1
|
Moghtaderi H, Mohahammadi S, Sadeghian G, Choudhury M, Al-Harrasi A, Rahman SM. Electrical impedance sensing in stem cell research: Insights, applications, and future directions. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:1-14. [PMID: 39557164 DOI: 10.1016/j.pbiomolbio.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
The exceptional differentiation abilities of stem cells make them ideal candidates for cell replacement therapies. Considering their great potential, researchers should understand how stem cells interact with other cell types. The production of high-quality differentiated cells is crucial for favorable treatment and makes them an ideal choice for clinical applications. Label-free stem cell monitoring approaches are anticipated to be more effective in this context, as they ensure quality of differentiation while preserving the therapeutic potential. Electric cell-substrate impedance sensing (ECIS) is a nonintrusive technique that enables cell quantification through continuous monitoring of adherent cell behavior using electronic transcellular impedance measurements. This technique also facilitates the study of cell growth, motility, differentiation, drug effects, and cell barrier functions. Therefore, numerous studies have identified ECIS as an effective method for monitoring stem cell quality and differentiation. In this review, we discuss the current understanding of ECIS's achievements in examining cell behaviors and the potential applications of ECIS arrays in preclinical stem cell research. Moreover, we highlight our present knowledge concerning ECIS's contributions in examining cell behaviors and speculate about the future uses of ECIS arrays in preclinical stem cell research. This review also aims to stimulate research on electrochemical biosensors for future applications in regenerative medicine.
Collapse
Affiliation(s)
- Hassan Moghtaderi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Sultanate of Oman
| | - Saeed Mohahammadi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Sultanate of Oman
| | - Golfam Sadeghian
- Advanced Micro and Nano Device Laboratory, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 1439957131, Iran
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas A & M University, College Station, TX, 77843, USA
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Sultanate of Oman
| | - Shaikh Mizanoor Rahman
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Sultanate of Oman.
| |
Collapse
|
2
|
Chow SKH, Gao Q, Pius A, Morita M, Ergul Y, Murayama M, Shinohara I, Cekuc MS, Ma C, Susuki Y, Goodman SB. The Advantages and Shortcomings of Stem Cell Therapy for Enhanced Bone Healing. Tissue Eng Part C Methods 2024; 30:415-430. [PMID: 39311464 DOI: 10.1089/ten.tec.2024.0252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
This review explores the regenerative potential of key progenitor cell types and therapeutic strategies to improve healing of complex fractures and bone defects. We define, summarize, and discuss the differentiation potential of totipotent, pluripotent, and multipotent stem cells, emphasizing the advantages and shortcomings of cell therapy for bone repair and regeneration. The fundamental role of mesenchymal stem cells is highlighted due to their multipotency to differentiate into the key lineage cells including osteoblasts, osteocytes, and chondrocytes, which are crucial for bone formation and remodeling. Hematopoietic stem cells (HSCs) also play a significant role; immune cells such as macrophages and T-cells modulate inflammation and tissue repair. Osteoclasts are multinucleated cells that are important to bone remodeling. Vascular progenitor (VP) cells are critical to oxygen and nutrient supply. The dynamic interplay among these lineages and their microenvironment is essential for effective bone restoration. Therapies involving cells that are more than "minimally manipulated" are controversial and include embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). ESCs, derived from early-stage embryos, possess pluripotent capabilities and have shown promise in preclinical studies for bone healing. iPSCs, reprogrammed from somatic cells, offer personalized medicine applications and can differentiate into various tissue-specific cell lines. Minimally manipulative cell therapy approaches such as the use of bone marrow aspirate concentrate (BMAC), exosomes, and various biomaterials for local delivery are explored for their effectiveness in bone regeneration. BMAC, which contains mostly immune cells but few mesenchymal and VPs, probably improves bone healing by facilitating paracrine-mediated intercellular communication. Exosome isolation harnesses the biological signals and cellular by-products that are a primary source for cell crosstalk and activation. Safe, efficacious, and cost-effective strategies to enhance bone healing using novel cellular therapies are part of a changing paradigm to modulate the inflammatory, repair, and regenerative pathways to achieve earlier more robust tissue healing and improved physical function. Impact Statement Stem cell therapy holds immense potential for bone healing due to its ability to regenerate damaged tissue. Nonmanipulated bone marrow aspirate contains mesenchymal stem cells that promote bone repair and reduce healing time. Induced pluripotent stem cells offer the advantage of creating patient-specific cells that can differentiate into osteoblasts, aiding in bone regeneration. Other delivery methods, such as scaffold-based techniques, enhance stem cell integration and function. Collectively, these approaches can improve treatment outcomes, reduce recovery periods, and advance our understanding of bone healing mechanisms, making them pivotal in orthopedic research and regenerative medicine.
