51
|
Kim M, Jee SC, Sung JS, Kadam AA. Supermagnetic Sugarcane Bagasse Hydrochar for Enhanced Osteoconduction in Human Adipose Tissue-Derived Mesenchymal Stem Cells. NANOMATERIALS 2020; 10:nano10091793. [PMID: 32916934 PMCID: PMC7557583 DOI: 10.3390/nano10091793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/21/2022]
Abstract
Hydrothermally carbonized sugarcane bagasse (SCB) has exceptional surface properties. Looking at the huge amount of SCB produced, its biocompatible nature, cheap-cost for carbonization, and its easy functionalization can give impeccable nano-biomaterials for tissue engineering applications. Herein, sugarcane bagasse was converted into hydrochar (SCB-H) by hydrothermal carbonation. The SCB-H produced was further modified with iron oxide (Fe3O4) nanoparticles (denoted as SCB-H@Fe3O4). Facile synthesized nano-bio-composites were characterized by SEM, HR-TEM, XRD, FT-IR, XPS, TGA, and VSM analysis. Bare Fe3O4 nanoparticles (NPs), SCB-H, and SCB-H@Fe3O4 were tested for cytocompatibility and osteoconduction enhancement of human adipose tissue-derived mesenchymal stem cells (hADMSCs). The results confirmed the cytocompatible and nontoxic nature of SCB-H@Fe3O4. SCB-H did not show enhancement in osteoconduction, whilst on the other hand, Fe3O4 NPs exhibited a 0.5-fold increase in the osteoconduction of hADMSCs. However, SCB-H@Fe3O4 demonstrated an excellent enhancement in osteoconduction of a 3-fold increase over the control, and a 2.5-fold increase over the bare Fe3O4 NPs. Correspondingly, the expression patterns assessment of osteoconduction marker genes (ALP, OCN, and RUNX2) confirmed the osteoconductive enhancement by SCB-H@Fe3O4. In the proposed mechanism, the surface of SCB-H@Fe3O4 might provide a unique topology, and anchoring to receptors of hADMSCs leads to accelerated osteogenesis. In conclusion, agriculture waste-derived sustainable materials like “SCB-H@Fe3O44” can be potentially applied in highly valued medicinal applications of stem cell differentiation.
Collapse
Affiliation(s)
- Min Kim
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University-Seoul, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyonggido 10326, Korea; (M.K.); (S.-C.J.); (J.-S.S.)
| | - Seung-Cheol Jee
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University-Seoul, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyonggido 10326, Korea; (M.K.); (S.-C.J.); (J.-S.S.)
| | - Jung-Suk Sung
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University-Seoul, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyonggido 10326, Korea; (M.K.); (S.-C.J.); (J.-S.S.)
| | - Avinash A. Kadam
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyonggido 10326, Korea
- Correspondence: or ; Tel.: +82-31-961-5616; Fax: 82-31-961-5108
| |
Collapse
|
52
|
Onak G, Gökmen O, Yaralı ZB, Karaman O. Enhanced osteogenesis of human mesenchymal stem cells by self-assembled peptide hydrogel functionalized with glutamic acid templated peptides. J Tissue Eng Regen Med 2020; 14:1236-1249. [PMID: 32615018 DOI: 10.1002/term.3095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
Self-assembling peptide (SAP) hydrogel has been shown to be an excellent biological material for three-dimensional cell culture and stimulatie cell migration and differentiation into the scaffold, as well as for repairing bone tissue defects. Herein, we designed one of the SAP scaffolds KLD (KLDLKLDLKLDL) through direct coupling to short bioactive motif O1 (EEGGC) and O2 (EEEEE) of which bioactivity on osteogenic differentiation was previously demonstrated and self-assembled in different concentrations (0.5%, 1%, and 2%). Our aim was to enhance osteogenesis and biomineralization of injectable SAP hydrogels with controlled mechanical properties so that the peptide hydrogel also becomes capable of being injected to bone defects. The molecular integration of the nanofibrous peptide scaffolds was observed using atomic force microscopy (AFM) and scanning electron microscopy (SEM). The rheological properties and degradation profile of SAP hydrogels were evaluated to ensure stability of SAPs. Compared with pure KLD scaffold, we found that these designed bioactive peptide scaffolds significantly promoted hMSCs proliferation depicted by biochemical analysis of alkaline phosphatase (ALP) activity, total calcium deposition. Moreover, key osteogenic markers of ALP activity, collagen type I (COL-1), osteopontin (OP), and osteocalcin (OCN) expression levels determined by real-time polymerase chain reaction (PCR) and immunofluorescence analysis were also significantly increased with the addition of glutamic acid residues to KLD. We demonstrated that the designed SAP scaffolds promoted the proliferation and osteogenic differentiation of hMSCs. Our results suggest that these designed bioactive peptide scaffolds may be useful for promoting bone tissue regeneration.
Collapse
Affiliation(s)
- Günnur Onak
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
| | - Oğuzhan Gökmen
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
| | - Ziyşan Buse Yaralı
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
| | - Ozan Karaman
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey.,Bonegraft Biomaterials Co., Ege University Technopolis, İzmir, Turkey
| |
Collapse
|
53
|
McColloch A, Liebman C, Liu H, Cho M. Alterted Adipogenesis of Human Mesenchymal Stem Cells by Photobiomodulation Using 1064 nm Laser Light. Lasers Surg Med 2020; 53:263-274. [PMID: 32495397 DOI: 10.1002/lsm.23278] [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: 02/26/2020] [Revised: 04/27/2020] [Accepted: 05/23/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Photobiomodulation (PBM) describes the influence of light irradiation on biological tissues. Laser light in the near-infrared (NIR) spectrum has been shown to mitigate pain, reduce inflammation, and promote wound healing. The cellular mechanism that mediates PBM's effects is generally accepted to be at the site of the mitochondria, leading to an increased flux through the electron transport chain and adenosine triphosphate (ATP) production. Moreover, PBM has been demonstrated to reduce oxidative stress through an increased production of reactive oxygen species (ROS)-sequestering enzymes. The aim of the study is to determine whether these PBM-induced effects expedite or interfere with the intended stem cell differentiation to the adipogenic lineage. STUDY DESIGN/MATERIALS AND METHODS To determine the effects of 1064 nm laser irradiation (fluence of 8.8-26.4 J/cm2 ) on human mesenchymal stem cells (hMSCs) undergoing adipogenic differentiation, the ATP and ROS levels, and adipogenic markers were quantitatively measured. RESULTS At a low fluence (8.8 J/cm2 ) the ATP increase was essentially negligible, whereas a higher fluence induced a significant increase. In the laser-stimulated cells, PBM over time decreased the ROS level compared with the non-treated control group and significantly reduced the extent of adipogenesis. A reduction in the ROS level was correlated with a diminished lipid accumulation, reduced production of adipose-specific genetic markers, and delayed the chemically intended adipogenesis. CONCLUSION We characterized the use of NIR light exposure to modulate adipogenesis. Both the ATP and ROS levels in hMSCs responded to different energy densities. The current study is expected to contribute significantly to the growing field of PBM as well as stem cell tissue engineering by demonstrating the wavelength-dependent responses of hMSC differentiation. Lasers Surg. Med. © 2020 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Andrew McColloch
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas, 76019
| | - Caleb Liebman
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas, 76019
| | - Hanli Liu
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas, 76019
| | - Michael Cho
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas, 76019
| |
Collapse
|
54
|
Zohora FT, Aldebs AI, Nosoudi N, Singh SP, Ramirez-Vick JE. Gene Expression Profiling of Human Adipose Tissue Stem Cells during 2D versus 3D Adipogenesis. Cells Tissues Organs 2020; 208:113-133. [PMID: 32464628 DOI: 10.1159/000507187] [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: 01/09/2020] [Accepted: 03/10/2020] [Indexed: 11/19/2022] Open
Abstract
Much of the current understanding on molecular and cellular events of adipose developmental biology comes from monolayer cell culture models using preadipocyte cell lines, although in vivo adipose tissue consists of a much more complex three-dimensional microenvironment of diverse cell types, extracellular network, and tissue-specific morphological and functional features. Added to this fact, the preadipocytes, on which the adipogenesis mechanisms are mostly explored, possess some serious limitations (e.g., time of initial subculture and adipogenic differentiation time), which, perhaps, can efficiently be replaced with progenitor cells such as adipose tissue-derived stem cells (ASCs). With the objective of developing a better in vitro model for adipose developmental biology, this project involves gene expression profiling of human ASCs (hASCs) during their differentiation to adipocytes in a 2D versus 3D culture model. This transcriptional-level analysis revealed that gene expression patterns of adipogenesis-induced hASCs in a 3D self-assembled polypeptide hydrogel are relatively different from the 2D monolayered cells on plastic hard substrate. Moreover, analysis of adipogenic lineage progression 9 days after adipogenic induction shows earlier differentiation of hASCs in 2D over their 3D counterparts. However, differentiation in 2D shows some unexpected behavior in terms of gene expression, which does not seem to be related to adipogenic lineage specification. Since hASCs are already being used in clinical trials due to their therapeutic potential, it is important to have a clear understanding of the molecular mechanisms in an in vivo model microenvironment like the one presented here.
