation, phenotypic analysis and differentiation are reported and contrasted in terms of donor gender. Heterogeneity in methodologies across studies likely contributes to the inconclusive findings presented here, with no discernible statistical disparity observed between genders in differentiation traits. Nevertheless, the proliferation results indicate a notable gender-related impact. Future investigations should aim to ascertain the potential influence of gender on MSC proliferation capacities more conclusively, emphasizing the necessity of standardized protocols for MSC analyses to enhance accuracy and comparability across studies.
Collapse
Affiliation(s)
- Antonia Vogt
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.)
| | - Anissa Faher
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 2SP, UK
| | - Joanna Kucharczak
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 2SP, UK
| | - Mark Birch
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.)
| | - Andrew McCaskie
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.)
| | - Wasim Khan
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.)
| |
Collapse
|
2
|
El-Husseiny HM, Kaneda M, Mady EA, Yoshida T, Doghish AS, Tanaka R. Impact of Adipose Tissue Depot Harvesting Site on the Multilineage Induction Capacity of Male Rat Adipose-Derived Mesenchymal Stem Cells: An In Vitro Study. Int J Mol Sci 2023; 24:7513. [PMID: 37108673 PMCID: PMC10138771 DOI: 10.3390/ijms24087513] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Recently, substantial attention has been paid toward adipose-derived mesenchymal stem cells (AdMSCs) as a potential therapy in tissue engineering and regenerative medicine applications. Rat AdMSCs (r-AdMSCs) are frequently utilized. However, the influence of the adipose depot site on the multilineage differentiation potential of the r-AdMSCs is still ambiguous. Hence, the main objective of this study was to explore the influence of the adipose tissue harvesting location on the ability of r-AdMSCs to express the stem-cell-related markers and pluripotency genes, as well as their differentiation capacity, for the first time. Herein, we have isolated r-AdMSCs from the inguinal, epididymal, peri-renal, and back subcutaneous fats. Cells were compared in terms of their phenotype, immunophenotype, and expression of pluripotency genes using RT-PCR. Additionally, we investigated their potential for multilineage (adipogenic, osteogenic, and chondrogenic) induction using special stains confirmed by the expression of the related genes using RT-qPCR. All cells could positively express stem cell marker CD 90 and CD 105 with no significant in-between differences. However, they did not express the hematopoietic markers as CD 34 and CD 45. All cells could be induced successfully. However, epididymal and inguinal cells presented the highest capacity for adipogenic and osteogenic differentiation (21.36-fold and 11.63-fold for OPN, 29.69-fold and 26.68-fold for BMP2, and 37.67-fold and 22.35-fold for BSP, respectively, in epididymal and inguinal cells (p < 0.0001)). On the contrary, the subcutaneous cells exhibited a superior potential for chondrogenesis over the other sites (8.9-fold for CHM1 and 5.93-fold for ACAN, (p < 0.0001)). In conclusion, the adipose tissue harvesting site could influence the differentiation capacity of the isolated AdMSCs. To enhance the results of their employment in various regenerative cell-based therapies, it is thus vital to take the collection site selection into consideration.
Collapse
Affiliation(s)
- Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Division of Animal Life Sciences, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan
| | - Eman A. Mady
- Department of Animal Hygiene, Behavior, and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Tadashi Yoshida
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Ahmed S. Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City 11829, Cairo, Egypt
- Department of Biochemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11651, Cairo, Egypt
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan
| |
Collapse
|
3
|
Frias F, Matos B, Jarnalo M, Freitas-Ribeiro S, Reis RL, Pirraco RP, Horta R. Stromal Vascular Fraction Obtained From Subcutaneous Adipose Tissue: Ex-Obese and Older Population as Main Clinical Targets. J Surg Res 2023; 283:632-639. [PMID: 36446251 DOI: 10.1016/j.jss.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Human adipose tissue contains a heterogeneous and synergistic mixture of cells called stromal vascular fraction (SVF) with highly proliferative and angiogenic properties, conferring promising applicability in the field of regenerative medicine. This study aims to investigate if age, body mass index (BMI), history of obesity and massive weight loss, and harvest site are related to SVF cell marker expression. METHODS A total of 26 samples of subcutaneous adipose tissue were harvested from patients admitted to the Plastic and Reconstructive department in University Hospital Center of São João, Porto, Portugal, for body contouring surgery. The percentage of cells expressing CD31, CD34, CD45, CD73, CD90, and CD105 was assessed and compared with patient's age, BMI, history of obesity and massive weight loss (ex-obese group), and harvest site. RESULTS In the ex-obese group, a significantly higher number of cells expressing CD90 (P = 0.002) was found. BMI, harvest site, and age appear to have no association with SVF subpopulations. CONCLUSIONS This study suggests that ex-obese patients have a higher percentage of SVF cells expressing CD90, which correlates with higher proliferative and angiogenic rates. The effect of former obesity and massive weight loss on the expression of CD90 is a new and relevant finding because it makes this population a suitable candidate for reconstructive and aesthetic surgery and other fields of regenerative medicine. The use of SVF appears also promising in older patients because no negative correlation between increasing age and different cell markers expression was found.
Collapse
Affiliation(s)
- Francisca Frias
- Department of Plastic, Reconstructive and Maxillo-Facial Surgery, and Burn Unit, Centro Hospitalar de São João, University of Porto, Porto, Portugal.
| | - Beatriz Matos
- Department of Plastic, Reconstructive and Maxillo-Facial Surgery, and Burn Unit, Centro Hospitalar de São João, University of Porto, Porto, Portugal
| | - Mariana Jarnalo
- Department of Plastic, Reconstructive and Maxillo-Facial Surgery, and Burn Unit, Centro Hospitalar de São João, University of Porto, Porto, Portugal
| | - Sara Freitas-Ribeiro
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Ricardo Horta
- Department of Plastic, Reconstructive and Maxillo-Facial Surgery, and Burn Unit, Centro Hospitalar de São João, University of Porto, Porto, Portugal
| |
Collapse
|
4
|
Sun X, Song W, Teng L, Huang Y, Liu J, Peng Y, Lu X, Yuan J, Zhao X, Zhao Q, Xu Y, Shen J, Peng X, Ren L. MiRNA 24-3p-rich exosomes functionalized DEGMA-modified hyaluronic acid hydrogels for corneal epithelial healing. Bioact Mater 2022; 25:640-656. [PMID: 37056274 PMCID: PMC10086767 DOI: 10.1016/j.bioactmat.2022.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 11/02/2022] Open
Abstract
The damage of corneal epithelium may lead to the formation of irreversible corneal opacities and even blindness. The migration rate of corneal epithelial cells directly affects corneal repair. Here, we explored ocu-microRNA 24-3p (miRNA 24-3p) that can promote rabbit corneal epithelial cells migration and cornea repair. Exosomes, an excellent transport carrier, were exacted from adipose derived mesenchymal stem cells for loading with miRNA 24-3p to prepare miRNA 24-3p-rich exosomes (Exos-miRNA 24-3p). It can accelerate corneal epithelial migration in vitro and in vivo. For application in cornea alkali burns, we further modified hyaluronic acid with di(ethylene glycol) monomethyl ether methacrylate (DEGMA) to obtain a thermosensitive hydrogel, also reported a thermosensitive DEGMA-modified hyaluronic acid hydrogel (THH) for the controlled release of Exos-miRNA 24-3p. It formed a highly uniform and clear thin layer on the ocular surface to resist clearance from blinking and extended the drug-ocular-epithelium contact time. The use of THH-3/Exos-miRNA 24-3p for 28 days after alkali burn injury accelerated corneal epithelial defect healing and epithelial maturation. It also reduced corneal stromal fibrosis and macrophage activation. MiRNA 24-3p-rich exosomes functionalized DEGMA-modified hyaluronic acid hydrogel as a multilevel delivery strategy has a potential use for cell-free therapy of corneal epithelial regeneration.