Collapse
Affiliation(s)
- Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Alexa Pius
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mayu Morita
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yasemin Ergul
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mehmet Sertac Cekuc
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Chao Ma
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yosuke Susuki
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
3
|
Dadashi Ouranj Z, Hosseini S, Alipour A, Homaeigohar S, Azari S, Ghazizadeh L, Shokrgozar M, Thomas S, Irian S, Shahsavarani H. The potent osteo-inductive capacity of bioinspired brown seaweed-derived carbohydrate nanofibrous three-dimensional scaffolds. MARINE LIFE SCIENCE & TECHNOLOGY 2024; 6:515-534. [PMID: 39219680 PMCID: PMC11358581 DOI: 10.1007/s42995-024-00241-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/11/2024] [Indexed: 09/04/2024]
Abstract
This study aimed to investigate the osteo-inductive capacity of a fucoidan polysaccharide network derived from brown algae on human adipose-derived stem cells (HA-MSCs) for bone regeneration. The physiochemical properties of the scaffold including surface morphology, surface chemistry, hydrophilicity, mechanical stiffness, and porosity were thoroughly characterized. Both in vitro and in vivo measurements implied a superior cell viability, proliferation, adhesion, and osteo-inductive performance of obtained scaffolds compared to using specific osteogenic induction medium with increased irregular growth of calcium crystallites, which mimic the structure of natural bones. That scaffold was highly biocompatible and suitable for cell cultures. Various examinations, such as quantification of mineralization, alkaline phosphatase, gene expression, and immunocytochemical staining of pre-osteocyte and bone markers confirmed that HAD-MSCs differentiate into osteoblasts, even without an osteogenic induction medium. This study provides evidence for the positive relationship and synergistic effects between the physical properties of the decellularized seaweed scaffold and the chemical composition of fucoidan in promoting the osteogenic differentiation of HA-MSCs. Altogether, the natural matrices derived from brown seaweed offers a sustainable, cost-effective, non-toxic bioinspired scaffold and holds promise for future clinical applications in orthopedics.
Collapse
Affiliation(s)
- Zahra Dadashi Ouranj
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, 15719-14911 Iran
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Saadi Hosseini
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Atefeh Alipour
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Shahin Homaeigohar
- School of Science and Engineering, University of Dundee, Dundee, DD1 4HN UK
| | - Shahram Azari
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Leila Ghazizadeh
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Mohammadali Shokrgozar
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Sabu Thomas
- School of Chemical Sciences, Mahatma Gandhi University, Kottayam, Kerala India
| | - Saeed Irian
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, 15719-14911 Iran
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, 19839-69411 Iran
- Iranian Biological Resource Center, Academic Center for Education, Culture and Research (ACECR), Tehran, 1533734716 Iran
| |
Collapse
|
4
|
Hu C, Huang R, Xia J, Hu X, Xie D, Jin Y, Qi W, Zhao C, Hu Z. A nanozyme-functionalized bilayer hydrogel scaffold for modulating the inflammatory microenvironment to promote osteochondral regeneration. J Nanobiotechnology 2024; 22:445. [PMID: 39069607 PMCID: PMC11283693 DOI: 10.1186/s12951-024-02723-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/14/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND The incidence of osteochondral defects caused by trauma, arthritis or tumours is increasing annually, but progress has not been made in terms of treatment methods. Due to the heterogeneous structure and biological characteristics of cartilage and subchondral bone, the integration of osteochondral repair is still a challenge. RESULTS In the present study, a novel bilayer hydrogel scaffold was designed based on anatomical characteristics to imitate superficial cartilage and subchondral bone. The scaffold showed favourable biocompatibility, and the addition of an antioxidant nanozyme (LiMn2O4) promoted reactive oxygen species (ROS) scavenging by upregulating antioxidant proteins. The cartilage layer effectively protects against chondrocyte degradation in the inflammatory microenvironment. Subchondral bionic hydrogel scaffolds promote osteogenic differentiation of rat bone marrow mesenchymal stem cells (BMSCs) by regulating the AMPK pathway in vitro. Finally, an in vivo rat preclinical osteochondral defect model confirmed that the bilayer hydrogel scaffold efficiently promoted cartilage and subchondral bone regeneration. CONCLUSIONS In general, our biomimetic hydrogel scaffold with the ability to regulate the inflammatory microenvironment can effectively repair osteochondral defects. This strategy provides a promising method for regenerating tissues with heterogeneous structures and biological characteristics.
Collapse
Affiliation(s)
- Chuan Hu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Ruipeng Huang
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jiechao Xia
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Xianjing Hu
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhu, 325000, China
| | - Dingqi Xie
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Yang Jin
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Weiming Qi
- Zhejiang Center for Medical Device Evaluation, Zhejiang Medical Products Administration, Hangzhou, 310009, China.
| | - Chengliang Zhao
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Zhijun Hu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
5
|
Yanli Z, Jiayao M, Chunqing Z, Yuting Z, Zhiyan Z, Yulin Z, Minghan L, Longquan S, Dehong Y, Wenjuan Y. MY-1-Loaded Nano-Hydroxyapatite Accelerated Bone Regeneration by Increasing Type III Collagen Deposition in Early-Stage ECM via a Hsp47-Dependent Mechanism. Adv Healthc Mater 2023; 12:e2300332. [PMID: 36999955 DOI: 10.1002/adhm.202300332] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/13/2023] [Indexed: 04/01/2023]
Abstract
The extracellular matrix (ECM) plays a crucial part in regulating stem cell function through its distinctive mechanical and chemical effect. Therefore, it is worth studying how to activate the driving force of osteoblast cells by dynamic changing of ECM and accelerate the bone regeneration. In this research, a novel peptide MY-1 is designed and synthesized. To achieve its sustained releasing, the nano-hydroxyapatite (nHA) is chosen as the carrier of MY-1 by mixed adsorption. The results reveal that the sustainable releasing of MY-1 regulates the synthesis and secretion of ECM from rat bone marrow mesenchymal stem cells (rBMSCs), which promotes the cell migration and osteogenic differentiation in the early stage of bone regeneration. Further analyses demonstrate that MY-1 increases the expression and nuclear translocation of β-catenin, and then upregulates the level of heat shock protein 47 (Hsp47), thereby accelerating the synthesis and secretion of type III collagen (Col III) at the early stage. Finally, the promoted rapid transformation of Col III to Col I at late stage benefits the bone regeneration. Hence, this study can provide a theoretical basis for the local application of MY-1 in bone regeneration.