Collapse
Affiliation(s)
- Fatema Tuj Zohora
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, Dayton, Ohio, USA
| | - Alyaa Isam Aldebs
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, Dayton, Ohio, USA
| | - Nasim Nosoudi
- Biomedical Engineering Program,Marshall University, Huntington, West Virginia, USA
| | - Surinder Pal Singh
- CSIR-National Physical Laboratory, Dr. K.S. Krishanan Marg, New Delhi, India
| | - Jaime Eduardo Ramirez-Vick
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, Dayton, Ohio, USA,
| |
Collapse
|
55
|
Rühle A, Thomsen A, Saffrich R, Voglstätter M, Bieber B, Sprave T, Wuchter P, Vaupel P, Huber PE, Grosu AL, Nicolay NH. Multipotent mesenchymal stromal cells are sensitive to thermic stress – potential implications for therapeutic hyperthermia. Int J Hyperthermia 2020; 37:430-441. [DOI: 10.1080/02656736.2020.1758350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Thomsen
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rainer Saffrich
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg-Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Maren Voglstätter
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Birgit Bieber
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tanja Sprave
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg-Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Peter Vaupel
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter E. Huber
- Department of Molecular Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H. Nicolay
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
56
|
Liu Z, Liu J, Cui X, Wang X, Zhang L, Tang P. Recent Advances on Magnetic Sensitive Hydrogels in Tissue Engineering. Front Chem 2020; 8:124. [PMID: 32211375 PMCID: PMC7068712 DOI: 10.3389/fchem.2020.00124] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Tissue engineering is a promising strategy for the repair and regeneration of damaged tissues or organs. Biomaterials are one of the most important components in tissue engineering. Recently, magnetic hydrogels, which are fabricated using iron oxide-based particles and different types of hydrogel matrices, are becoming more and more attractive in biomedical applications by taking advantage of their biocompatibility, controlled architectures, and smart response to magnetic field remotely. In this literature review, the aim is to summarize the current development of magnetically sensitive smart hydrogels in tissue engineering, which is of great importance but has not yet been comprehensively viewed.
Collapse
Affiliation(s)
- Zhongyang Liu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jianheng Liu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Xiang Cui
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| |
Collapse
|
57
|
Varma DM, DiNicolas MS, Nicoll SB. Injectable, redox-polymerized carboxymethylcellulose hydrogels promote nucleus pulposus-like extracellular matrix elaboration by human MSCs in a cell density-dependent manner. J Biomater Appl 2019; 33:576-589. [PMID: 30326804 DOI: 10.1177/0885328218805216] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Low back pain is a major cause for disability and is closely linked to intervertebral disc degeneration. Mechanical and biological dysfunction of the nucleus pulposus in the disc has been found to initiate intradiscal degenerative processes. Replacing or enriching the diseased nucleus pulposus with an injectable, stem cell-laden biomaterial that mimics its material properties can provide a minimally invasive strategy for biological and structural repair of the tissue. In this study, injectable, in situ-gelling carboxymethylcellulose hydrogels were developed for nucleus pulposus tissue engineering using encapsulated human marrow-derived mesenchymal stromal cells (hMSCs). With the goal of obtaining robust extracellular matrix deposition and faster construct maturation, two cell-seeding densities, 20 × 106 cells/ml and 40 × 106 cells/ml, were examined. The constructs were fabricated using a redox initiation system to yield covalently crosslinked, cell-seeded hydrogels via radical polymerization. Chondrogenic culture of the hydrogels over 35 days exhibited high cell viability along with deposition of proteoglycan and collagen-rich extracellular matrix, and mechanical and swelling properties similar to native human nucleus pulposus. Further, the matrix production and distribution in the carboxymethylcellulose hydrogels was found to be strongly influenced by hMSC-seeding density, with the lower cell-seeding density yielding a more favorable nucleus pulposus-specific matrix phenotype, while the rate of construct maturation was less dependent on the cell-seeding density. These findings are the first to demonstrate the utility of redox-polymerized carboxymethylcellulose hydrogels as hMSC carriers for potential minimally invasive treatment strategies for nucleus pulposus replacement.
Collapse
Affiliation(s)
- Devika M Varma
- The City College of the City University of New York, New York, NY, USA
| | | | - Steven B Nicoll
- The City College of the City University of New York, New York, NY, USA
| |
Collapse
|
58
|
Mao Y, Hoffman T, Dhall S, Singal A, Sathyamoorthy M, Danilkovitch A, Kohn J. Endogenous viable cells in lyopreserved amnion retain differentiation potential and anti-fibrotic activity in vitro. Acta Biomater 2019; 94:330-339. [PMID: 31176843 DOI: 10.1016/j.actbio.2019.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023]
Abstract
Human amniotic membrane (AM) has intrinsic anti-inflammatory, anti-fibrotic and antimicrobial properties. Tissue preservation methods have helped to overcome the short shelf life of fresh AM allowing "on demand" use of AM grafts. Cryopreserved AM that retains all native tissue components, including viable cells, has clinical benefits in treating chronic wounds. However, cryopreservation requires ultra-low temperature storage, limiting the use of cryopreserved products. To overcome this limitation, a new lyopreservation method has been developed for ambient storage of living tissues. The goal of this study was to investigate the viability and functionality of AM cells following lyopreservation. Fresh AM and devitalized lyopreserved AM (DLAM) served as positive and negative controls, respectively. Using live/dead staining, we confirmed the presence of living cells in viable lyopreserved AM (VLAM) and showed that these cells persisted up to 21 days in culture medium. The functionality of cells in VLAM was assessed by their differentiation potential and anti-fibrotic activity in vitro. With osteogenic induction, cells in VLAM deposited calcium within the membrane, a marker of osteogenic cells, in a time-dependent manner. The migration of human lung fibrotic fibroblasts in a scratch wound assay was reduced significantly in the presence of VLAM-derived conditioned medium. Quantitative PCR analyses indicated that VLAM reduced the expression of pro-fibrotic factors such as type I collagen and increased the expression of anti-fibrotic factors such as hepatocyte growth factor and anti-fibrotic microRNA in fibrotic fibroblasts. Taken together, these results demonstrate that endogenous cells in VLAM remain viable and functional post-lyophilization. STATEMENT OF SIGNIFICANCE: This study, for the first time, provides direct evidence showing that tissue viability and functional cells can be preserved by lyophilization. Similar to fresh amniotic membrane (AM), viable lyopreserved AM (VLAM) retains viable cells for extended periods of time. More importantly, these cells are functional and maintain their osteogenic differentiation potential and anti-fibrotic activity. Our results confirmed that the novel lyophilization method preserves tissue viability.
Collapse
|
59
|
Thermal cycling effect on osteogenic differentiation of MC3T3-E1 cells loaded on 3D-porous Biphasic Calcium Phosphate (BCP) scaffolds for early osteogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110027. [PMID: 31546388 DOI: 10.1016/j.msec.2019.110027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/21/2019] [Accepted: 07/26/2019] [Indexed: 12/30/2022]
Abstract
The application of heat stress on a defect site during the healing process is a promising technique for early bone regeneration. The primary goal of this study was to investigate the effect of periodic heat shock on bone formation. MC3T3-E1 cells were seeded onto biphasic calcium phosphate (BCP) scaffolds, followed by periodic heating to evaluate osteogenic differentiation. Heat was applied to cells seeded onto scaffolds at 41 °C for 1 h once, twice, and four times a day for seven days and their viability, morphology, and differentiation were analyzed. BCP scaffolds with interconnected porous structures mimic bone biology for cellular studies. MTT and confocal studies have shown that heat shock significantly increased cell proliferation without any toxic effects. Compared to non-heated samples, heat shock enhanced calcium deposition and mineralization, which could be visualized by SEM observation and Alizarin red S staining. Immunostaining images showed the localization of osteogenic proteins ALP and OPN on heat-shocked cells. qRT-PCR analysis revealed the presence of more osteospecific markers, osteopontin (OPN), osteocalcin, collagen type X, and Runx2, in the heat-shocked samples than in the non-heated sample. Periodic heat shock significantly upregulated both heat shock proteins (HSP70 and HSP27) in differentiated MC3T3-E1 cells. The results of this study demonstrated that periodically heat applied especially two times a day was better approach for osteogenic differentiation. Hence, this work provides a define temperature and time schedule for the development of a clinical heating device in future for early bone regeneration during the postsurgical period.
Collapse
|
60
|
Saadeldin IM, Swelum AAA, Noreldin AE, Tukur HA, Abdelazim AM, Abomughaid MM, Alowaimer AN. Isolation and Culture of Skin-Derived Differentiated and Stem-Like Cells Obtained from the Arabian Camel ( Camelus dromedarius). Animals (Basel) 2019; 9:ani9060378. [PMID: 31226810 PMCID: PMC6616910 DOI: 10.3390/ani9060378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 12/22/2022] Open
Abstract
Simple Summary This is the first comprehensive study to isolate different cellular types and stem-like cells from the camel skin. We reported the multipotency of the isolated stem cells. Moreover, some unique cells were observed, such as dermal cyst-forming cells. This discovery represents a cheap and easy source for camel stem cells that is essential for development of the elite camel regenerative medicine and provides a good source of camel fibroblast required for camel cloning. Abstract Elite camels often suffer from massive injuries. Thus, there is a pivotal need for a cheap and readily available regenerative medicine source. We isolated novel stem-like cells from camel skin and investigated their multipotency and resistance against various stresses. Skin samples were isolated from ears of five camels. Fibroblasts, keratinocytes, and spheroid progenitors were extracted. After separation of different cell lines by trypsinization, all cell lines were exposed to heat shock. Then, fibroblasts and dermal cyst-forming cells were examined under cryopreservation. Dermal cyst-forming cells were evaluated for resistance against osmotic pressure. The results revealed that resistance periods against trypsin were 1.5, 4, and 7 min for fibroblasts, keratinocytes, and spheroid progenitors, respectively. Furthermore, complete recovery of different cell lines after heat shock along with the differentiation of spheroid progenitors into neurons was observed. Fibroblasts and spheroid progenitors retained cell proliferation after cryopreservation. Dermal cyst-forming cells regained their normal structure after collapsing by osmotic pressure. The spheroid progenitors incubated in the adipogenic, osteogenic, and neurogenic media differentiated into adipocyte-, osteoblast-, and neuron-like cells, respectively. To the best of our knowledge, we isolated different unique cellular types and stem-like cells from the camel skin and examined their multipotency for the first time.
Collapse
Affiliation(s)
- Islam M Saadeldin
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Ayman Abdel-Aziz Swelum
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Ahmed E Noreldin
- Histology and Cytology Department, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| | - Hammed A Tukur
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Aaser M Abdelazim
- Department of Basic Medical Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia.