Collapse
|
5
|
Adipose-derived exosomes block muscular stem cell proliferation in aged mouse by delivering miRNA Let-7d-3p that targets transcription factor HMGA2. J Biol Chem 2022; 298:102098. [PMID: 35679898 PMCID: PMC9257422 DOI: 10.1016/j.jbc.2022.102098] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/22/2022] Open
Abstract
Sarcopenia is an aging-associated attenuation of muscular volume and strength and is the major cause of frailty and falls in elderly individuals. The number of individuals with sarcopenia is rapidly increasing worldwide; however, little is known about the underlying mechanisms of the disease. Sarcopenia often copresents with obesity, and some patients with sarcopenia exhibit accumulation of peri-organ or intra-organ adipose tissue as ectopic fat deposition, including atrophied skeletal muscle. In this study, we showed that transplantation of the perimuscular adipose tissue (PMAT) to the hindlimb thigh muscles of young mice decreased the number of integrin α7/CD29-double positive muscular stem/progenitor cells and that the reaction was mediated by PMAT-derived exosomes. We also found that the inhibition of cell proliferation was induced by Let-7d-3p miRNA that targets HMGA2, which is an important transcription factor for stem cell self-renewal, in muscular stem/progenitor cells and the composite molecular reaction in aged adipocytes. Reduction of Let-7 miRNA repressor Lin28 A/B and activation of nuclear factor-kappa B signaling can lead to the accumulation of Let-7d-3p in the exosomes of aged PMAT. These findings suggest a novel crosstalk between adipose tissue and skeletal muscle in the development of aging-associated muscular atrophy and indicate that adipose tissue–derived miRNAs may play a key role in sarcopenia.
Collapse
|
6
|
Nebel S, Lux M, Kuth S, Bider F, Dietrich W, Egger D, Boccaccini AR, Kasper C. Alginate Core-Shell Capsules for 3D Cultivation of Adipose-Derived Mesenchymal Stem Cells. Bioengineering (Basel) 2022; 9:66. [PMID: 35200419 PMCID: PMC8869374 DOI: 10.3390/bioengineering9020066] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are primary candidates in tissue engineering and stem cell therapies due to their intriguing regenerative and immunomodulatory potential. Their ability to self-assemble into three-dimensional (3D) aggregates further improves some of their therapeutic properties, e.g., differentiation potential, secretion of cytokines, and homing capacity after administration. However, high hydrodynamic shear forces and the resulting mechanical stresses within commercially available dynamic cultivation systems can decrease their regenerative properties. Cells embedded within a polymer matrix, however, lack cell-to-cell interactions found in their physiological environment. Here, we present a "semi scaffold-free" approach to protect the cells from high shear forces by a physical barrier, but still allow formation of a 3D structure with in vivo-like cell-to-cell contacts. We highlight a relatively simple method to create core-shell capsules by inverse gelation. The capsules consist of an outer barrier made from sodium alginate, which allows for nutrient and waste diffusion and an inner compartment for direct cell-cell interactions. Next to capsule characterization, a harvesting procedure was established and viability and proliferation of human adipose-derived MSCs were investigated. In the future, this encapsulation and cultivation technique might be used for MSC-expansion in scalable dynamic bioreactor systems, facilitating downstream procedures, such as cell harvest and differentiation into mature tissue grafts.
Collapse
Affiliation(s)
- Sabrina Nebel
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences BOKU Vienna, 1190 Vienna, Austria; (S.N.); (M.L.); (D.E.)
| | - Manuel Lux
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences BOKU Vienna, 1190 Vienna, Austria; (S.N.); (M.L.); (D.E.)
| | - Sonja Kuth
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (S.K.); (F.B.); (A.R.B.)
| | - Faina Bider
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (S.K.); (F.B.); (A.R.B.)
| | - Wolf Dietrich
- Department of Gynecology and Obstetrics, Karl Landsteiner University of Health Sciences, Alter Ziegelweg 10, 3430 Tulln, Austria;
| | - Dominik Egger
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences BOKU Vienna, 1190 Vienna, Austria; (S.N.); (M.L.); (D.E.)
| | - Aldo R. Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (S.K.); (F.B.); (A.R.B.)
| | - Cornelia Kasper
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences BOKU Vienna, 1190 Vienna, Austria; (S.N.); (M.L.); (D.E.)
| |
Collapse
|
7
|
A Comparative Study of the Effect of Anatomical Site on Multiple Differentiation of Adipose-Derived Stem Cells in Rats. Cells 2021; 10:cells10092469. [PMID: 34572123 PMCID: PMC8465004 DOI: 10.3390/cells10092469] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived from adipose tissue are evolved into various cell-based regenerative approaches. Adipose-derived stem cells (ASCs) isolated from rats are commonly used in tissue engineering studies. Still, there is a gap in knowledge about how the harvest locations influence and guide cell differentiation. This study aims to investigate how the harvesting site affects stem-cell-specific surface markers expression, pluripotency, and differentiation potential of ASCs in female Sprague Dawley rats. ASCs were extracted from the adipose tissue of the peri-ovarian, peri-renal, and mesenteric depots and were compared in terms of cell morphology. MSCs phenotype was validated by cell surfaces markers using flow cytometry. Moreover, pluripotent gene expression of Oct4, Nanog, Sox2, Rex-1, and Tert was evaluated by reverse transcriptase-polymerase chain reaction (RT-PCR). ASCs multipotency was evaluated by specific histological stains, and the results were confirmed by quantitative polymerase chain reaction (RT-qPCR) expression analysis of specific genes. There was a non-significant difference detected in the cell morphology and immunophenotype between different harvesting sites. ASCs from multiple locations were significantly varied in their capacity to differentiate into adipocytes, osteoblastic cells, and chondrocytes. To conclude, depot selection is a critical element that should be considered when using ASCs in tissue-specific cell-based regenerative therapies research.