Collapse
Affiliation(s)
- Zhang Yanli
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Mo Jiayao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Zheng Chunqing
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Zeng Yuting
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Zhou Zhiyan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Zhang Yulin
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Li Minghan
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Shao Longquan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yang Dehong
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yan Wenjuan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
6
|
Galefi A, Nourany M, Hosseini S, Alipour A, Azari S, Jahanfar M, Farrokhi N, Homaeigohar S, Shahsavarani H. Enhanced osteogenesis on proantocyanidin-loaded date palm endocarp cellulosic matrices: A novel sustainable approach for guided bone regeneration. Int J Biol Macromol 2023; 242:124857. [PMID: 37187421 DOI: 10.1016/j.ijbiomac.2023.124857] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/17/2023]
Abstract
Developing inexpensive, biocompatible natural scaffolds that can support the differentiation and proliferation of stem cells has been recently emphasized by the research community to faster obtain the FDA approvals for regenerative medicine. In this regard, plant-derived cellulose materials are a novel class of sustainable scaffolding materials with high potentials for bone tissue engineering (BTE). However, low bioactivity of the plant-derived cellulose scaffolds restricts cell proliferation and cell differentiation. This limitation can be addressed though surface-functionalization of cellulose scaffolds with natural antioxidant polyphenols, e.g., grape seed proanthocyanidin (PCA)-rich extract (GSPE). Despite the various merits of GSPE as a natural antioxidant, its impact on the proliferation and adhesion of osteoblast precursor cells, and on their osteogenic differentiation is an as-yet unknown issue. Here, we investigated the effects of GSPE surface functionalization on the physicochemical properties of decellularized date (Phoenix dactyliferous) fruit inner layer (endocarp) (DE) scaffold. In this regard, various physiochemical characteristics of the DE-GSPE scaffold such as hydrophilicity, surface roughness, mechanical stiffness, porosity, and swelling, and biodegradation behavior were compared with those of the DE scaffold. Additionally, the impact of the GSPE treatment of the DE scaffold on the osteogenic response of human mesenchymal stem cells (hMSCs) was thoroughly studied. For this purpose, cellular activities including cell adhesion, calcium deposition and mineralization, alkaline phosphatase (ALP) activity, and expression levels of bone-related genes were monitored. Taken together, the GSPE treatment enhanced the physicochemical and biological properties of the DE-GSPE scaffold, thereby raising its potentials as a promising candidate for guided bone regeneration.
Collapse
Affiliation(s)
- Atena Galefi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 19839-69411, Iran; Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran 13169-43551, Iran
| | - Mohammad Nourany
- Amirkabir University of Technology, Polymer Engineering and Color Technology, Tehran, Iran; Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran 13169-43551, Iran
| | - Saadi Hosseini
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran 13169-43551, Iran
| | - Atefeh Alipour
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran.
| | - Shahram Azari
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran 13169-43551, Iran
| | - Mehdi Jahanfar
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 19839-69411, Iran
| | - Naser Farrokhi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 19839-69411, Iran
| | - Shahin Homaeigohar
- School of Science & Engineering, University of Dundee, Dundee DD1 4HN, UK
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 19839-69411, Iran; Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran 13169-43551, Iran.
| |
Collapse
|
7
|
Liang C, Liao L, Tian W. Advances Focusing on the Application of Decellularized Extracellular Matrix in Periodontal Regeneration. Biomolecules 2023; 13:673. [PMID: 37189420 PMCID: PMC10136219 DOI: 10.3390/biom13040673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/01/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
The decellularized extracellular matrix (dECM) is capable of promoting stem cell proliferation, migration, adhesion, and differentiation. It is a promising biomaterial for application and clinical translation in the field of periodontal tissue engineering as it most effectively preserves the complex array of ECM components as they are in native tissue, providing ideal cues for regeneration and repair of damaged periodontal tissue. dECMs of different origins have different advantages and characteristics in promoting the regeneration of periodontal tissue. dECM can be used directly or dissolved in liquid for better flowability. Multiple ways were developed to improve the mechanical strength of dECM, such as functionalized scaffolds with cells that harvest scaffold-supported dECM through decellularization or crosslinked soluble dECM that can form injectable hydrogels for periodontal tissue repair. dECM has found recent success in many periodontal regeneration and repair therapies. This review focuses on the repairing effect of dECM in periodontal tissue engineering, with variations in cell/tissue sources, and specifically discusses the future trend of periodontal regeneration and the future role of soluble dECM in entire periodontal tissue regeneration.