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Mosleh M Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Abdullah N Alowaimer
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
61
|
Mohammad K, Dakik P, Medkour Y, Mitrofanova D, Titorenko VI. Quiescence Entry, Maintenance, and Exit in Adult Stem Cells. Int J Mol Sci 2019; 20:ijms20092158. [PMID: 31052375 PMCID: PMC6539837 DOI: 10.3390/ijms20092158] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/24/2019] [Accepted: 04/28/2019] [Indexed: 12/13/2022] Open
Abstract
Cells of unicellular and multicellular eukaryotes can respond to certain environmental cues by arresting the cell cycle and entering a reversible state of quiescence. Quiescent cells do not divide, but can re-enter the cell cycle and resume proliferation if exposed to some signals from the environment. Quiescent cells in mammals and humans include adult stem cells. These cells exhibit improved stress resistance and enhanced survival ability. In response to certain extrinsic signals, adult stem cells can self-renew by dividing asymmetrically. Such asymmetric divisions not only allow the maintenance of a population of quiescent cells, but also yield daughter progenitor cells. A multistep process of the controlled proliferation of these progenitor cells leads to the formation of one or more types of fully differentiated cells. An age-related decline in the ability of adult stem cells to balance quiescence maintenance and regulated proliferation has been implicated in many aging-associated diseases. In this review, we describe many traits shared by different types of quiescent adult stem cells. We discuss how these traits contribute to the quiescence, self-renewal, and proliferation of adult stem cells. We examine the cell-intrinsic mechanisms that allow establishing and sustaining the characteristic traits of adult stem cells, thereby regulating quiescence entry, maintenance, and exit.
Collapse
Affiliation(s)
- Karamat Mohammad
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Paméla Dakik
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Younes Medkour
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Darya Mitrofanova
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Vladimir I Titorenko
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| |
Collapse
|
62
|
Deller RC, Richardson T, Richardson R, Bevan L, Zampetakis I, Scarpa F, Perriman AW. Artificial cell membrane binding thrombin constructs drive in situ fibrin hydrogel formation. Nat Commun 2019; 10:1887. [PMID: 31015421 PMCID: PMC6478844 DOI: 10.1038/s41467-019-09763-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
Cell membrane re-engineering is emerging as a powerful tool for the development of next generation cell therapies, as it allows the user to augment therapeutic cells to provide additional functionalities, such as homing, adhesion or hypoxia resistance. To date, however, there are few examples where the plasma membrane is re-engineered to display active enzymes that promote extracellular matrix protein assembly. Here, we report on a self-contained matrix-forming system where the membrane of human mesenchymal stem cells is modified to display a novel thrombin construct, giving rise to spontaneous fibrin hydrogel nucleation and growth at near human plasma concentrations of fibrinogen. The cell membrane modification process is realised through the synthesis of a membrane-binding supercationic thrombin-polymer surfactant complex. Significantly, the resulting robust cellular fibrin hydrogel constructs can be differentiated down osteogenic and adipogenic lineages, giving rise to self-supporting monoliths that exhibit Young's moduli that reflect their respective extracellular matrix compositions.
Collapse
Affiliation(s)
- Robert C Deller
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- School of Engineering, University of Liverpool, Liverpool, L69 3GH, UK
| | - Thomas Richardson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol, BS8 1FD, UK
| | - Rebecca Richardson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Laura Bevan
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Ioannis Zampetakis
- Bristol Composites Institute (ACCIS), University of Bristol, Bristol, BS8 1TR, UK
| | - Fabrizio Scarpa
- Bristol Composites Institute (ACCIS), University of Bristol, Bristol, BS8 1TR, UK
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
63
|
Panek M, Antunović M, Pribolšan L, Ivković A, Gotić M, Vukasović A, Caput Mihalić K, Pušić M, Jurkin T, Marijanović I. Bone Tissue Engineering in a Perfusion Bioreactor Using Dexamethasone-Loaded Peptide Hydrogel. MATERIALS 2019; 12:ma12060919. [PMID: 30893951 PMCID: PMC6470940 DOI: 10.3390/ma12060919] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/10/2019] [Accepted: 03/18/2019] [Indexed: 01/08/2023]
Abstract
The main goal of this study was the formation of bone tissue using dexamethasone (DEX)-loaded [COCH₃]-RADARADARADARADA-[CONH₂] (RADA 16-I) scaffold that has the ability to release optimal DEX concentration under perfusion force. Bone-marrow samples were collected from three patients during a hip arthroplasty. Human mesenchymal stem cells (hMSCs) were isolated and propagated in vitro in order to be seeded on scaffolds made of DEX-loaded RADA 16-I hydrogel in a perfusion bioreactor. DEX concentrations were as follows: 4 × 10-3, 4 × 10-4 and 4 × 10-5 M. After 21 days in a perfusion bioreactor, tissue was analyzed by scanning electron microscopy (SEM) and histology. Markers of osteogenic differentiation were quantified by real-time polymerase chain reaction (RT-PCR) and immunocytochemistry. Minerals were quantified and detected by the von Kossa method. In addition, DEX release from the scaffold in a perfusion bioreactor was assessed. The osteoblast differentiation was confirmed by the expression analysis of osteoblast-related genes (alkaline phosphatase (ALP), collagen I (COL1A1) and osteocalcin (OC). The hematoxylin/eosin staining confirmed the presence of cells and connective tissue, while SEM revealed morphological characteristics of cells, extracellular matrix and minerals-three main components of mature bone tissue. Immunocytochemical detection of collagen I is in concordance with given results, supporting the conclusion that scaffold with DEX concentration of 4 × 10-4 M has the optimal engineered tissue morphology. The best-engineered bone tissue is produced on scaffold loaded with 4 × 10-4 M DEX with a perfusion rate of 0.1 mL/min for 21 days. Differentiation of hMSCs on DEX-loaded RADA 16-I scaffold under perfusion force has a high potential for application in regenerative orthopedics.
Collapse
Affiliation(s)
- Marina Panek
- Department of Biology, Faculty of Science, University of Zagreb, 10 000 Zagreb, Croatia.
- Center for Translational and Clinical Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia.
| | - Maja Antunović
- Department of Biology, Faculty of Science, University of Zagreb, 10 000 Zagreb, Croatia.
| | - Lidija Pribolšan
- Department of Biology, Faculty of Science, University of Zagreb, 10 000 Zagreb, Croatia.
| | - Alan Ivković
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia.
- Department of Orthopaedic Surgery, University Hospital Sveti Duh, 10000 Zagreb, Croatia.
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia.
| | - Marijan Gotić
- Department of Material Chemistry, Rudjer Boskovic Institute, 10000 Zagreb, Croatia.
| | - Andreja Vukasović
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia.
| | - Katarina Caput Mihalić
- Department of Biology, Faculty of Science, University of Zagreb, 10 000 Zagreb, Croatia.
| | - Maja Pušić
- Department of Biology, Faculty of Science, University of Zagreb, 10 000 Zagreb, Croatia.
| | - Tanja Jurkin
- Department of Material Chemistry, Rudjer Boskovic Institute, 10000 Zagreb, Croatia.
| | - Inga Marijanović
- Department of Biology, Faculty of Science, University of Zagreb, 10 000 Zagreb, Croatia.
| |
Collapse
|
64
|
Antunovic M, Matic I, Nagy B, Caput Mihalic K, Skelin J, Stambuk J, Josipovic P, Dzinic T, Paradzik M, Marijanovic I. FADD-deficient mouse embryonic fibroblasts undergo RIPK1-dependent apoptosis and autophagy after NB-UVB irradiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 194:32-45. [PMID: 30904584 DOI: 10.1016/j.jphotobiol.2019.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/28/2019] [Accepted: 03/13/2019] [Indexed: 11/24/2022]
Abstract
Sun or therapy-related ultraviolet B (UVB) irradiation induces different cell death modalities such as apoptosis, necrosis/necroptosis and autophagy. Understanding of mechanisms implicated in regulation and execution of cell death program is imperative for prevention and treatment of skin diseases. An essential component of death-inducing complex is Fas-associated protein with death domain (FADD), involved in conduction of death signals of different death modalities. The purpose of this study was to enlighten the role of FADD in the selection of cell death mode after narrow-band UVB (NB-UVB) irradiation using specific cell death inhibitors (carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]- fluoromethylketone (zVAD-fmk), Necrostatin-1 and 3-Methyladenine) and FADD-deficient (FADD-/-) mouse embryonic fibroblasts (MEFs) and their wild type (wt) counterparts. The results imply that lack of FADD sensitized MEFs to induction of receptor-interacting protein 1 (RIPK1)-dependent apoptosis by the generation of reactive oxygen species (ROS), but without activation of the proteins p53, Bax and Bcl-2 as well as without the enrolment of calpain-2. Autophagy was established as a contributing factor to NB-UVB-induced death execution. By contrast, wt cells triggered intrinsic apoptotic pathway that was resistant to the inhibition by zVAD-fmk and Necrostatin-1 pointing to the mechanism overcoming the cell survival. These findings support the role of FADD in prevention of autophagy-dependent apoptosis.