Collapse
|
8
|
Wu Y, Zhang Q, Zhao B, Wang X. Effect and mechanism of propranolol on promoting osteogenic differentiation and early implant osseointegration. Int J Mol Med 2021; 48:191. [PMID: 34414453 PMCID: PMC8416142 DOI: 10.3892/ijmm.2021.5024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022] Open
Abstract
The present study aimed to investigate the effect of β‑receptor blocker propranolol on early osseointegration of pure titanium implants and the underlying molecular regulatory mechanisms. An implant osseointegration model using the tibial metaphysis of New Zealand rabbits was established. The rabbits were divided into control and low‑, medium‑ and high‑dose propranolol groups. The formation of implant osseointegration was detected by X‑ray scanning. Mesenchymal stem cells (MSCs) and osteoblasts (OBs) were isolated and cultured in vitro, isoproterenol was supplemented to simulate sympathetic action and propranolol was subsequently administrated. The effect of propranolol on cell proliferation and osteogenic differentiation were assessed by EdU, flow cytometry, alizarin red staining and alkaline phosphatase (ALP) detection. The expression levels of bone morphogenetic protein (BMP)2, RUNX family transcription factor (RunX)2, collagen (COL)‑1, osteocalcin (OCN) and β2‑adrenergic receptor (AR) were detected by immunofluorescence, reverse transcription‑quantitative PCR and western blot assay. Propranolol effectively promoted implant osseointegration in vivo, facilitated proliferation of OBs, inhibited proliferation of MSCs and enhanced osteogenic differentiation of OBs and MSCs. The calcium content and ALP activity of cells treated with propranolol were markedly higher than in the control group. Propranolol also elevated mRNA and protein expression levels of BMP2, RunX2, COL‑1 and OCN in tissue and cells, and decreased the expression of β2‑AR. The present study demonstrated that the β‑receptor blocker propranolol promoted osteogenic differentiation of OBs and MSCs and enhanced implant osseointegration. The present study provided a novel insight into the application and regulatory mechanisms of propranolol.
Collapse
Affiliation(s)
- Yupeng Wu
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Qi Zhang
- School of Stomatology, Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Baodong Zhao
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Xiaojing Wang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
9
|
Zupan J, Strazar K, Kocijan R, Nau T, Grillari J, Marolt Presen D. Age-related alterations and senescence of mesenchymal stromal cells: Implications for regenerative treatments of bones and joints. Mech Ageing Dev 2021; 198:111539. [PMID: 34242668 DOI: 10.1016/j.mad.2021.111539] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/18/2022]
Abstract
The most common clinical manifestations of age-related musculoskeletal degeneration are osteoarthritis and osteoporosis, and these represent an enormous burden on modern society. Mesenchymal stromal cells (MSCs) have pivotal roles in musculoskeletal tissue development. In adult organisms, MSCs retain their ability to regenerate tissues following bone fractures, articular cartilage injuries, and other traumatic injuries of connective tissue. However, their remarkable regenerative ability appears to be impaired through aging, and in particular in age-related diseases of bones and joints. Here, we review age-related alterations of MSCs in musculoskeletal tissues, and address the underlying mechanisms of aging and senescence of MSCs. Furthermore, we focus on the properties of MSCs in osteoarthritis and osteoporosis, and how their changes contribute to onset and progression of these disorders. Finally, we consider current treatments that exploit the enormous potential of MSCs for tissue regeneration, as well as for innovative cell-free extracellular-vesicle-based and anti-aging treatment approaches.
Collapse
Affiliation(s)
- Janja Zupan
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Klemen Strazar
- Department of Orthopaedic Surgery, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Roland Kocijan
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; Medical Faculty of Bone Diseases, Sigmund Freud University Vienna, 1020, Vienna, Austria
| | - Thomas Nau
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Centre, 1200, Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200, Vienna, Austria; Building 14, Mohamed Bin Rashid University of Medicine and Health Sciences Dubai, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Centre, 1200, Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200, Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, 1180, Vienna, Austria
| | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Centre, 1200, Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200, Vienna, Austria.
| |
Collapse
|
10
|
Naderi A, Zhang B, Belgodere JA, Sunder K, Palardy G. Improved Biocompatible, Flexible Mesh Composites for Implant Applications via Hydroxyapatite Coating with Potential for 3-Dimensional Extracellular Matrix Network and Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2021; 13:26824-26840. [PMID: 34097380 PMCID: PMC8289173 DOI: 10.1021/acsami.1c09034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 05/24/2021] [Indexed: 06/02/2023]
Abstract
Hydroxyapatite (HA)-coated metals are biocompatible composites, which have potential for various applications for bone replacement and regeneration in the human body. In this study, we proposed the design of biocompatible, flexible composite implants by using a metal mesh as substrate and HA coating as bone regenerative stimulant derived from a simple sol-gel method. Experiments were performed to understand the effect of coating method (dip-coating and drop casting), substrate material (titanium and stainless steel) and substrate mesh characteristics (mesh size, weave pattern) on implant's performance. HA-coated samples were characterized by X-ray diffractometer, transmission electron microscope, field-emission scanning electron microscope, nanoindenter, polarization and electrochemical impedance spectroscopy, and biocompatibility test. Pure or biphasic nanorod HA coating was obtained on mesh substrates with thicknesses varying from 4.0 to 7.9 μm. Different coating procedures and number of layers did not affect crystal structure, shape, or most intense plane reflections of the HA coating. Moduli of elasticity below 18.5 GPa were reported for HA-coated samples, falling within the range of natural skull bone. Coated samples led to at least 90% cell viability and up to 99.5% extracellular matrix coverage into a 3-dimensional network (16.4% to 76.5% higher than bare substrates). Fluorescent imaging showed no antagonistic effect of the coatings on osteogenic differentiation. Finer mesh size enhanced coating coverage and adhesion, but a low number of HA layers was preferable to maintain open mesh areas promoting extracellular matrix formation. Finally, electrochemical behavior studies revealed that, although corrosion protection for HA-coated samples was generally higher than bare samples, galvanic corrosion occurred on some samples. Overall, the results indicated that while HA-coated titanium grade 1 showed the best performance as a potential implant, HA-coated stainless steel 316 with the finest mesh size constitutes an adequate, lower cost alternative.