Collapse
Affiliation(s)
| | - Li Liao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Sichuan 610041, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Sichuan 610041, China
| |
Collapse
|
8
|
Okutan B, Schwarze UY, Berger L, Martinez DC, Herber V, Suljevic O, Plocinski T, Swieszkowski W, Santos SG, Schindl R, Löffler JF, Weinberg AM, Sommer NG. The combined effect of zinc and calcium on the biodegradation of ultrahigh-purity magnesium implants. BIOMATERIALS ADVANCES 2023; 146:213287. [PMID: 36669235 DOI: 10.1016/j.bioadv.2023.213287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 12/15/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
Magnesium (Mg)-based implants are promising candidates for orthopedic interventions, because of their biocompatibility, good mechanical features, and ability to degrade completely in the body, eliminating the need for an additional removal surgery. In the present study, we synthesized and investigated two Mg-based materials, ultrahigh-purity ZX00 (Mg-Zn-Ca; <0.5 wt% Zn and <0.5 wt% Ca, in wt%; Fe-content <1 ppm) and ultrahigh-purity Mg (XHP-Mg, >99.999 wt% Mg; Fe-content <1 ppm), in vitro and in vivo in juvenile healthy rats to clarify the effect of the alloying elements Zn and Ca on mechanical properties, microstructure, cytocompatibility and degradation rate. Potential differences in bone formation and bone in-growth were also assessed and compared with state-of-the-art non-degradable titanium (Ti)-implanted, sham-operated, and control (non-intervention) groups, using micro-computed tomography, histology and scanning electron microscopy. At 6 and 24 weeks after implantation, serum alkaline phosphatase (ALP), calcium (Ca), and Mg level were measured and bone marrow stromal cells (BMSCs) were isolated for real-time PCR analysis. Results show that ZX00 implants have smaller grain size and superior mechanical properties than XHP-Mg, and that both reveal good biocompatibility in cytocompatibilty tests. ZX00 homogenously degraded with an increased gas accumulation 12 and 24 weeks after implantation, whereas XHP-Mg exhibited higher gas accumulation already at 2 weeks. Serum ALP, Ca, and Mg levels were comparable among all groups and both Mg-based implants led to similar relative expression levels of Alp, Runx2, and Bmp-2 genes at weeks 6 and 24. Histologically, Mg-based implants are superior for new bone tissue formation and bone in-growth compared to Ti implants. Furthermore, by tracking the sequence of multicolor fluorochrome labels, we observed higher mineral apposition rate at week 2 in both Mg-based implants compared to the control groups. Our findings suggest that (i) ZX00 and XHP-Mg support bone formation and remodeling, (ii) both Mg-based implants are superior to Ti implants in terms of new bone tissue formation and osseointegration, and (iii) ZX00 is more favorable due to its lower degradation rate and moderate gas accumulation.
Collapse
Affiliation(s)
- Begüm Okutan
- Department of Orthopedics and Traumatology, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| | - Uwe Y Schwarze
- Department of Orthopedics and Traumatology, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria; Department of Dentistry and Oral Health, Division of Oral Surgery and Orthodontics, Medical University of Graz, Billrothgasse 4, 8010 Graz, Austria.
| | - Leopold Berger
- Laboratory of Metal Physics and Technology, Department of Materials, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland.
| | - Diana C Martinez
- Department of Materials Science and Engineering, Warsaw University of Technology, Woloska 141, 02-507 Warsaw, Poland.
| | - Valentin Herber
- Department of Dentistry and Oral Health, Division of Oral Surgery and Orthodontics, Medical University of Graz, Billrothgasse 4, 8010 Graz, Austria; Department of Oral Surgery, University Center for Dental Medicine, University of Basel, Mattenstrasse 40, 4058 Basel, Switzerland.
| | - Omer Suljevic
- Department of Orthopedics and Traumatology, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| | - Tomasz Plocinski
- Department of Materials Science and Engineering, Warsaw University of Technology, Woloska 141, 02-507 Warsaw, Poland.
| | - Wojciech Swieszkowski
- Department of Materials Science and Engineering, Warsaw University of Technology, Woloska 141, 02-507 Warsaw, Poland.
| | - Susana G Santos
- i3S - Instituto de Investigação e Inovação em Saúde, and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal.
| | - Rainer Schindl
- Gottfried Schatz Research Center, Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria.
| | - Jörg F Löffler
- Laboratory of Metal Physics and Technology, Department of Materials, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland.
| | - Annelie M Weinberg
- Department of Orthopedics and Traumatology, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| | - Nicole G Sommer
- Department of Orthopedics and Traumatology, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| |
Collapse
|
9
|
Wang B, Qinglai T, Yang Q, Li M, Zeng S, Yang X, Xiao Z, Tong X, Lei L, Li S. Functional acellular matrix for tissue repair. Mater Today Bio 2023; 18:100530. [PMID: 36601535 PMCID: PMC9806685 DOI: 10.1016/j.mtbio.2022.100530] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022] Open
Abstract
In view of their low immunogenicity, biomimetic internal environment, tissue- and organ-like physicochemical properties, and functionalization potential, decellularized extracellular matrix (dECM) materials attract considerable attention and are widely used in tissue engineering. This review describes the composition of extracellular matrices and their role in stem-cell differentiation, discusses the advantages and disadvantages of existing decellularization techniques, and presents methods for the functionalization and characterization of decellularized scaffolds. In addition, we discuss progress in the use of dECMs for cartilage, skin, nerve, and muscle repair and the transplantation or regeneration of different whole organs (e.g., kidneys, liver, uterus, lungs, and heart), summarize the shortcomings of using dECMs for tissue and organ repair after refunctionalization, and examine the corresponding future prospects. Thus, the present review helps to further systematize the application of functionalized dECMs in tissue/organ transplantation and keep researchers up to date on recent progress in dECM usage.