Collapse
Affiliation(s)
- Maja Antunovic
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Igor Matic
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Biserka Nagy
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Katarina Caput Mihalic
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Josipa Skelin
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Jerko Stambuk
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Pavle Josipovic
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Tamara Dzinic
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Mladen Paradzik
- Department of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia
| | - Inga Marijanovic
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia.
| |
Collapse
|
65
|
Tong L, Liao Q, Zhao Y, Huang H, Gao A, Zhang W, Gao X, Wei W, Guan M, Chu PK, Wang H. Near-infrared light control of bone regeneration with biodegradable photothermal osteoimplant. Biomaterials 2019; 193:1-11. [DOI: 10.1016/j.biomaterials.2018.12.008] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/22/2018] [Accepted: 12/08/2018] [Indexed: 11/25/2022]
|
66
|
Quiescent Human Mesenchymal Stem Cells Are More Resistant to Heat Stress than Cycling Cells. Stem Cells Int 2018; 2018:3753547. [PMID: 30675168 PMCID: PMC6323451 DOI: 10.1155/2018/3753547] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/20/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
Quiescence is the prevailing state of many cell types under homeostatic conditions. Yet, surprisingly, little is known about how quiescent cells respond to environmental challenges. The aim of the present study is to compare stress responses of cycling and quiescent mesenchymal stem cells (MSC). Human endometrial mesenchymal cells (eMSС) were employed as adult stem cells. eMSC quiescence was modeled by serum starvation. Sublethal heat shock (HS) was used as a stress factor. Both quiescent and cycling cells were heated at 45°C for 30 min and then returned to standard culture conditions for their recovery. HS response was monitored by DNA damage response, stress-induced premature senescence (SIPS), cell proliferation activity, and oxidative metabolism. It has been found that quiescent cells repair DNA more rapidly, resume proliferation, and undergo SIPS less than proliferating cells. HS-enforced ROS production in heated cycling cells was accompanied with increased expression of genes regulating redox-active proteins. Quiescent cells exposed to HS did not intensify the ROS production, and genes involved in antioxidant defense were mostly silent. Altogether, the results have shown that quiescent cells are more resistant to heat stress than cycling cells. Next-generation sequencing (NGS) demonstrates that HS-survived cells retain differentiation capacity and do not exhibit signs of spontaneous transformation.
Collapse
|
67
|
Moise S, Byrne JM, El Haj AJ, Telling ND. The potential of magnetic hyperthermia for triggering the differentiation of cancer cells. NANOSCALE 2018; 10:20519-20525. [PMID: 30397703 DOI: 10.1039/c8nr05946b] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Magnetic hyperthermia is a potential technique for cancer therapy that exploits heat generated by magnetic nanoparticles to kill cancerous cells. Many studies have shown that magnetic hyperthermia is effective at killing cancer cells both in vitro and in vivo, however little attention has been paid to the cellular functioning of the surviving cells. We report here new evidence demonstrating the onset of thermally triggered differentiation in osteosarcoma cancer cells that survive magnetic hyperthermia treatment. This raises the possibility that in addition to causing cell death, magnetic hyperthermia could induce surviving cancer cells to form more mature cell types and thereby inhibit their capacity to self-renew. Such processes could prove to be as important as cell death when considering magnetic hyperthermia for treating cancer.
Collapse
Affiliation(s)
- Sandhya Moise
- Department of Chemical Engineering, University of Bath, Bath BA2 7AY, UK.
| | | | | | | |
Collapse
|
68
|
He B, Zhao J, Ou Y, Jiang D. Biofunctionalized peptide nanofiber-based composite scaffolds for bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 90:728-738. [PMID: 29853144 DOI: 10.1016/j.msec.2018.04.063] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 04/15/2018] [Accepted: 04/19/2018] [Indexed: 12/21/2022]
Abstract
Bone tissue had moderate self-healing capabilities, but biomaterial scaffolds were required for the repair of some defects such as large bone defects. Peptide nanofiber scaffolds demonstrated important potential in regenerative medicine. Functional modification and controlled release of signal molecules were two significant approaches to increase the bioactivity of biofunctionalized peptide nanofiber scaffolds, but peptide scaffolds were limited by insufficient mechanical strength. Thus, it was necessary to combine peptide scaffolds with other materials including polymers, hydroxyapatite, demineralized bone matrix (DBM) and metal materials based on the requirement of different bone defects. As the development of peptide-based composite scaffolds continued to evolve, ultimate translation to the clinical environment may allow for improved therapeutic outcomes for bone repair.
Collapse
Affiliation(s)
- Bin He
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jinqiu Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yunsheng Ou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Dianming Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
69
|
Effect of nanoheat stimulation mediated by magnetic nanocomposite hydrogel on the osteogenic differentiation of mesenchymal stem cells. SCIENCE CHINA-LIFE SCIENCES 2018; 61:448-456. [DOI: 10.1007/s11427-017-9287-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/05/2018] [Indexed: 12/20/2022]
|
70
|
Sun Z, Nair LS, Laurencin CT. The Paracrine Effect of Adipose-Derived Stem Cells Inhibits IL-1β-induced Inflammation in Chondrogenic Cells through the Wnt/β-Catenin Signaling Pathway. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018. [DOI: 10.1007/s40883-018-0047-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
71
|
Li C, Sunderic K, Nicoll SB, Wang S. Downregulation of Heat Shock Protein 70 Impairs Osteogenic and Chondrogenic Differentiation in Human Mesenchymal Stem Cells. Sci Rep 2018; 8:553. [PMID: 29323151 PMCID: PMC5765044 DOI: 10.1038/s41598-017-18541-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/12/2017] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) show promise for bone and cartilage regeneration. Our previous studies demonstrated that hMSCs with periodic mild heating had enhanced osteogenic and chondrogenic differentiation with significantly upregulated heat shock protein 70 (HSP70). However, the role of HSP70 in adult tissue regeneration is not well studied. Here, we revealed an essential regulatory mechanism of HSP70 in osteogenesis and chondrogenesis using adult hMSCs stably transfected with specific shRNAs to knockdown HSP70. Periodic heating at 39 °C was applied to hMSCs for up to 26 days. HSP70 knockdown resulted in significant reductions of alkaline phosphatase activity, calcium deposition, and gene expression of Runx2 and Osterix during osteogenesis. In addition, knockdown of HSP70 led to significant decreases of collagens II and X during chondrogenesis. Thus, downregulation of HSP70 impaired hMSC osteogenic and chondrogenic differentiation as well as the enhancement of these processes by thermal treatment. Taken together, these findings suggest a putative mechanism of thermal-enhanced bone and cartilage formation and underscore the importance of HSP70 in adult bone and cartilage differentiation.
Collapse
Affiliation(s)
- Chenghai Li
- Department of Biomedical Engineering, City University of New York-City College, 160 Convent Avenue, New York, NY, 10031, USA
| | - Kristifor Sunderic
- Department of Biomedical Engineering, City University of New York-City College, 160 Convent Avenue, New York, NY, 10031, USA
| | - Steven B Nicoll
- Department of Biomedical Engineering, City University of New York-City College, 160 Convent Avenue, New York, NY, 10031, USA
| | - Sihong Wang
- Department of Biomedical Engineering, City University of New York-City College, 160 Convent Avenue, New York, NY, 10031, USA.
| |
Collapse
|
72
|
Heat-Shock Protein 70 Overexpression in Adipose-Derived Stem Cells Enhances Fat Graft Survival. Ann Plast Surg 2017; 78:460-466. [PMID: 28106629 DOI: 10.1097/sap.0000000000000968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Autologous fat grafting is a prevalent technique used for soft-tissue augmentation; however, the poor survival rate of the grafted tissue remains a drawback of this method. Although adipose-derived stem cells (ASCs) are an attractive candidate for enhancing graft retention, the poor posttransplantation viability of these cells limits their application. Here we investigated whether overexpression of the antiapoptotic protein heat-shock protein 70 (Hsp70) could enhance ASCs' therapeutic potential for fat transplant survival. METHODS Recombinant adenoviral vectors were used to overexpress Hsp70 in ASCs isolated from a healthy woman. The Hsp70 expression was assessed by quantitative real-time polymerase chain reaction and Western blot analyses. The adipose tissue granules aspirated from another woman were mixed with ASCs expressing green fluorescent protein (GFP)-tagged Hsp70 (group A) or GFP alone (group B), untreated ASCs (group C), and phosphate-buffered saline (group D). Fat mixtures were then injected subcutaneously into the backs of nude mice, and graft survival was compared after 3 months. RESULTS Adipose-derived stem cells transduced with recombinant adenoviral vectors exhibited significantly increased Hsp70 expression in vitro. Meanwhile, weight retention analyses demonstrated that fat grafts using the group A cell population exhibited significantly higher survival rates than the other treatment groups in vivo. Moreover, histological analyses revealed that fat grafts containing GFP-Hsp70-expressing ASCs yielded significantly lower levels of tissue fibrosis and fat cysts/vacuoles, higher capillary densities, and increased numbers of viable adipocytes than the control groups. CONCLUSIONS Our data indicate that Hsp70 overexpression enhances the efficacy of ASC therapy by improving the survival and quality of the transplanted fat tissues.
Collapse
|
73
|
Ota T, Nishida Y, Ikuta K, Kato R, Kozawa E, Hamada S, Sakai T, Ishiguro N. Heat-stimuli-enhanced osteogenesis using clinically available biomaterials. PLoS One 2017; 12:e0181404. [PMID: 28719650 PMCID: PMC5515435 DOI: 10.1371/journal.pone.0181404] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/02/2017] [Indexed: 11/18/2022] Open
Abstract
A recent study reported that heat stress stimulates osteogenesis in an in vivo rat model using alginate gel and magnetite cationic liposomes. However, for clinical use, the efficacy for promoting osteogenesis needs to be investigated using clinically approved materials, and preferably with animals larger than rats. The aim of this study was to evaluate multiple heat stimuli-triggered osteogenesis in rat tibial defect models using already clinically applicable materials (Resovist® and REGENOS®) and determine the efficacy also in the rabbit. Fifty-eight rats and 10 rabbits were divided into two groups, respectively, with or without hyperthermia treatment at 45°C for 15 min. (hyperthermia; 20 rats once a week, 8 rats three times a week, 5 rabbits once a week, control; 30 rats and 5 rabbits). Micro-CT assessment at 4 weeks revealed that a significantly stimulated osteogenesis was observed in the once a week group of both rats and rabbits as compared to the control group (p = 0.018 and 0.036, respectively). In contrast, the three times a week group did not show enhanced osteogenesis. Histological examination and image analysis showed consistent results in which the area of mineralized bone formation in the once a week hyperthermia group was significantly increased compared with that in the control group at four weeks (rat; p = 0.026, rabbit; p = 0.031). Newly formed bone was observed in the grafted materials from the periphery toward the center, and more osteoclasts were found in the once a week group. Heat stress also induced enhanced alkaline phosphatase expression in cultured osteoblastic cells, MC3T3, in vitro (p = 0.03). On the other hand, heat stress had no obvious effects on chondrogenic differentiation using ATDC5 cells. Our study demonstrates that heat-stimuli with clinically applicable novel heating materials can promote significant osteogenesis, and may thus be a promising treatment option for diseases associated with bone defects.