Collapse
Affiliation(s)
- Armaghan Naderi
- Department
of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Bin Zhang
- Department
of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Jorge A. Belgodere
- Department
of Biological & Agricultural Engineering, Louisiana State University and Agricultural Center, Baton Rouge, Louisiana 70803, United States
| | - Kaushik Sunder
- Department
of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Genevieve Palardy
- Department
of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| |
Collapse
|
11
|
Comparison of Osteogenic Differentiation Potential of Human Dental-Derived Stem Cells Isolated from Dental Pulp, Periodontal Ligament, Dental Follicle, and Alveolar Bone. Stem Cells Int 2021; 2021:6631905. [PMID: 33927769 PMCID: PMC8049831 DOI: 10.1155/2021/6631905] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 03/10/2021] [Accepted: 03/24/2021] [Indexed: 01/09/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) have become promising candidates for regeneration medicine due to their multidifferentiation potential and immunomodulatory ability. Compared with classic MSCs derived from the bone marrow and fat, dental-derived MSCs show high plasticity, accessibility, and applicability. Therefore, they are considered alternative sources for regeneration medicine. Methods Four types of MSCs were isolated from the dental pulp, periodontal ligament, dental follicle, and alveolar bone of the same donor, and there were five different individuals. We analyzed their morphology, immunophenotype, proliferation rate, apoptosis, trilineage differentiation potential, and the gene expression during osteogenic differentiation. Results Our research demonstrated that DPSCs, PDLSCs, DFPCs and ABMMSCs exhibited similar morphology and immunophenotype. DFPCs showed a higher rate of proliferation and apoptosis. When cultured in the trilineage differentiation medium, all types of MSCs presented the differentiation potential of osteogenesis, adipogenesis, and chondrogenesis. Through staining and genetic analysis during osteogenic induction, ABMMSCs and PDLSCs showed the highest osteogenic ability, followed by DPSCs, and DFPCs were the lowest. Conclusions Overall, our results indicated that different dental-derived stem cells possessed different biological characteristics. For bone tissue engineering, ABMMSCs and PDLSCs can be used as optimal candidates of seed cells.
Collapse
|
12
|
Li D, Liu J, Yang C, Tian Y, Yin C, Hu L, Chen Z, Zhao F, Zhang R, Lu A, Zhang G, Qian A. Targeting long noncoding RNA PMIF facilitates osteoprogenitor cells migrating to bone formation surface to promote bone formation during aging. Am J Cancer Res 2021; 11:5585-5604. [PMID: 33859765 PMCID: PMC8039942 DOI: 10.7150/thno.54477] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: The migration of mesenchymal osteoprogenitor cells (OPCs) to bone formation surface is the initial step of osteoblastogenesis before they undergo osteoblast differentiation and maturation for governing bone formation. However, whether the migration capacity of OPCs is compromised during aging and how it contributes to the aging-related bone formation reduction remain unexplored. In the present study, we identified a migration inhibitory factor (i.e., long noncoding RNA PMIF) and examined whether targeting lnc-PMIF could facilitate osteoprogenitor cells migrating to bone formation surface to promote bone formation during aging. Methods: Primary OPCs from young (6-momth-old) and aged (18-momth-old) C57BL/6 mice and stable lnc-PMIF knockdown/overexpression cell lines were used for in vitro and in vivo cell migration assay (i.e., wound healing assay, transwell assay and cell intratibial injection assay). RNA pulldown-MS/WB and RIP-qPCR were performed to identify the RNA binding proteins (RBPs) of lnc-PMIF. Truncations of lnc-PMIF and the identified RBP were engaged to determine the interaction motif between them by RNA pulldown-WB and EMSA. By cell-based therapy approach and by pharmacological approach, small interfering RNA (siRNA)-mediated lnc-PMIF knockdown were used in aged mice. The cell migration ability was evaluated by transwell assay and cell intratibial injection assay. The bone formation was evaluated by microCT analysis and bone morphometry analysis. Results: We reported that the decreased bone formation was accompanied by the reduced migration capacity of the bone marrow mesenchymal stem cells (BMSCs, the unique source of OPCs in bone marrow) in aged mice. We further identified that the long non-coding RNA PMIF (postulated migration inhibitory factor) (i.e., lnc-PMIF) was highly expressed in BMSCs from aged mice and responsible for the reduced migration capacity of aged OPCs to bone formation surface. Mechanistically, we found that lnc-PMIF could bind to human antigen R (HuR) for interrupting the HuR-β-actin mRNA interaction, therefore inhibit the expression of β-actin for suppressing the migration of aged OPCs. We also authenticated a functionally conserved human lncRNA ortholog of the murine lnc-PMIF. By cell-based therapy approach, we demonstrated that replenishing the aged BMSCs with small interfering RNA (siRNA)-mediated lnc-PMIF knockdown could promote bone formation in aged mice. By pharmacological approach, we showed that targeted delivery of lnc-PMIF siRNA approaching the OPCs around the bone formation surface could also promote bone formation in aged mice. Conclusion: Toward translational medicine, this study hints that targeting lnc-PMIF to facilitate aged OPCs migrating to bone formation surface could be a brand-new anabolic strategy for aging-related osteoporosis.
Collapse
|
13
|
Liu Y, Holmes C. Tissue Regeneration Capacity of Extracellular Vesicles Isolated From Bone Marrow-Derived and Adipose-Derived Mesenchymal Stromal/Stem Cells. Front Cell Dev Biol 2021; 9:648098. [PMID: 33718390 PMCID: PMC7952527 DOI: 10.3389/fcell.2021.648098] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cell (MSC)-based therapies have demonstrated tissue repair and regeneration capacity in various preclinical models. These therapeutic effects have recently been largely attributed to the paracrine effects of the MSC secretome, including proteins and extracellular vesicles (EVs). EVs are cell-secreted nano-sized vesicles with lipid bilayer membranes that facilitate cell–cell signaling. Treatments based on MSC-derived EVs are beginning to be explored as an alternative to MSC transplantation-based therapies. However, it remains to be determined which MSC source produces EVs with the greatest therapeutic potential. This review compares the tissue regeneration capacity of EVs isolated from the two most common clinical sources of adult MSCs, bone marrow and adipose tissue, with a particular focus on their angiogenic, osteogenic, and immunomodulatory potentials. Other important issues in the development of MSC-derived EV based therapies are also discussed.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, Tallhassee, FL, United States
| | - Christina Holmes
- Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, Tallhassee, FL, United States
| |
Collapse
|
14
|
Khazaei S, Keshavarz G, Bozorgi A, Nazari H, Khazaei M. Adipose tissue-derived stem cells: a comparative review on isolation, culture, and differentiation methods. Cell Tissue Bank 2021; 23:1-16. [PMID: 33616792 DOI: 10.1007/s10561-021-09905-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/01/2021] [Indexed: 01/14/2023]
Abstract
Adipose tissue-derived stem cells (ADSCs) are an available source of mesenchymal stem cells with the appropriate capacity to in vitro survive, propagate, and differentiate into cells from three lineages of ectoderm, mesoderm, and endoderm. The biological features of ADSCs depend on the donor physiology and health status, isolation procedure, culture conditions, and differentiation protocols used. Adipose tissue samples are provided by surgery and lipoaspiration-based methods and subjected to various mechanical and chemical digestion techniques to finally generate a heterogeneous mixture named stromal vascular fraction (SVF). ADSCs are purified through varied cell populations that exist within SVF and cultured under standard conditions to give rise to a highly rich resource of stem cells directly applied in the clinic or differentiated into a wide range of cells. The development and optimization of conventional isolation, expansion, and differentiation methods seem noteworthy to preserve the desirable biological functions of ADSCs in pre-clinical and clinical investigations.