Collapse
Affiliation(s)
- Bin Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Tang Qinglai
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Zian Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinying Tong
- Department of Hemodialysis, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
10
|
TiO 2/HA and Titanate/HA Double-Layer Coatings on Ti6Al4V Surface and Their Influence on In Vitro Cell Growth and Osteogenic Potential. J Funct Biomater 2022; 13:jfb13040271. [PMID: 36547531 PMCID: PMC9787412 DOI: 10.3390/jfb13040271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022] Open
Abstract
Hydroxyapatite (HA) layers are appropriate biomaterials for use in the modification of the surface of implants produced inter alia from a Ti6Al4V alloy. The issue that must be solved is to provide implants with appropriate biointegration properties, enabling the permanent link between them and bone tissues, which is not so easy with the HA layer. Our proposition is the use of the intermediate layer ((IL) = TiO2, and titanate layers) to successfully link the HA coating to a metal substrate (Ti6Al4V). The morphology, structure, and chemical composition of Ti6Al4V/IL/HA systems were characterized by scanning electron microscopy (SEM), X-ray diffraction (XRD), and energy-dispersive X-ray spectrometry (EDS). We evaluated the apatite-forming ability on the surface of the layer in simulated body fluid. We investigated the effects of the obtained systems on the viability and growth of human MG-63 osteoblast-like cells, mouse L929 fibroblasts, and adipose-derived human mesenchymal stem cells (ADSCs) in vitro, as well as on their osteogenic properties. Based on the obtained results, we can conclude that both investigated systems reflect the physiological environment of bone tissue and create a biocompatible surface supporting cell growth. However, the nanoporous TiO2 intermediate layer with osteogenesis-supportive activity seems most promising for the practical application of Ti6Al4V/TiO2/HA as a system of bone tissue regeneration.
Collapse
|
11
|
Alghfeli L, Parambath D, Tag Eldeen LA, El-Serafi I, El-Serafi AT. Non-additive effect of the DNA methylation inhibitor, 5-Aza-dC, and glass as a culture surface on osteogenic differentiation. Heliyon 2022; 8:e12433. [PMID: 36590514 PMCID: PMC9794900 DOI: 10.1016/j.heliyon.2022.e12433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/31/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
The clinical need for bone regenerative solutions is expanding with increasing life expectancy and escalating incidence of accidents. Several strategies are being investigated to enhance the osteogenic differentiation of stem cells. We previously reported two different approaches for this purpose, in monolayer and three-dimensional cell culture. The first approach was based on pretreating cells with 5-Aza-dC, a DNA methylation inhibitor, before the applying the differentiation media. The second approach was based on culturing cells on a glass surface during differentiation. In this study, we investigated the potential effect of combining both methods. Our results suggested that both approaches were associated with decreasing global DNA methylation levels. Cells cultured as a monolayer on glass surface showed enhancement in alkaline phosphatase activity at day 10, while 5-Aza-dC pretreatment enhanced the activity at day 5, irrespective of the culture surface. In three-dimensional pellet culture, 5-Aza-dC pretreatment enhanced osteogenesis through Runx-2 and TGF-β1 upregulation while the glass surface induced Osterix. Furthermore, pellets cultured on glass showed upregulation of a group of miRNAs, including pro-osteogenesis miR- 20a and miR -148b and anti-osteogenesis miR -125b, miR -31, miR -138, and miR -133a. Interestingly, 5-Aza-dC was not associated with a change of miRNAs in cells cultured on tissue culture plastic but reverted the upregulated miRNAs on the glass to the basal level. This study confirms the two approaches for enhancing osteogenic differentiation and contradicts their combination.
Collapse
Affiliation(s)
- Latifa Alghfeli
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Divyasree Parambath
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Loaa A. Tag Eldeen
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Egypt
| | - Ibrahim El-Serafi
- Basic Medical Sciences Department, College of Medicine, Ajman University, United Arab Emirates
- Department of Biochemistry, Faculty of Medicine, Port-Said University, Egypt
| | - Ahmed T. El-Serafi
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Egypt
- Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| |
Collapse
|
12
|
Effects of vitamin B12 in culture medium for calcified nodule formation by rat dental pulp cells. J Dent Sci 2022. [DOI: 10.1016/j.jds.2022.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
13
|
Turner NJ, Quijano LM, Hussey GS, Jiang P, Badylak SF. Matrix Bound Nanovesicles have Tissue Specific Characteristics that Suggest a Regulatory Role. Tissue Eng Part A 2022; 28:879-892. [PMID: 35946072 DOI: 10.1089/ten.tea.2022.0091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recent studies have identified an extracellular vesicle population that is tightly anchored within the extracellular matrix of tissues and organs until released by matrix turnover events. Evidence suggests that these matrix-bound nanovesicles (MBV) are a ubiquitous component of the ECM, raising questions regarding their tissue specific identity and their biologic function(s). The primary objective of this study was to examine MBV isolated from six different tissues and compare their physical and compositional characteristics to determine the common and differentially expressed features. Accordingly, the results of this characterization show that while MBV are a ubiquitous component of the ECM they contain a protein and miRNA cargo that is tissue specific. The results furthermore suggest that MBV have an important role in regulating tissue homeostasis.