Collapse
Affiliation(s)
- Takehiro Ota
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Yoshihiro Nishida
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
- * E-mail:
| | - Kunihiro Ikuta
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Ryuji Kato
- Department of Basic Medical Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Eiji Kozawa
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Shunsuke Hamada
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Tomohisa Sakai
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| | - Naoki Ishiguro
- Department of Orthopaedic Surgery, Nagoya University Graduate School and School of Medicine, Nagoya, Japan
| |
Collapse
|
74
|
Carreras MP, Wang S. A multifunctional microfluidic platform for generation, trapping and release of droplets in a double laminar flow. J Biotechnol 2017; 251:106-111. [PMID: 28450257 DOI: 10.1016/j.jbiotec.2017.04.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 04/17/2017] [Accepted: 04/24/2017] [Indexed: 10/19/2022]
Abstract
Droplet microfluidics, involving micrometer-sized emulsion of droplets is a growing subfield of microfluidics which attracts broad interest due to its application on biological assays. Droplet-based systems have been used as microreactors as well as to encapsulate many biological entities for biomedical and biotechnological applications. Here, a novel microfluidic device is presented for the generation, trapping and release of aqueous including hydrogel droplets in a double laminar oil flow. This platform enables the storage and release of picoliter-sized droplets in two different carrier oils by using hydrodynamic forces without the need of electrical forces or optical actuators. Furthermore, this design allows droplets to be selectively and simultaneously exposed to two different conditions and collected on demand. Successful encapsulation of hepatoma H35 cells was performed on-chip. Viability of cell-laden droplets was performed off-chip to assess the potential applications in 3D encapsulation cell culture and drug discovery assays.
Collapse
Affiliation(s)
- Maria Pilar Carreras
- Department of Biomedical Engineering, City University of New York - City College, New York, NY 10031, USA
| | - Sihong Wang
- Department of Biomedical Engineering, City University of New York - City College, New York, NY 10031, USA.
| |
Collapse
|
75
|
Shaik S, Hayes D, Gimble J, Devireddy R. Inducing Heat Shock Proteins Enhances the Stemness of Frozen-Thawed Adipose Tissue-Derived Stem Cells. Stem Cells Dev 2017; 26:608-616. [PMID: 28052723 PMCID: PMC5393415 DOI: 10.1089/scd.2016.0289] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/04/2017] [Indexed: 01/01/2023] Open
Abstract
Extensive research has been performed to determine the effect of freezing protocol and cryopreservation agents on the viability of adipose tissue-derived stromal/stem cells (ASCs) as well as other cells. Unfortunately, the conclusion one may draw after decades of research utilizing fundamentally similar cryopreservation techniques is that a barrier exists, which precludes full recovery. We hypothesize that agents capable of inducing a subset of heat shock proteins (HSPs) and chaperones will reduce the intrinsic barriers to the post-thaw recovery of ASCs. ASCs were exposed to 43°C for 1 h to upregulate HSPs, and the temporal HSP expression profile postheat shock was determined by performing quantitative polymerase chain reaction (PCR) and western blotting assays. The expression levels of HSP70 and HSP32 were found to be maximum at 3 h after the heat shock, whereas HSP90 and HSP27 remain unchanged. The heat shocked ASCs cryopreserved during maximal HSPs expression exhibited increased post-thaw viability than the nonheat shocked samples. Histochemical staining and quantitative reverse transcription-PCR indicated that the ASC differentiation potential was retained. Thus, suggesting that the upregulation of HSPs before a freezing insult is beneficial to ASCs and a potential alternative to the use of harmful cryoprotective agents.
Collapse
Affiliation(s)
- Shahensha Shaik
- Bioengineering Laboratory, Department of Mechanical Engineering, Louisiana State University, Baton Rouge, Louisiana
| | - Daniel Hayes
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania
| | - Jeffrey Gimble
- La Cell LLC and Center for Stem Cell Research and Regenerative Medicine and Departments of Medicine, Structural and Cellular Biology, and Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ram Devireddy
- Bioengineering Laboratory, Department of Mechanical Engineering, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
76
|
He B, Ou Y, Chen S, Zhao W, Zhou A, Zhao J, Li H, Jiang D, Zhu Y. Designer bFGF-incorporated d-form self-assembly peptide nanofiber scaffolds to promote bone repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 74:451-458. [PMID: 28254316 DOI: 10.1016/j.msec.2016.12.042] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 11/21/2016] [Accepted: 12/09/2016] [Indexed: 10/20/2022]
Abstract
d-Form and l-form peptide nanofiber scaffolds can spontaneously form stable β-sheet secondary structures and nanofiber hydrogel scaffolds, and hold some promise in hemostasis and wound healing. We report here on the synthetic self-assembling peptide d-RADA16 and l-RADA16 are both found to produce stable β-sheet secondary structure and nanofiber hydrogel scaffolds based on circular dichroism (CD) spectroscopy, transmission electron microscopy (TEM) and rheology analysis etc. d-RADA16 hydrogel and l-RADA16 hydrogel can enhance obvious bone repair in femoral condyle defects of the Sprague-Dawley (SD) rat model compared to PBS treatment. Based on micro-computed tomography (CT), it was revealed that d-RADA16 hydrogel and l-RADA16 hydrogel were capable to obtain the extensive bone healing. Histological evaluation also found that these two hydrogels facilitate the presence of more mature bone tissue within the femoral condyle defects. Additionally, d-RADA16 hydrogel showed some potential in storing and releasing basic-fibroblast growth factor (bFGF) which was able to further promote bone regeneration based on micro-CT analysis. These results indicate that d-form peptide nanofiber hydrogel have some special capacity for bone repair.
Collapse
Affiliation(s)
- Bin He
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Yunsheng Ou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shuo Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Weikang Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jinqiu Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hong Li
- School of Physical Science and Technology, Sichuan University, Chengdu 610000, China
| | - Dianming Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
77
|
Meka SRK, Jain S, Chatterjee K. Strontium eluting nanofibers augment stem cell osteogenesis for bone tissue regeneration. Colloids Surf B Biointerfaces 2016; 146:649-56. [DOI: 10.1016/j.colsurfb.2016.07.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/13/2016] [Accepted: 07/04/2016] [Indexed: 01/13/2023]
|
78
|
Castillo Diaz LA, Elsawy M, Saiani A, Gough JE, Miller AF. Osteogenic differentiation of human mesenchymal stem cells promotes mineralization within a biodegradable peptide hydrogel. J Tissue Eng 2016; 7:2041731416649789. [PMID: 27493714 PMCID: PMC4959308 DOI: 10.1177/2041731416649789] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/20/2016] [Indexed: 01/07/2023] Open
Abstract
An attractive strategy for the regeneration of tissues has been the use of extracellular matrix analogous biomaterials. Peptide-based fibrillar hydrogels have been shown to mimic the structure of extracellular matrix offering cells a niche to undertake their physiological functions. In this study, the capability of an ionic-complementary peptide FEFEFKFK (F, E, and K are phenylalanine, glutamic acid, and lysine, respectively) hydrogel to host human mesenchymal stem cells in three dimensions and induce their osteogenic differentiation is demonstrated. Assays showed sustained cell viability and proliferation throughout the hydrogel over 12 days of culture and these human mesenchymal stem cells differentiated into osteoblasts simply upon addition of osteogenic stimulation. Differentiated osteoblasts synthesized key bone proteins, including collagen-1 (Col-1), osteocalcin, and alkaline phosphatase. Moreover, mineralization occurred within the hydrogel. The peptide hydrogel is a naturally biodegradable material as shown by oscillatory rheology and reversed-phase high-performance liquid chromatography, where both viscoelastic properties and the degradation of the hydrogel were monitored over time, respectively. These findings demonstrate that a biodegradable octapeptide hydrogel can host and induce the differentiation of stem cells and has the potential for the regeneration of hard tissues such as alveolar bone.
Collapse
Affiliation(s)
- Luis A Castillo Diaz
- School of Chemical Engineering and Analytical Science, The University of Manchester, Manchester, UK; Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Mohamed Elsawy
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Alberto Saiani
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK; The School of Materials, The University of Manchester, Manchester, UK
| | - Julie E Gough
- The School of Materials, The University of Manchester, Manchester, UK
| | - Aline F Miller
- School of Chemical Engineering and Analytical Science, The University of Manchester, Manchester, UK; Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| |
Collapse
|
79
|
3D patterned stem cell differentiation using thermo-responsive methylcellulose hydrogel molds. Sci Rep 2016; 6:29408. [PMID: 27381562 PMCID: PMC4933913 DOI: 10.1038/srep29408] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/16/2016] [Indexed: 12/20/2022] Open
Abstract
Tissue-specific patterned stem cell differentiation serves as the basis for the development, remodeling, and regeneration of the multicellular structure of the native tissues. We herein proposed a cytocompatible 3D casting process to recapitulate this patterned stem cell differentiation for reconstructing multicellular tissues in vitro. We first reconstituted the 2D culture conditions for stem cell fate control within 3D hydrogel by incorporating the sets of the diffusible signal molecules delivered through drug-releasing microparticles. Then, utilizing thermo-responsivity of methylcellulose (MC), we developed a cytocompatible casting process to mold these hydrogels into specific 3D configurations, generating the targeted spatial gradients of diffusible signal molecules. The liquid phase of the MC solution was viscous enough to adopt the shapes of 3D impression patterns, while the gelated MC served as a reliable mold for patterning the hydrogel prepolymers. When these patterned hydrogels were integrated together, the stem cells in each hydrogel distinctly differentiated toward individually defined fates, resulting in the formation of the multicellular tissue structure bearing the very structural integrity and characteristics as seen in vascularized bones and osteochondral tissues.