Collapse
Affiliation(s)
- Saber Khazaei
- Department of Endodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghazal Keshavarz
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Azam Bozorgi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Nazari
- Department of Orofacial Surgery, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
15
|
Multiplex Analysis of Adipose-Derived Stem Cell (ASC) Immunophenotype Adaption to In Vitro Expansion. Cells 2021; 10:cells10020218. [PMID: 33499095 PMCID: PMC7911224 DOI: 10.3390/cells10020218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
In order to enhance the therapeutic potential, it is important that sufficient knowledge regarding the dynamic changes of adipose-derived stem cell (ASC) immunophenotypical and biological properties during in vitro growth is available. Consequently, we embarked on a study to follow the evolution of highly defined cell subsets from three unrelated donors in the course of eight passages on tissue culture polystyrene. The co-expression patterns were defined by panels encompassing seven and five cell surface markers, including CD34, CD146, CD166, CD200, CD248, CD271, and CD274 and CD29, CD31, CD36, CD201, and Stro-1, respectively. The analysis was performed using multichromatic flow cytometry. We observed a major paradigm shift, where the CD166-CD34+ combination which was found across all cell subsets early in the culture was replaced by the CD166+ phenotype as the population homogeneity increased with time. At all analysis points, the cultures were dominated by a few major clones that were highly prevalent in most of the donors. The selection process resulted in two predominant clones in the larger panel (CD166+CD34-CD146-CD271- CD274-CD248-CD200- and CD166+CD34+ CD146-CD271-CD274-CD248-CD200-) and one clone in the smaller panel (CD29+CD201+CD36- Stro-1- CD31-). The minor subsets, including CD166+CD34-CD146-CD271+CD274-CD248-CD200- and CD166+CD34+CD146+CD271-CD274-CD248-CD200-, and CD29+CD201-CD36-Stro-1-CD31-, CD29+CD201+CD36-Stro-1+CD31-, and CD29+CD201+CD36+Stro-1-CD31-, in the seven and five marker panels, respectively, were, on the other, hand highly fluctuating and donor-dependent. The results demonstrate that only a limited number of phenotypical repertoires are possible in ASC cultures. Marked differences in their relative occurrence between distinct individuals underscore the need for potency standardization of different ASC preparation to improve the clinical outcome.
Collapse
|
16
|
Kuhlmann C, Schenck TL, Haas EM, Giunta R, Wiggenhauser PS. [Current review of factors in the stem cell donor that influence the regenerative potential of adipose tissue-derived stem cells]. HANDCHIR MIKROCHIR P 2020; 52:521-532. [PMID: 33291167 DOI: 10.1055/a-1250-7878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Regenerative therapies like cell-assisted lipotransfer or preclinical experimental studies use adipose tissue-derived stem cells (ASCs) as the main therapeutic agent. But there are also factors depending on the clinical donor that influence the cell yield and regenerative potential of human ASCs and stromal vascular fraction (SVF). Therefore, the aim of this review was to identify and evaluate these factors according to current literature. METHODS For this purpose, a systematic literature review was performed with focus on factors affecting the regenerative potential of ASCs and SVF using the National Library of Medicine. RESULTS Currently, there is an abundance of studies regarding clinical donor factors influencing ASCs properties. But there is some contradiction and need for further investigation. Nevertheless, we identified several recurrent factors: age, sex, weight, diabetes, lipoedema, use of antidepressants, anti-hormonal therapy and chemotherapy. CONCLUSION We recommend characterisation of the ASC donor cohort in all publications, regardless of whether they are experimental studies or clinical trials. By these means, donor factors that influence experimental or clinical findings can be made transparent and results are more comparable. Moreover, this knowledge can be used for study design to form a homogenous donor cohort by precise clinical history and physical examination.
Collapse
Affiliation(s)
| | | | | | | | - Paul Severin Wiggenhauser
- Klinikum der Universität München, Abteilung Handchirurgie, Plastische Chirurgie, Ästhetische Chirurgie
| |
Collapse
|
17
|
Effects of Harvest Sites on Cryopreserved Adipose-Derived Stem Cells and ASC-Enriched Fat Grafts. Aesthetic Plast Surg 2020; 44:2286-2296. [PMID: 32754834 DOI: 10.1007/s00266-020-01900-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Enrichment of adipose-derived stem cells (ASCs) with fat grafts has demonstrated benefit for graft retention and histologic appearance. There is no consensus on the optimal harvest site for adipose-derived stem cells. This study aimed to investigate the effects of harvest sites on the characteristics of cryopreserved adipose-derived stem cells and the graft retention of cell-assisted lipotransfer. METHODS Lipoaspirates were harvested from 18 healthy volunteers who underwent liposuctions for body contouring. Twenty milliliters of lipoaspirates was, respectively, obtained from four sites, including the upper limb, abdomen, waist, and thighs, by the Coleman technique. Adipose-derived stem cells were ex vivo cultured and cryopreserved for four weeks. The biological characteristics of ASCs from four harvest sites were analyzed: MSC surface markers, cell proliferation, migration ability, and multipotential differentiation. The fat grafts were co-implanted with ASCs from four harvest sites and injected subcutaneously in mice. The ASC-enriched fat grafts were analyzed three months after transplantation. RESULTS Cryopreserved ASCs from the abdomen and thighs maintained more significant cell proliferation, migration ability, and differentiation potential, compared with cells from the upper limb and waist. Moreover, we achieved better graft retention of cell-assisted fat grafts with cryopreserved ASC from the abdomen and thighs. CONCLUSIONS The harvest site of adipose tissue affects the cellular activity and differentiation potential of cryopreserved ASCs. Improved understanding of harvest sites for ASCs can optimize the outcomes of cell-assisted fat grafts. Fat grafts enriched with cryopreserved ASCs from the abdomen or thighs are the optimal choices. LEVEL OF EVIDENCE V This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
18
|
Nie F, Bi H, Zhang C, Ding P. Differentiation potential and mRNA profiles of human dedifferentiated adipose cells and adipose‑derived stem cells from young donors. Mol Med Rep 2020; 23:47. [PMID: 33200799 PMCID: PMC7705993 DOI: 10.3892/mmr.2020.11685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