Collapse
Affiliation(s)
- Neill J Turner
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, 450 Technology Drive, Suite 300, Pittsburgh, Pennsylvania, United States, 15212;
| | - Lina Maria Quijano
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States;
| | - George S Hussey
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, 450 Technology Dr., Pittsburgh, Pennsylvania, United States, 15219;
| | - Peng Jiang
- Cleveland State University, Center for Gene Regulation in Health and Disease, Cleveland, Ohio, United States;
| | - Stephen F Badylak
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, 450 Technology Drive, Suite 300, Pittsburgh, Pennsylvania, United States, 15219;
| |
Collapse
|
14
|
Xu F, Zheng Z, Yao M, Zhu F, Shen T, Li J, Zhu C, Yang T, Shao M, Wan Z, Fang C. A regulatory mechanism of a stepwise osteogenesis-mimicking decellularized extracellular matrix on the osteogenic differentiation of bone marrow-derived mesenchymal stem cells. J Mater Chem B 2022; 10:6171-6180. [PMID: 35766339 DOI: 10.1039/d2tb00721e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A cell-derived decellularized extracellular matrix (dECM) plays a vital role in controlling cell functions because of its similarity to the in vivo microenvironment. In the process of stem cell differentiation, the composition of the dECM is not constant but is dynamically remolded. However, there is little information regarding the dynamic regulation by the dECM of the osteogenic differentiation of stem cells. Herein, four types of stepwise dECMs (0, 7, 14, and 21 d-ECM) were prepared from bone marrow-derived mesenchymal stem cells (BMSCs) undergoing osteogenic differentiation for 0, 7, 14, and 21 days after decellularization. In vitro experiments were designed to study the regulation of BMSC osteogenesis by dECMs. The results showed that all the dECMs could support the activity and proliferation of BMSCs but had different effects on their osteogenic differentiation. The 14d-ECM promoted the osteogenesis of BMSCs significantly compared with the other dECMs. Proteomic analysis demonstrated that the composition of dECMs changed over time. The 14d ECM had higher amounts of collagen type IV alpha 2 chain (COL4A2) than the other dECMs. Furthermore, COL4A2 was obviously enriched in the activated focal adhesion kinase (FAK)/phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways. Thus, the 14d-ECM could promote the osteogenic differentiation of BMSCs, which might be related to the high content of COL4A2 in the 14d-ECM by activating the FAK/PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Fei Xu
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Ziran Zheng
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Mianfeng Yao
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Feiya Zhu
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Ting Shen
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Jiang Li
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Chao Zhu
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Tianru Yang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Mengying Shao
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Zicheng Wan
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Changyun Fang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| |
Collapse
|
15
|
Bloise N, Waldorff EI, Montagna G, Bruni G, Fassina L, Fang S, Zhang N, Jiang J, Ryaby JT, Visai L. Early Osteogenic Marker Expression in hMSCs Cultured onto Acid Etching-Derived Micro- and Nanotopography 3D-Printed Titanium Surfaces. Int J Mol Sci 2022; 23:7070. [PMID: 35806083 PMCID: PMC9266831 DOI: 10.3390/ijms23137070] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 12/13/2022] Open
Abstract
Polyetheretherketone (PEEK) titanium composite (PTC) is a novel interbody fusion device that combines a PEEK core with titanium alloy (Ti6Al4V) endplates. The present study aimed to investigate the in vitro biological reactivity of human bone-marrow-derived mesenchymal stem cells (hBM-MSCs) to micro- and nanotopographies produced by an acid-etching process on the surface of 3D-printed PTC endplates. Optical profilometer and scanning electron microscopy were used to assess the surface roughness and identify the nano-features of etched or unetched PTC endplates, respectively. The viability, morphology and the expression of specific osteogenic markers were examined after 7 days of culture in the seeded cells. Haralick texture analysis was carried out on the unseeded endplates to correlate surface texture features to the biological data. The acid-etching process modified the surface roughness of the 3D-printed PTC endplates, creating micro- and nano-scale structures that significantly contributed to sustaining the viability of hBM-MSCs and triggering the expression of early osteogenic markers, such as alkaline phosphatase activity and bone-ECM protein production. Finally, the topography of 3D-printed PTC endplates influenced Haralick's features, which in turn correlated with the expression of two osteogenic markers, osteopontin and osteocalcin. Overall, these data demonstrate that the acid-etching process of PTC endplates created a favourable environment for osteogenic differentiation of hBM-MSCs and may potentially have clinical benefit.
Collapse
Affiliation(s)
- Nora Bloise
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, 27100 Pavia, Italy;
- Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100 Pavia, Italy
| | - Erik I. Waldorff
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Giulia Montagna
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, 27100 Pavia, Italy;
- Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (CHT), University of Pavia, 27100 Pavia, Italy;
| | - Giovanna Bruni
- C.S.G.I.-Department of Chemistry, Section of Physical Chemistry, University of Pavia, 27100 Pavia, Italy;
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (CHT), University of Pavia, 27100 Pavia, Italy;
| | - Samuel Fang
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Nianli Zhang
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Jiechao Jiang
- Department of Material Science, University of Texas, Arlington, TX 76019, USA;
| | - James T. Ryaby
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Livia Visai
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, 27100 Pavia, Italy;
- Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100 Pavia, Italy
| |
Collapse
|
16
|
Wu H, Yin G, Pu X, Wang J, Liao X, Huang Z. Coordination of Osteoblastogenesis and Osteoclastogenesis by the Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Matrix To Promote Bone Regeneration. ACS APPLIED BIO MATERIALS 2022; 5:2913-2927. [DOI: 10.1021/acsabm.2c00264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Huan Wu