Collapse
|
80
|
Sou K, Chan LY, Lee CLK. Temperature Tracking in a Three-Dimensional Matrix Using Thermosensitive Liposome Platform. ACS Sens 2016. [DOI: 10.1021/acssensors.6b00222] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Keitaro Sou
- Waseda Bioscience Research Institute in Singapore (WABIOS), 11 Biopolis Way, #05-02 Helios, Singapore 138667, Singapore
- Organization
for University Research Initiatives, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Li Yan Chan
- Centre
for Biomedical and Life Sciences, Department for Technology, Innovation
and Enterprise (TIE), Singapore Polytechnic, 500 Dover Road, Singapore 139651, Singapore
| | - Chi-Lik Ken Lee
- Centre
for Biomedical and Life Sciences, Department for Technology, Innovation
and Enterprise (TIE), Singapore Polytechnic, 500 Dover Road, Singapore 139651, Singapore
| |
Collapse
|
81
|
Identification of Differential Genes Expression Profiles and Pathways of Bone Marrow Mesenchymal Stem Cells of Adolescent Idiopathic Scoliosis Patients by Microarray and Integrated Gene Network Analysis. Spine (Phila Pa 1976) 2016; 41:840-55. [PMID: 26679893 DOI: 10.1097/brs.0000000000001394] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Microarray approach and integrated gene network analysis. OBJECTIVE To explore the differential genetic expression profile, gene ontology terms, and Kyoto Encyclopedia of Genes and Genomes pathways in bone marrow mesenchymal stem cells (BM-MSCs) of idiopathic scoliosis (AIS) and non-AIS controls. SUMMARY OF BACKGROUND DATA The pathogenesis of adolescent AIS and the accompanying generalized osteopenia remain unclear. Our previous study suggested increased proliferation ability and decreased osteogenic differentiation ability of BM-MSCs of AIS. Therefore, we hypothesized that MSCs may play a significant role in the etiology and pathogenesis of AIS. METHODS In this study, microarray analysis was used to identify differentially expressed genes (DEGs) of BM-MSCs from AIS patients compared with those from healthy individuals. Comprehensive bioinformatics analyses were then used to enrich datasets for gene ontology and pathway. Based on the gene signal transduction network analysis of DEGs contained in significant pathways, 24 potential crucial genes were selected for validation by reverse transcription polymerase chain reaction. RESULTS There are 1027 previously unrecognized DEGs in BM-MSCs from AIS patients. Pathway analysis revealed dysregulated mitogen-activated protein kinase (MAPK) signaling pathway, PI3K-Akt signaling pathway, calcium signaling pathway, peroxisome proliferator-activated receptor (PPAR) signaling pathway, ubiquitin-mediated proteolysis, and Notch signaling pathway, all of which have been reported to play an important role in regulating the osteogenic or adipogenic differentiation of MSCs. Furthermore, gene signal transduction networks analysis indicated that mitogen-activated protein kinase kinase 1 (MAP2K1), SMAD family member 3 (SMAD3), homeobox C6 (HOXC6), heat shock 70kDa protein 6 (HSPA6), general transcription factor IIi (GTF2I), CREB binding protein (CREBBP), phosphoinositide-3-kinase, regulatory subunit 2 (PIK3R2), and dual specificity phosphatase 2 (DUSP2) may play essential roles in AIS pathogenesis and accompanied osteopenia. CONCLUSION This study reports the differential genes expression profiles of BM-MSCs from AIS patients and related potential pathways for the first time. These previously unrecognized genes and molecular pathways might play a significant role in not only the causal mechanism of osteopenia in AIS, but also the AIS initiation and development. The identification of these candidate genes provides novel insight into the underlying etiological mechanisms of AIS. LEVEL OF EVIDENCE N/A.
Collapse
|
82
|
He B, Ou Y, Zhou A, Chen S, Zhao W, Zhao J, Li H, Zhu Y, Zhao Z, Jiang D. Functionalized d-form self-assembling peptide hydrogels for bone regeneration. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1379-88. [PMID: 27114701 PMCID: PMC4833366 DOI: 10.2147/dddt.s97530] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bone defects are very common in orthopedics, and there is great need to develop suitable bone grafts for transplantation in vivo. However, current bone grafts still encounter some limitations, including limited availability, immune rejection, poor osteoinduction and osteoconduction, poor biocompatibility and degradation properties, etc. Self-assembling peptide nanofiber scaffolds have emerged as an important substrate for cell culture and bone regeneration. We report on the structural features (eg, Congo red staining, circular dichroism spectroscopy, transmission electron microscopy, and rheometry assays) and osteogenic ability of d-RADA16-RGD peptide hydrogels (with or without basic fibroblast growth factor) due to the better stability of peptide bonds formed by these peptides compared with those formed by l-form peptides, and use them to fill the femoral condyle defect of Sprague Dawley rat model. The bone morphology change, two-dimensional reconstructions using microcomputed tomography, quantification of the microcomputed tomography analyses as well as histological analyses have demonstrated that RGD-modified d-form peptide scaffolds are able to enhance extensive bone regeneration.
Collapse
Affiliation(s)
- Bin He
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yunsheng Ou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Shuo Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Weikang Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jinqiu Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Hong Li
- School of Physical Science and Technology, Sichuan University, Chengdu, People's Republic of China
| | - Yong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zenghui Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dianming Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
83
|
Tatman PD, Muhonen EG, Wickers ST, Gee AO, Kim ES, Kim DH. Self-assembling peptides for stem cell and tissue engineering. Biomater Sci 2016; 4:543-54. [PMID: 26878078 PMCID: PMC4803621 DOI: 10.1039/c5bm00550g] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Regenerative medicine holds great potential to address many shortcomings in current medical therapies. An emerging avenue of regenerative medicine is the use of self-assembling peptides (SAP) in conjunction with stem cells to improve the repair of damaged tissues. The specific peptide sequence, mechanical properties, and nanotopographical cues vary widely between different SAPs, many of which have been used for the regeneration of similar tissues. To evaluate the potential of SAPs to guide stem cell fate, we extensively reviewed the literature for reports of SAPs and stem cell differentiation. To portray the most accurate summary of these studies, we deliberately discuss both the successes and pitfalls, allowing us to make conclusions that span the breadth of this exciting field. We also expand on these conclusions by relating these findings to the fields of nanotopography, mechanotransduction, and the native composition of the extracellular matrix in specific tissues to identify potential directions for future research.
Collapse
Affiliation(s)
- Philip D Tatman
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Colorado, Aurora, Colorado, USA
| | - Ethan G Muhonen
- School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Sean T. Wickers
- Department of Chemistry, University of Colorado, Denver, Colorado, USA
| | - Albert O. Gee
- Department of Orthopedics and Sports Medicine, University of Washington, Seattle, WA 98195, USA
| | - Eung-Sam Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Biological Sciences, Chonnam National University, Gwangju, Korea
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
84
|
Chen E, Xue D, Zhang W, Lin F, Pan Z. Extracellular heat shock protein 70 promotes osteogenesis of human mesenchymal stem cells through activation of the ERK signaling pathway. FEBS Lett 2015; 589:4088-96. [PMID: 26608032 DOI: 10.1016/j.febslet.2015.11.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/22/2015] [Accepted: 11/13/2015] [Indexed: 12/30/2022]
Abstract
Heat shock proteins have protective effects when cells are exposed to stress. However, the relationship between extracellular heat shock protein 70 (eHSP70) and osteogenesis of hMSCs has not been reported. The results of this study showed that HSP70 (200 ng/ml) increases alkaline phosphatase activity and promotes hMSC mineralization. Under osteogenic induction conditions, HSP70 significantly upregulated the expression of osteo-specific genes, such as the runt family transcription factor Runx2 and osterix (OSX). Comparative expression profiling by microarray and pathway analyses revealed that HSP70 promotes osteogenesis of hMSCs through activation of the ERK signaling pathway. HSP70 may be a potential therapeutic agent for the treatment of bone nonunion.
Collapse
Affiliation(s)
- Erman Chen
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, 310000 Hangzhou, People's Republic of China
| | - Deting Xue
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, 310000 Hangzhou, People's Republic of China
| | - Wei Zhang
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, 310000 Hangzhou, People's Republic of China
| | - Feng Lin
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, 310000 Hangzhou, People's Republic of China
| | - Zhijun Pan
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, 310000 Hangzhou, People's Republic of China.
| |
Collapse
|
85
|
Li G, Liu X, Du Q, Gao M, An J. Three dimensional de novo micro bone marrow and its versatile application in drug screening and regenerative medicine. Exp Biol Med (Maywood) 2015; 240:1029-38. [PMID: 26283705 DOI: 10.1177/1535370215594583] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The finding that bone marrow hosts several types of multipotent stem cell has prompted extensive research aimed at regenerating organs and building models to elucidate the mechanisms of diseases. Conventional research depends on the use of two-dimensional (2D) bone marrow systems, which imposes several obstacles. The development of 3D bone marrow systems with appropriate molecules and materials however, is now showing promising results. In this review, we discuss the advantages of 3D bone marrow systems over 2D systems and then point out various factors that can enhance the 3D systems. The intensive research on 3D bone marrow systems has revealed multiple important clinical applications including disease modeling, drug screening, regenerative medicine, etc. We also discuss some possible future directions in the 3D bone marrow research field.