Dedifferentiated adipose cells (DAs) and adipose-derived stem cells (ADSCs) are two of the primary types of stem cells derived from adipose tissue, which have been reported to possess similar characteristics, but also exhibit unique phenotypic and functional advantages. However, several reports have described inconsistent results regarding their differences in multilineage differentiation function. Moreover, to the best of our knowledge, there are no studies assessing their myogenic ability, or the differences in the transcriptome between the two cell types derived from lipoaspirates via tumescent liposuction from the same donors. The aim of the present study was to compare the properties and expression profiles of these cell types. Subcutaneous adipose tissue of three female patients (aged 23–30 years) with a physiological BMI (19.1–23.9 kg/m2) were obtained during tumescent liposuction of the abdomen or the thigh. The stromal vascular fraction and mature adipocytes were obtained via collagenase digestion, and ADSCs and DAs were cultured successively. To determine the differences between DAs and ADSCs after 6–7 passages, cell proliferation assays, phenotypic assessment, differentiation assays and high-throughput RNA sequencing (seq) were used. Similar cell morphologies, proliferation dynamics, surface markers and transcriptome expression profiles were observed between the DAs and ADSCs. Whilst there were notable individual differences in the osteogenic, lipogenic, chondrogenic and myogenic abilities of the DAs and ADSCs, it was difficult to determine their differentiation potential based only on the cell source. Interestingly, the myogenic ability was relatively stronger in cells with relatively weaker lipogenic ability. Only 186 differentially expressed genes between the two groups were identified using RNAseq. Several of these genes were involved in biological functions such as transcription regulation, protein translation regulation, cytokine interactions and energy metabolism regulation. The results of the present study suggested a similar functional potential of DAs and ADSCs from young donors undergoing tumescent liposuction operation in regeneration areas and the balance of the differentiative ability of the same cell populations. These data may provide a foundation for further clinical administration of stem cells derived from adipose tissues in therapy.
Collapse
Affiliation(s)
- Fangfei Nie
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Hongsen Bi
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Chen Zhang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Pengbing Ding
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| |
Collapse
|
19
|
Adipose Tissue: A Source of Stem Cells with Potential for Regenerative Therapies for Wound Healing. J Clin Med 2020; 9:jcm9072161. [PMID: 32650555 PMCID: PMC7408846 DOI: 10.3390/jcm9072161] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/25/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
Interest in adipose tissue is fast becoming a focus of research after many years of being considered as a simple connective tissue. It is becoming increasingly apparent that adipose tissue contains a number of diverse cell types, including adipose-derived stem cells (ASCs) with the potential to differentiate into a number of cell lineages, and thus has significant potential for developing therapies for regenerative medicine. Currently, there is no gold standard treatment for scars and impaired wound healing continues to be a challenge faced by clinicians worldwide. This review describes the current understanding of the origin, different types, anatomical location, and genetics of adipose tissue before discussing the properties of ASCs and their promising applications for tissue engineering, scarring, and wound healing.
Collapse
|
20
|
Xiang Q, Xu F, Li Y, Liu X, Chen Q, Huang J, Yu N, Zeng Z, Yuan M, Zhang Q, Long X, Zhou Z. Transcriptome analysis and functional identification of adipose-derived mesenchymal stem cells in secondary lymphedema. Gland Surg 2020; 9:558-574. [PMID: 32420291 DOI: 10.21037/gs.2020.02.09] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Secondary lymphedema is a common condition that affects patients with malignant tumors. Conservative treatments fail to provide lasting relief because they do not address the underlying pathological accumulation of excessive fat. Our aim is to clarify the molecular mechanisms of abnormal adipogenic differentiation in lymphedema adipose tissue. Methods We compared the proliferation and adipogenesis potential of adipose-derived mesenchymal stem cells (ASCs) from the lymphedema adipose tissue from liposuction specimens of 10 patients with extremity lymphedema with that of ASCs from adipose tissue from the normal upper abdomen of the same patients. Transcriptome analysis were performed to identify the differences between the two kinds of ASCs. Cyclin-dependent kinase 1 (CDK1) inhibitors were used to treat the abnormal ASCs in lymphedema adipose tissue. Results Our results demonstrate that significant functional and transcriptomic differences exist between the two kinds of ASCs. Up-regulated genes were mainly involved in cell proliferation and division while down-regulated genes were mainly associated with immune responses and inflammatory as well as osteogenic and myogenic differentiation. Furthermore, we find that the excessive proliferation and adipogenesis of ASCs from lymphedema adipose tissue returned to the normal phenotype by CDK1 inhibitors. ASCs from lymphedema adipose tissues have higher immunosuppressive effect and the cytokines related to immunosuppressive was significantly up-regulated. Conclusions In conclusion, lymphedema-associated ASCs had more rapid proliferation and a higher adipogenic differentiation capacity. CDK1 may be a key driver of proliferation and adipogenic differentiation in these cells, which might expound the accumulation of adipose tissue extensively observed in secondary lymphedema. ASCs from lymphedema adipose tissues showed immunomodulation dysfunction and immunomodulation may play an important role in the pathogenesis of lymphedema.
Collapse
Affiliation(s)
- Qinqin Xiang
- Prenatal Diagnosis Center, Department of Obstetrics & Gynecologic, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Center of Laboratory Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Fen Xu
- Center of Laboratory Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yunzhu Li
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xuanyu Liu
- Center of Laboratory Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Qianlong Chen
- Center of Laboratory Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jiuzuo Huang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Nanze Yu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ziyi Zeng
- Center of Laboratory Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Meng Yuan
- Center of Laboratory Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Qixu Zhang
- Plastic Surgery Department, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiao Long
- Department of Plastic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zhou Zhou
- Center of Laboratory Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
21
|
Peng Q, Alipour H, Porsborg S, Fink T, Zachar V. Evolution of ASC Immunophenotypical Subsets During Expansion In Vitro. Int J Mol Sci 2020; 21:E1408. [PMID: 32093036 PMCID: PMC7073142 DOI: 10.3390/ijms21041408] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 12/18/2022] Open
Abstract
Adipose-derived stromal/stem cells (ASCs) are currently being considered for clinical use for a number of indications. In order to develop standardized clinical protocols, it is paramount to have a full characterization of the stem cell preparations. The surface marker expression of ASCs has previously been characterized in multiple studies. However, most of these studies have provided a cross-sectional description of ASCs in either earlier or later passages. In this study, we evaluate the dynamic changes of 15 different surface molecules during culture. Using multichromatic flow cytometry, ASCs from three different donors each in passages 1, 2, 4, 6, and 8 were analyzed for their co-expression of markers associated with mesenchymal stem cells, wound healing, immune regulation, ASC markers, and differentiation capacity, respectively. We confirmed that at an early stage, ASC displayed a high heterogeneity with a plethora of subpopulations, which by culturing became more homogeneous. After a few passages, virtually all ASCs expressed CD29, CD166 and CD201, in addition to canonical markers CD73, CD90, and CD105. However, even at passage 8, there were several predominant lineages that differed with respect to the expression of CD34, CD200 and CD271. Although the significance of remaining subpopulations still needs to be elucidated, our results underscore the necessity to fully characterize ASCs prior to clinical use.