- College of Biomedical Engineering, Sichuan University, No.24, South 1st Section, 1st Ring Road, Chengdu 610064, P. R. China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, No.24, South 1st Section, 1st Ring Road, Chengdu 610064, P. R. China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, No.24, South 1st Section, 1st Ring Road, Chengdu 610064, P. R. China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, No.24, South 1st Section, 1st Ring Road, Chengdu 610064, P. R. China
| | - Xiaoming Liao
- College of Biomedical Engineering, Sichuan University, No.24, South 1st Section, 1st Ring Road, Chengdu 610064, P. R. China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, No.24, South 1st Section, 1st Ring Road, Chengdu 610064, P. R. China
| |
Collapse
|
17
|
Qi D, Su J, Li S, Zhu H, Cheng L, Hua S, Yuan X, Jiang J, Shu Z, Shi Y, Xiao J. 3D printed magnesium-doped β-TCP gyroid scaffold with osteogenesis, angiogenesis, immunomodulation properties and bone regeneration capability in vivo. BIOMATERIALS ADVANCES 2022; 136:212759. [PMID: 35929304 DOI: 10.1016/j.bioadv.2022.212759] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/25/2022] [Accepted: 03/10/2022] [Indexed: 06/15/2023]
Abstract
Bioceramics have been used in orthopedic surgery for several years. Magnesium (Mg) is an essential element in bone tissue and plays an important role in bone metabolism. Mg-doped bioceramics has attracted the attention of researchers recently. However, the optimal doping amount of Mg in β-TCP and the immunomodulatory property of Mg-doped β-TCP (Mg-TCP) have not been determined yet. In this study, β-TCP scaffolds doped with different contents of magnesium oxide (0 wt%, 1 wt%, 3 wt%, and 5 wt%) with gyroid structure were printed by digital light processing (DLP) method, and the physicochemical and biological functions were then investigated. Mg-doping improved the physicochemical properties of the β-TCP scaffolds. In vitro experiments confirmed that the doping of Mg in β-TCP scaffolds promoted the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and angiogenic differentiation of endothelial progenitor cells (EPCs), where the 5Mg-TCP has the optimal properties when using the "one cell type" method. It was also found that all Mg-TCP facilitated the polarization of RAW264.7 cells to the M2 phenotype, especially the 3Mg-TCP. However, 3Mg-TCP displayed the optimal osteogenic and angiogenic potential when using a "multiple cell type" method, which referred to culturing the BMSCs or EPCs in the macrophage-conditioned medium. Finally, the in vivo experiments were conducted and the results confirmed that the 3Mg-TCP scaffolds possessed the satisfying bone defect repair capability both after 6 and 12 weeks of implantation. This study suggests that 3Mg-TCP scaffolds provide the optimal biological performance and thus have the potential for clinical translation.
Collapse
Affiliation(s)
- Dahu Qi
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Song Li
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Hao Zhu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Lijin Cheng
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shuaibin Hua
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xi Yuan
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jiawei Jiang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Zixing Shu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yusheng Shi
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Jun Xiao
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
18
|
Wan HY, Shin RLY, Chen JCH, Assunção M, Wang D, Nilsson SK, Tuan RS, Blocki A. Dextran sulfate-amplified extracellular matrix deposition promotes osteogenic differentiation of mesenchymal stem cells. Acta Biomater 2022; 140:163-177. [PMID: 34875356 DOI: 10.1016/j.actbio.2021.11.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022]
Abstract
The development of bone-like tissues in vitro that exhibit key features similar to those in vivo is needed to produce tissue models for drug screening and the study of bone physiology and disease pathogenesis. Extracellular matrix (ECM) is a predominant component of bone in vivo; however, as ECM assembly is sub-optimal in vitro, current bone tissue engineering approaches are limited by an imbalance in ECM-to-cell ratio. We amplified the deposition of osteoblastic ECM by supplementing dextran sulfate (DxS) into osteogenically induced cultures of human mesenchymal stem cells (MSCs). DxS, previously implicated to act as a macromolecular crowder, was recently demonstrated to aggregate and co-precipitate major ECM components, including collagen type I, thereby amplifying its deposition. This effect was re-confirmed for MSC cultures undergoing osteogenic induction, where DxS supplementation augmented collagen type I deposition, accompanied by extracellular osteocalcin accumulation. The resulting differentiated osteoblasts exhibited a more mature osteogenic gene expression profile, indicated by a strong upregulation of the intermediate and late osteogenic markers ALP and OCN, respectively. The associated cellular microenvironment was also enriched in bone morphogenetic protein 2 (BMP-2). Interestingly, the resulting decellularized matrices exhibited the strongest osteo-inductive effects on re-seeded MSCs, promoted cell proliferation, osteogenic marker expression and ECM calcification. Taken together, these findings suggest that DxS-mediated enhancement of osteogenic differentiation by MSCs is mediated by the amplified ECM, which is enriched in osteo-inductive factors. We have thus established a simple and reproducible approach to generate ECM-rich bone-like tissue in vitro with sequestration of osteo-inductive factors. STATEMENT OF SIGNIFICANCE: As extracellular matrix (ECM) assembly is significantly retarded in vitro, the imbalance in ECM-to-cell ratio hampers current in vitro bone tissue engineering approaches in their ability to faithfully resemble their in vivo counterpart. We addressed this limitation by leveraging a poly-electrolyte mediated co-assembly and amplified deposition of ECM during osteogenic differentiation of human mesenchymal stem cells (MSCs). The resulting pericelluar space in culture was enriched in organic and inorganic bone ECM components, as well as osteo-inductive factors, which promoted the differentiation of MSCs towards a more mature osteoblastic phenotype. These findings thus demonstrated a simple and reproducible approach to generate ECM-rich bone-like tissue in vitro with a closer recapitulation of the in vivo tissue niche.