Collapse
Affiliation(s)
- Guanqun Li
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13202, USA
| | - Xujun Liu
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13202, USA
| | - Qian Du
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13202, USA
| | - Mei Gao
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13202, USA SUNY Upstate Cancer Research Institute, State University of New York, Upstate Medical University, Syracuse, NY 13202, USA
| | - Jing An
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13202, USA SUNY Upstate Cancer Research Institute, State University of New York, Upstate Medical University, Syracuse, NY 13202, USA
| |
Collapse
|
86
|
Wang W, Kratz K, Behl M, Yan W, Liu Y, Xu X, Baudis S, Li Z, Kurtz A, Lendlein A, Ma N. The interaction of adipose-derived human mesenchymal stem cells and polyether ether ketone. Clin Hemorheol Microcirc 2015; 61:301-21. [DOI: 10.3233/ch-152001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Helmholtz Virtual Institute - Multifunctional Materials in Medicine, Berlin and Teltow, Teltow, Germany
| | - Marc Behl
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Wan Yan
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Yue Liu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Stefan Baudis
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Zhengdong Li
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Andreas Kurtz
- Berlin-Brandenburg Center for Regenerative Therapies, Charité - University Medicine Berlin, Berlin, Germany
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Gwangk-ro 1, Gwanak-gu, Seoul, Korea
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Helmholtz Virtual Institute - Multifunctional Materials in Medicine, Berlin and Teltow, Teltow, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Helmholtz Virtual Institute - Multifunctional Materials in Medicine, Berlin and Teltow, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
87
|
Piñeiro Y, Vargas Z, Rivas J, López-Quintela MA. Iron Oxide Based Nanoparticles for Magnetic Hyperthermia Strategies in Biological Applications. Eur J Inorg Chem 2015. [DOI: 10.1002/ejic.201500598] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
88
|
Li Z, Hou T, Deng M, Luo F, Wu X, Xing J, Chang Z, Xu J. The Osteogenetic Efficacy of Goat Bone Marrow-Enriched Self-Assembly Peptide/Demineralized Bone Matrix In Vitro and In Vivo. Tissue Eng Part A 2015; 21:1398-408. [PMID: 25518911 DOI: 10.1089/ten.tea.2014.0294] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Zhiqiang Li
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China
- Tissue Engineering Laboratory of Chongqing City, Chongqing, China
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, China
| | - Tianyong Hou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China
- Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Moyuan Deng
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China
- Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Fei Luo
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China
- Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Xuehui Wu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China
- Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Junchao Xing
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China
- Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Zhengqi Chang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China
- Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Jianzhong Xu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China
- Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| |
Collapse
|
89
|
Sun ZB, Wang JW, Xiao H, Zhang QS, Kan WS, Mo FB, Hu S, Ye SN. Icariin may benefit the mesenchymal stem cells of patients with steroid-associated osteonecrosis by ABCB1-promoter demethylation: a preliminary study. Osteoporos Int 2015; 26:187-97. [PMID: 25112719 DOI: 10.1007/s00198-014-2809-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 07/02/2014] [Indexed: 12/25/2022]
Abstract
UNLABELLED In this study, we found out a previously undefined function of icariin which restored the dynamic balance between osteogenic and adipogenic differentiation of mesenchymal stem cells (MSCs) in patients with osteonecrosis of femoral head (ONFH) via ABCB1-promoter demethylation. These findings provided important information regarding potential implication of icariin targeting epigenetic changes for the treatment of steroid -associated ONFH. INTRODUCTION Here, we investigated whether icariin can also exert a beneficial role in the reactivation of MSCs in the patients with steroid-associated ONFH via ABCB1-promoter demethylation. METHODS Bone marrow was collected from the proximal femur in patients with steroid-associated ONFH (n = 20) and patients with new femoral neck fractures (n = 22), and then MSCs were isolated. We investigated cell viability, intracellular reactive oxygen species (ROS) level, mitochondrial membrane potential (MMP), P-glycoprotein (P-gp) activity, the transcript levels of ABCB1 and oxidative stress-related genes, methylation extent at CpG islands of ABCB1 promoter, and osteogenic and adipogenic differentiation ability of MSCs from the femoral neck fractures group and from the steroid-associated ONFH group treated with or without icariin. RESULTS We observed that MSCs from the steroid-associated ONFH group showed reduced proliferation ability, elevated ROS level, depressed MMP, weakened osteogenesis, and enhanced adipogenesis while low P-gp activity, transcription level of ABCB1, and oxidative stress-related genes as well as aberrant CpG islands hypermethylation of ABCB1 were also noted in steroid-associated ONFH group. Treatment with icariin obviously induced de novo P-gp expression, decreased oxidative stress, and promoted osteogenesis. CONCLUSION Icariin may be a potential drug targeting epigenetic changes for the treatment of steroid-associated ONFH.
Collapse
Affiliation(s)
- Z-B Sun
- Department of Orthopedics, Pu-Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Ikuta K, Urakawa H, Kozawa E, Hamada S, Ota T, Kato R, Honda H, Kobayashi T, Ishiguro N, Nishida Y. In vivoheat-stimulus-triggered osteogenesis. Int J Hyperthermia 2014; 31:58-66. [DOI: 10.3109/02656736.2014.988662] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
91
|
Patil S, Paul S. A comprehensive review on the role of various materials in the osteogenic differentiation of mesenchymal stem cells with a special focus on the association of heat shock proteins and nanoparticles. Cells Tissues Organs 2014; 199:81-102. [PMID: 25401759 DOI: 10.1159/000362226] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2014] [Indexed: 11/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have important roles in the area of regenerative medicine and clinical applications due to their pluripotent nature. Osteogenic differentiation of MSCs has been studied extensively using various stimulants to develop models of bone repair. There are several factors that enhance the differentiation of MSCs into bone tissues. This review focuses on the effects of various inducers on the osteoblast differentiation of MSCs at different stages of cellular development. We discuss the various growth factors, hormones, vitamins, cytokines, chemical stimulants, and mechanical forces applied in bioreactors that play an essential role in the proliferation, differentiation, and matrix mineralization of stem cells during osteogenesis. Various nanoparticles have also been used recently for the same purpose and the results are promising. Moreover, we review the role of various stresses, including thermal stress, and the subsequent involvement of heat shock proteins as inducers of the proliferation and differentiation of osteoblasts. We also report how various proteasome inhibitors have been shown to induce proliferation and osteogenic differentiation of MSCs in a number of cases. In this communication, the role of peptide-based scaffolds in osteoblast proliferation and differentiation is also reviewed. Based on the reviewed information, this article proposes novel possibilities for the enhancement of proliferation, differentiation, and migration of osteoblasts from MSCs. © 2014 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Supriya Patil
- Structural Biology and Nanomedicine Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela, India
| | | |
Collapse
|
92
|
Gupta MS, Nicoll SB. Duration of TGF-β3 Exposure Impacts the Chondrogenic Maturation of Human MSCs in Photocrosslinked Carboxymethylcellulose Hydrogels. Ann Biomed Eng 2014; 43:1145-57. [DOI: 10.1007/s10439-014-1179-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/29/2014] [Indexed: 12/11/2022]
|
93
|
Yanagi T, Kajiya H, Kawaguchi M, Kido H, Fukushima T. Photothermal stress triggered by near infrared-irradiated carbon nanotubes promotes bone deposition in rat calvarial defects. J Biomater Appl 2014; 29:1109-18. [DOI: 10.1177/0885328214556913] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The bone regenerative healing process is often prolonged, with a high risk of infection particularly in elderly and diseased patients. A reduction in healing process time usually requires mechanical stress devices, chemical cues, or laser/thermal therapies. Although these approaches have been used extensively for the reduction of bone healing time, the exact mechanisms involved in thermal stress-induced bone regeneration remain unclear. In this study, we investigated the effect of optimal hyperthermia on rat calvarial defects in vivo and on osteogenesis in vitro. Photothermal stress stimulation was carried out using a new photothermal device, composed of an alginate gel including in carbon nanotubes and their irradiator with near-infrared light. Photothermal stress (15 min at 42℃, every day), trigged by near-infrared-induced carbon nanotube, promoted bone deposition in critical-sized calvarial defects compared with nonthermal stress controls. We recently reported that our novel DNA/protamine complex scaffold induces bone regeneration in calvarial defects. In this study, photothermal stress upregulated bone deposition in DNA/protamine-engrafted calvarial defects. Furthermore, photothermal stress significantly induced expression of osteogenic related genes in a time-dependent manner, including alkaline phosphatase, osterix, and osteocalcin. This was observed in DNA/protamine cells, which were expanded from regenerated tissue engrafted into the DNA/protamine scaffold, as well as in human MG63 preosteoblasts. In summary, this novel carbon nanotube-based photothermal stress approach upregulated expression of osteogenic-related genes in preosteoblasts, resulting in promotion of mineral deposition for enhanced bone repair.
Collapse
Affiliation(s)
- Tsukasa Yanagi
- Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Hiroshi Kajiya
- Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Minoru Kawaguchi
- Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan
- Department of Dental Engineering, Fukuoka Dental College, Fukuoka, Japan
| | - Hirofumi Kido
- Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
| | - Tadao Fukushima
- Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
94
|
Gupta MS, Nicoll SB. Functional nucleus pulposus-like matrix assembly by human mesenchymal stromal cells is directed by macromer concentration in photocrosslinked carboxymethylcellulose hydrogels. Cell Tissue Res 2014; 358:527-39. [PMID: 25092545 DOI: 10.1007/s00441-014-1962-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/08/2014] [Indexed: 02/07/2023]
Abstract
Intervertebral disc (IVD) degeneration is associated with several pathophysiologic changes of the IVD, including dehydration of the nucleus pulposus (NP). Tissue engineering strategies may be used to restore both biological and mechanical function of the IVD following removal of NP tissue during surgical intervention. Recently, photocrosslinked carboxymethylcellulose (CMC) hydrogels were shown to support chondrogenic, NP-like extracellular matrix (ECM) elaboration by human mesenchymal stromal cells (hMSCs) when supplemented with TGF-β3; however, mechanical properties of these constructs did not reach native values. Fabrication parameters (i.e., composition, crosslinking density) can influence the bulk mechanical properties of hydrogel scaffolds, as well as cellular behavior and differentiation patterns. The objective of this study was to evaluate the influence of CMC macromer concentration (1.5, 2.5 and 3.5 % weight/volume) on bulk hydrogel properties and NP-like matrix elaboration by hMSCs. The lowest macromer concentration of 1.5 % exhibited the highest gene expression levels of aggrecan and collagen II at day 7, corresponding with the largest accumulation of glycosaminoglycans and collagen II by day 42. The ECM elaboration in the 1.5 % constructs was more homogeneously distributed compared to primarily pericellular localization in 3.5 % gels. The 1.5 % gels also displayed significant improvements in mechanical functionality by day 42 compared to earlier time points, which was not seen in the other groups. The effects of macromer concentration on matrix accumulation and organization are likely attributed to quantifiable differences in polymer crosslinking density and diffusive properties between the various hydrogel formulations. Taken together, these results demonstrate that macromer concentration of CMC hydrogels can direct hMSC matrix elaboration, such that a lower polymer concentration allows for greater NP-like ECM assembly and improvement of mechanical properties over time.