Collapse
Affiliation(s)
| | | | | | | | - Vladimir Zachar
- Department of Health Science and Technology, Regenerative Medicine Group, Aalborg University, Fredrik Bajers Vej 3B, 9220 Aalborg, Denmark; (Q.P.); (H.A.); (S.P.); (T.F.)
| |
Collapse
|
22
|
Marolt Presen D, Traweger A, Gimona M, Redl H. Mesenchymal Stromal Cell-Based Bone Regeneration Therapies: From Cell Transplantation and Tissue Engineering to Therapeutic Secretomes and Extracellular Vesicles. Front Bioeng Biotechnol 2019; 7:352. [PMID: 31828066 PMCID: PMC6890555 DOI: 10.3389/fbioe.2019.00352] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Effective regeneration of bone defects often presents significant challenges, particularly in patients with decreased tissue regeneration capacity due to extensive trauma, disease, and/or advanced age. A number of studies have focused on enhancing bone regeneration by applying mesenchymal stromal cells (MSCs) or MSC-based bone tissue engineering strategies. However, translation of these approaches from basic research findings to clinical use has been hampered by the limited understanding of MSC therapeutic actions and complexities, as well as costs related to the manufacturing, regulatory approval, and clinical use of living cells and engineered tissues. More recently, a shift from the view of MSCs directly contributing to tissue regeneration toward appreciating MSCs as "cell factories" that secrete a variety of bioactive molecules and extracellular vesicles with trophic and immunomodulatory activities has steered research into new MSC-based, "cell-free" therapeutic modalities. The current review recapitulates recent developments, challenges, and future perspectives of these various MSC-based bone tissue engineering and regeneration strategies.
Collapse
Affiliation(s)
- Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andreas Traweger
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Spinal Cord Injury & Tissue Regeneration Center Salzburg, Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
23
|
Paula ACC, Carvalho PH, Martins TMM, Boeloni JN, Cunha PS, Novikoff S, Correlo VM, Reis RL, Goes AM. Improved vascularisation but inefficient in vivo bone regeneration of adipose stem cells and poly-3-hydroxybutyrate-co-3-hydroxyvalerate scaffolds in xeno-free conditions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 107:110301. [PMID: 31761156 DOI: 10.1016/j.msec.2019.110301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/24/2019] [Accepted: 10/10/2019] [Indexed: 01/26/2023]
Abstract
Bone defects are a common clinical situation. However, bone regeneration remains a challenge and faces the limitation of poor engraftment due to deficient vascularisation. Poly-3-hydroxybutyrate-co-3-hydroxyvalerate (PHB-HV) and human adipose stem cells (hASC) are promising for vascularisation and bone regeneration. Therefore, we sought to investigate the bone regenerative capacity of hASCs cultured in allogeneic human serum (aHS) and PHB-HV scaffolds in a nude mouse model of the critical-sized calvarial defect. We evaluated bone healing for three treatment groups: empty (control), PHB-HV and PHB-HV + hASCs. The pre-implant analysis showed that hASCs colonised the PHB-HV scaffolds maintaining cell viability before implantation. Histological analysis revealed that PHB-HV scaffolds were tolerated in vivo; they integrated with adjacent tissue eliciting a response like a foreign body reaction, and tiny primary bone was observed only in the PHB-HV group. Also, the μ-CT analysis revealed only approximately 10% of new bone in the bone defect area in both the PHB-HV and PHB-HV + hASCs groups. The expression of BGLAP and its protein (osteocalcin) by PHB-HV + hASCs group and native bone was similar while the other bone markers RUNX2, ALPL and COL1A1 were upregulated, but this expression remained significantly lower compared to the native bone. Nevertheless, the PHB-HV group showed neovascularisation at 12 weeks post-implantation while PHB-HV + hASCs group also exhibited higher VEGFA expression as well as a higher number of vessels at 4 weeks post-implantation, and, consequently, earlier neovascularisation. This neovascularisation must be due to scaffold architecture, improved by hASCs, that survived for the long term in vivo in the PHB-HV + hASCs group. These results demonstrated that hASCs cultured in aHS combined with PHB-HV scaffolds were ineffective to promote bone regeneration, although the construct of hASCs + PHB-HV in xeno-free conditions improved scaffold vascularisation representing a strategy potentially promising for other tissue engineering applications.
Collapse
Affiliation(s)
- Ana C C Paula
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte, 31270-901, MG, Brazil; Department of Pharmaceutical Sciences, School of Pharmacy, Federal University of Juiz de Fora, R. José Lourenço Kelmer- s/n, Juiz de Fora, 36036-900, MG, Brazil.
| | - Pablo H Carvalho
- Department of Clinical and Surgery, College of Veterinary Medicine, Federal University of Minas Gerais, Av. Presidente Antônio Carlos- 6627, Belo Horizonte, 31270-901, MG, Brazil
| | - Thaís M M Martins
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Presidente Antônio Carlos- 6627, Belo Horizonte, 31270-901, MG, Brazil
| | - Jankerle N Boeloni
- Department of Veterinary Medicine, Federal University of Espírito Santo, Alto Universitário, Alegre, 29500-000, ES, Brazil
| | - Pricila S Cunha
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte, 31270-901, MG, Brazil
| | - Silviene Novikoff
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte, 31270-901, MG, Brazil; Transplants Immunobiology Laboratory, Department of Immunology, University of São Paulo, Brazil
| | - Vitor M Correlo
- 3B´s Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial de Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - Associate Laboratory, PT Government Associate Laboratory, Campus de Gualtar, 4710-057, Braga, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Rui L Reis
- 3B´s Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial de Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - Associate Laboratory, PT Government Associate Laboratory, Campus de Gualtar, 4710-057, Braga, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Alfredo M Goes
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte, 31270-901, MG, Brazil; Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Presidente Antônio Carlos- 6627, Belo Horizonte, 31270-901, MG, Brazil
| |
Collapse
|
24
|
Musorina AS, Zenin VV, Turilova VI, Yakovleva TK, Poljanskaya GG. Characterization of a Nonimmortalized Mesenchymal Stem Cell Line Isolated from Human Epicardial Adipose Tissue. ACTA ACUST UNITED AC 2019. [DOI: 10.1134/s1990519x19040060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
25
|
Häussling V, Deninger S, Vidoni L, Rinderknecht H, Ruoß M, Arnscheidt C, Athanasopulu K, Kemkemer R, Nussler AK, Ehnert S. Impact of Four Protein Additives in Cryogels on Osteogenic Differentiation of Adipose-Derived Mesenchymal Stem Cells. Bioengineering (Basel) 2019; 6:E67. [PMID: 31394780 PMCID: PMC6784125 DOI: 10.3390/bioengineering6030067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/30/2019] [Accepted: 08/05/2019] [Indexed: 12/21/2022] Open
Abstract
Human adipose-derived mesenchymal stem/stromal cells (Ad-MSCs) have great potential for bone tissue engineering. Cryogels, mimicking the three-dimensional structure of spongy bone, represent ideal carriers for these cells. We developed poly(2-hydroxyethyl methacrylate) cryogels, containing hydroxyapatite to mimic inorganic bone matrix. Cryogels were additionally supplemented with different types of proteins, namely collagen (Coll), platelet-rich plasma (PRP), immune cells-conditioned medium (CM), and RGD peptides (RGD). The different protein components did not affect scaffolds' porosity or water-uptake capacity, but altered pore size and stiffness. Stiffness was highest in scaffolds with PRP (82.3 kPa), followed by Coll (55.3 kPa), CM (45.6 kPa), and RGD (32.8 kPa). Scaffolds with PRP, CM, and Coll had the largest pore diameters (~60 µm). Ad-MSCs were osteogenically differentiated on these scaffolds for 14 days. Cell attachment and survival rates were comparable for all four scaffolds. Runx2 and osteocalcin levels only increased in Ad-MSCs on Coll, PRP and CM cryogels. Osterix levels increased slightly in Ad-MSCs differentiated on Coll and PRP cryogels. With differentiation alkaline phosphatase activity decreased under all four conditions. In summary, besides Coll cryogel our PRP cryogel constitutes as an especially suitable carrier for bone tissue engineering. This is of special interest, as this scaffold can be generated with patients' PRP.
Collapse
Affiliation(s)
- Victor Häussling
- Siegfried Weller Research Institute, BG Unfallklinik Tuebingen, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Sebastian Deninger
- Siegfried Weller Research Institute, BG Unfallklinik Tuebingen, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Laura Vidoni
- Siegfried Weller Research Institute, BG Unfallklinik Tuebingen, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Helen Rinderknecht
- Siegfried Weller Research Institute, BG Unfallklinik Tuebingen, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Marc Ruoß
- Siegfried Weller Research Institute, BG Unfallklinik Tuebingen, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Christian Arnscheidt
- Siegfried Weller Research Institute, BG Unfallklinik Tuebingen, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Kiriaki Athanasopulu
- Department of Applied Chemistry Reutlingen University, 72762 Reutlingen, Germany
| | - Ralf Kemkemer
- Department of Applied Chemistry Reutlingen University, 72762 Reutlingen, Germany
| | - Andreas K Nussler
- Siegfried Weller Research Institute, BG Unfallklinik Tuebingen, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, 72074 Tübingen, Germany.
| | - Sabrina Ehnert
- Siegfried Weller Research Institute, BG Unfallklinik Tuebingen, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| |
Collapse
|
26
|
Abstract
Adipose stem cells (ASCs) are the basis of procedures intended for tissue regeneration. These cells are heterogeneous, owing to various factors, including the donor age, sex, body mass index, and clinical condition; the isolation procedure (liposuction or fat excision); the place from where the cells were sampled (body site and depth of each adipose depot); culture surface; type of medium (whether supplemented with fetal bovine serum or xeno-free), that affect the principal phenotypic features of ASCs. The features related to ASCs heterogeneity are relevant for the success of therapeutic procedures; these features include proliferation capacity, differentiation potential, immunophenotype, and the secretome. These are important characteristics for the success of regenerative tissue engineering, not only because of their effects upon the reconstruction and healing exerted by ASCs themselves, but also because of the paracrine signaling of ASCs and its impact on recipient tissues. Knowledge of sources of heterogeneity will be helpful in the standardization of ASCs-based procedures. New avenues of research could include evaluation of the effects of the use of more homo1geneous ASCs for specific purposes, the study of ASCs-recipient interactions in heterologous cell transplantation, and the characterization of epigenetic changes in ASCs, as well as investigations of the effect of the metabolome upon ASCs behavior in culture.
Collapse
|
27
|
Silicon-Substituted Hydroxyapatite Particles and Response of Adipose Stem Cells In Vitro. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00108-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
28
|
Shen C, Yang C, Xu S, Zhao H. Comparison of osteogenic differentiation capacity in mesenchymal stem cells derived from human amniotic membrane (AM), umbilical cord (UC), chorionic membrane (CM), and decidua (DC). Cell Biosci 2019; 9:17. [PMID: 30792848 PMCID: PMC6371545 DOI: 10.1186/s13578-019-0281-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/04/2019] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have been extensively explored as a promising therapeutic agent in the field of bone tissue engineering due to their osteogenic differentiation ability. In this study, the osteogenic differential ability and the effect of fibronectin and laminin on the osteogenic differentiation in four types of MSCs derived from placental tissue are compared to determine the ideal source for bone reconstruction tissue engineering. RESULTS The present study examines the osteogenic differentiation levels of four types of MSCs using alizarin red staining and quantifies the calcium levels and alkaline phosphatase (ALP) activity. In addition, this study examines the osteoblast differentiation protein markers osterix, collagen I, osteopontin, and osteocalcin using a Western blot assay. qPCR and EdU labeling assays were employed to identify the kinetics of osteogenic differentiation. Calcium deposit levels, ALP activity, and osteopontin and osteocalcin concentrations were determined to confirm the role of Extracellular matrix (ECM) components role on the osteogenic differentiation of MSCs. The data demonstrated that MSCs isolated from different layers of placenta had different potentials to differentiate into osteogenic cells. Importantly, AM-MSCs and UC-MSCs differentiated into the osteoblast stage more efficiently and quickly than CM-MSCs and DC-MSCs, which was associated with a decrease in their proliferation ability. Among the different types of MSCs, AM-MSCs and UC-MSCs had a higher osteogenic differentiation potential induced by fibronectin due to enhanced phosphorylation during the Akt and ERK pathways. CONCLUSIONS Taken together, these results indicate that AM-MSCs and UC-MSCs possess a higher osteogenic potential, and fibronectin can robustly enhance the osteogenic potential of the Akt and ERK pathways.
Collapse
Affiliation(s)
- Chongyang Shen
- Basic Medicine School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Chuan Yang
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Shijun Xu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Hai Zhao
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|