Collapse
Affiliation(s)
- Michelle S Gupta
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | | |
Collapse
|
95
|
Bone marrow enriched graft, modified by self-assembly peptide, repairs critically-sized femur defects in goats. INTERNATIONAL ORTHOPAEDICS 2014; 38:2391-8. [DOI: 10.1007/s00264-014-2388-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 05/17/2014] [Indexed: 12/12/2022]
|
96
|
Kim JE, Lee SM, Kim SH, Tatman P, Gee AO, Kim DH, Lee KE, Jung Y, Kim SJ. Effect of self-assembled peptide-mesenchymal stem cell complex on the progression of osteoarthritis in a rat model. Int J Nanomedicine 2014; 9 Suppl 1:141-57. [PMID: 24872709 PMCID: PMC4024982 DOI: 10.2147/ijn.s54114] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To evaluate the efficacy of mesenchymal stem cells (MSCs) encapsulated in self-assembled peptide (SAP) hydrogels in a rat knee model for the prevention of osteoarthritis (OA) progression. MATERIALS AND METHODS Nanostructured KLD-12 SAPs were used as the injectable hydrogels. Thirty-three Sprague Dawley rats were used for the OA model. Ten rats were used for the evaluation of biotin-tagged SAP disappearance. Twenty-three rats were divided into four groups: MSC (n=6), SAP (n=6), SAP-MSC (n=6), and no treatment (n=5). MSCs, SAPs, and SAP-MSCs were injected into the knee joints 3 weeks postsurgery. Histologic examination, immunofluorescent staining, measurement of cytokine levels, and micro-computed tomography analysis were conducted 6 weeks after injections. Behavioral studies were done to establish baseline measurements before treatment, and repeated 3 and 6 weeks after treatment to measure the efficacy of SAP-MSCs. RESULTS Concentration of biotinylated SAP at week 1 was not significantly different from those at week 3 and week 6 (P=0.565). Bone mineral density was significantly lower in SAP-MSC groups than controls (P=0.002). Significant differences in terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling staining between the control group and all other groups were observed. Caspase-8, tissue inhibitor of metalloproteinases 1, and matrix metalloproteinase 9 were diffusely stained in controls, whereas localized or minimal staining was observed in other groups. Modified Mankin scores were significantly lower in the SAP and SAP-MSC groups than in controls (P=0.001 and 0.013). Although not statistically significant, synovial inflammation scores were lower in the SAP (1.3±0.3) and SAP-MSC (1.3±0.2) groups than in controls (2.6±0.2). However, neither the cytokine level nor the behavioral score was significantly different between groups. CONCLUSION Injection of SAP-MSC hydrogels showed evidence of chondroprotection, as measured by the histologic grading and decreased expression of biochemical markers of inflammation and apoptosis. It also lowered subchondral bone mineral density, which can be increased by OA. This suggests that the SAP-MSC complex may have clinical potential to inhibit OA progression.
Collapse
Affiliation(s)
- Ji Eun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Sang Mok Lee
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Seoul, South Korea
| | - Soo Hyun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Phil Tatman
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Albert O Gee
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA, USA ; Institute for Stem Cell and Regenerative Medicine and Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Kyung Eun Lee
- Advanced Analysis Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Sang Jun Kim
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
97
|
Alekseenko LL, Zemelko VI, Domnina AP, Lyublinskaya OG, Zenin VV, Pugovkina NA, Kozhukharova IV, Borodkina AV, Grinchuk TM, Fridlyanskaya II, Nikolsky NN. Sublethal heat shock induces premature senescence rather than apoptosis in human mesenchymal stem cells. Cell Stress Chaperones 2014; 19:355-66. [PMID: 24078383 PMCID: PMC3982025 DOI: 10.1007/s12192-013-0463-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 12/12/2022] Open
Abstract
Stem cells in adult organism are responsible for cell turnover and tissue regeneration. The study of stem cell stress response contributes to our knowledge on the mechanisms of damaged tissue repair. Previously, we demonstrated that sublethal heat shock (HS) induced apoptosis in human embryonic stem cells. This study aimed to investigate HS response of human adult stem cells. Human mesenchymal stem cells (MSCs) cultivated in vitro were challenged with sublethal HS. It was found that sublethal HS did not affect the cell viability assessed by annexin V/propidium staining. However, MSCs subjected to severe HS exhibited features of stress-induced premature senescence (SIPS): irreversible cell cycle arrest, altered morphology, increased expression of senescence-associated β-galactosidase (SA-β-gal) activity, and induction of cyclin-dependent kinase inhibitor p21 protein. High level of Hsp70 accumulation induced by sublethal HS did not return to the basal level, at least, after 72 h of the cell recovery when most cells exhibited SIPS hallmarks. MSCs survived sublethal HS, and resumed proliferation sustained the properties of parental MSCs: diploid karyotype, replicative senescence, expression of the cell surface markers, and capacity for multilineage differentiation. Our results showed for the first time that in human MSCs, sublethal HS induced premature senescence rather than apoptosis or necrosis. MSC progeny that survived sublethal HS manifested stem cell properties of the parental cells: limited replicative life span and multilineage capacity.
Collapse
Affiliation(s)
| | | | - Alisa P. Domnina
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | | | - Valery V. Zenin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | | | | | | | | | | | | |
Collapse
|
98
|
Chen J, Li C, Wang S. Periodic heat shock accelerated the chondrogenic differentiation of human mesenchymal stem cells in pellet culture. PLoS One 2014; 9:e91561. [PMID: 24632670 PMCID: PMC3954764 DOI: 10.1371/journal.pone.0091561] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 02/12/2014] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is one of diseases that seriously affect elderly people's quality of life. Human mesenchymal stem cells (hMSCs) offer a potential promise for the joint repair in OA patients. However, chondrogenic differentiation from hMSCs in vitro takes a long time (∼6 weeks) and differentiated cells are still not as functionally mature as primary isolated chondrocytes, though chemical stimulations and mechanical loading have been intensively studied to enhance the hMSC differentiation. On the other hand, thermal stimulations of hMSC chondrogenesis have not been well explored. In this study, the direct effects of mild heat shock (HS) on the differentiation of hMSCs into chondrocytes in 3D pellet culture were investigated. Periodic HS at 41°C for 1 hr significantly increased sulfated glycosaminoglycan in 3D pellet culture at Day 10 of chondrogenesis. Immunohistochemical and Western Blot analyses revealed an increased expression of collagen type II and aggrecan in heat-shocked pellets than non heat-shocked pellets on Day 17 of chondrogenesis. In addition, HS also upregulated the expression of collagen type I and X as well as heat shock protein 70 on Day 17 and 24 of differentiation. These results demonstrate that HS accelerated the chondrogenic differentiation of hMSCs and induced an early maturation of chondrocytes differentiated from hMSCs. The results of this study will guide the design of future protocols using thermal treatments to facilitate cartilage regeneration with human mesenchymal stem cells.
Collapse
Affiliation(s)
- Jing Chen
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York, United States of America
| | - Chenghai Li
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York, United States of America
| | - Sihong Wang
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
99
|
Leskinen JJ, Olkku A, Mahonen A, Hynynen K. Nonuniform Temperature Rise in In Vitro Osteoblast Ultrasound Exposures With Associated Bioeffect. IEEE Trans Biomed Eng 2014; 61:920-7. [DOI: 10.1109/tbme.2013.2292546] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
100
|
Hsiao ST, Dilley RJ, Dusting GJ, Lim SY. Ischemic preconditioning for cell-based therapy and tissue engineering. Pharmacol Ther 2013; 142:141-53. [PMID: 24321597 DOI: 10.1016/j.pharmthera.2013.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/02/2013] [Indexed: 01/07/2023]
Abstract
Cell- and tissue-based therapies are innovative strategies to repair and regenerate injured hearts. Despite major advances achieved in optimizing these strategies in terms of cell source and delivery method, the clinical outcome of cell-based therapy remains unsatisfactory. The non-genetic approach of ischemic/hypoxic preconditioning to enhance cell- and tissue-based therapies has received much attention in recent years due to its non-invasive drug-free application. Here we discuss the current development of hypoxic/ischemic preconditioning to enhance stem cell-based cardiac repair and regeneration.
Collapse
Affiliation(s)
- Sarah T Hsiao
- Department of Cardiovascular Science, University of Sheffield, United Kingdom
| | - Rodney J Dilley
- Ear Science Institute Australia and Ear Sciences Centre, School of Surgery, University of Western Australia, Nedlands, Western Australia, Australia
| | - Gregory J Dusting
- Centre for Eye Research Australia and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Department of Ophthalmology, University of Melbourne, East Melbourne, Victoria, Australia; Department of Surgery, University of Melbourne, Fitzroy, Victoria, Australia; O'Brien Institute, Fitzroy, Victoria, Australia
| | - Shiang Y Lim
- Department of Surgery, University of Melbourne, Fitzroy, Victoria, Australia; O'Brien Institute, Fitzroy, Victoria, Australia.
| |
Collapse
|