Collapse
|
19
|
Liu C, Pei M, Li Q, Zhang Y. Decellularized extracellular matrix mediates tissue construction and regeneration. Front Med 2022; 16:56-82. [PMID: 34962624 PMCID: PMC8976706 DOI: 10.1007/s11684-021-0900-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023]
Abstract
Contributing to organ formation and tissue regeneration, extracellular matrix (ECM) constituents provide tissue with three-dimensional (3D) structural integrity and cellular-function regulation. Containing the crucial traits of the cellular microenvironment, ECM substitutes mediate cell-matrix interactions to prompt stem-cell proliferation and differentiation for 3D organoid construction in vitro or tissue regeneration in vivo. However, these ECMs are often applied generically and have yet to be extensively developed for specific cell types in 3D cultures. Cultured cells also produce rich ECM, particularly stromal cells. Cellular ECM improves 3D culture development in vitro and tissue remodeling during wound healing after implantation into the host as well. Gaining better insight into ECM derived from either tissue or cells that regulate 3D tissue reconstruction or organ regeneration helps us to select, produce, and implant the most suitable ECM and thus promote 3D organoid culture and tissue remodeling for in vivo regeneration. Overall, the decellularization methodologies and tissue/cell-derived ECM as scaffolds or cellular-growth supplements used in cell propagation and differentiation for 3D tissue culture in vitro are discussed. Moreover, current preclinical applications by which ECM components modulate the wound-healing process are reviewed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27109, USA.
| |
Collapse
|
20
|
Hamid HA, Sarmadi VH, Prasad V, Ramasamy R, Miskon A. Electromagnetic field exposure as a plausible approach to enhance the proliferation and differentiation of mesenchymal stem cells in clinically relevant scenarios. J Zhejiang Univ Sci B 2022; 23:42-57. [PMID: 35029087 PMCID: PMC8758935 DOI: 10.1631/jzus.b2100443] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapy has been regarded as one of the most revolutionary breakthroughs in the history of modern medicine owing to its myriad of immunoregulatory and regenerative properties. With the rapid progress in the fields of osteo- and musculoskeletal therapies, the demand for MSC-based treatment modalities is becoming increasingly prominent. In this endeavor, researchers around the world have devised new and innovative techniques to support the proliferation of MSCs while minimizing the loss of hallmark features of stem cells. One such example is electromagnetic field (EMF) exposure, which is an alternative approach with promising potential. In this review, we present a critical discourse on the efficiency, practicability, and limitations of some of the relevant methods, with insurmountable evidence backing the implementation of EMF as a feasible strategy for the clinically relevant expansion of MSCs.
Collapse
Affiliation(s)
- Haslinda Abdul Hamid
- Bio-artificial Organ and Regenerative Medicine Unit, National Defense University of Malaysia, Kuala Lumpur 57000, Malaysia
| | - Vahid Hosseinpour Sarmadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran 144961 4535, Iran.,Institutes of Regenerative Medicine, Iran University of Medical Sciences, Tehran 199671 4353, Iran
| | - Vivek Prasad
- Stem Cell and Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia
| | - Rajesh Ramasamy
- Stem Cell and Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia
| | - Azizi Miskon
- Bio-artificial Organ and Regenerative Medicine Unit, National Defense University of Malaysia, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
21
|
Xu W, Tan W, Li C, Wu K, Zeng X, Xiao L. Metformin-loaded β-TCP/CTS/SBA-15 composite scaffolds promote alveolar bone regeneration in a rat model of periodontitis. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:145. [PMID: 34862928 PMCID: PMC8643296 DOI: 10.1007/s10856-021-06621-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/07/2021] [Indexed: 06/07/2023]
Abstract
Periodontitis is a progressive infectious inflammatory disease, which leads to alveolar bone resorption and loss of periodontal attachment. It is imperative for us to develop a therapeutic scaffold to repair the alveolar bone defect of periodontitis. In this study, we designed a new composite scaffold loading metformin (MET) by using the freeze-drying method, which was composed of β-tricalcium phosphate (β-TCP), chitosan (CTS) and the mesoporous silica (SBA-15). The scaffolds were expected to combine the excellent biocompatibility of CTS, the good bioactivity of β-TCP, and the anti-inflammatory properties of MET. The MET-loaded β-TCP/CTS/SBA-15 scaffolds showed improved cell adhesion, appropriate porosity and good biocompatibility in vitro. This MET composite scaffold was implanted in the alveolar bone defects area of rats with periodontitis. After 12 weeks, Micro-CT and histological analysis were performed to evaluate different degrees of healing and mineralization. Results showed that the MET-loaded β-TCP/CTS/SBA-15 scaffolds promoted alveolar bone regeneration in a rat model of periodontitis. To our knowledge, this is the first report that MET-loaded β-TCP/CTS/SBA-15 scaffolds have a positive effect on alveolar bone regeneration in periodontitis. Our findings might provide a new and promising strategy for repairing alveolar bone defects under the condition of periodontitis.
Collapse
Affiliation(s)
- Wanghan Xu
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, PR China
- Department of Stomatology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311202, Zhejiang, PR China
| | - Wei Tan
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410011, Hunan, PR China
| | - Chan Li
- Department of Metabolism and Endocrinology, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, PR China
| | - Xinyi Zeng
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, PR China
| | - Liwei Xiao
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, PR China.
| |
Collapse
|
22
|
Dikici S, Aldemir Dikici B, MacNeil S, Claeyssens F. Decellularised extracellular matrix decorated PCL PolyHIPE scaffolds for enhanced cellular activity, integration and angiogenesis. Biomater Sci 2021; 9:7297-7310. [PMID: 34617526 PMCID: PMC8547328 DOI: 10.1039/d1bm01262b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Wound healing involves a complex series of events where cell–cell and cell-extracellular matrix (ECM) interactions play a key role. Wounding can be simple, such as the loss of the epithelial integrity, or deeper and more complex, reaching to subcutaneous tissues, including blood vessels, muscles and nerves. Rapid neovascularisation of the wounded area is crucial for wound healing as it has a key role in supplying oxygen and nutrients during the highly demanding proliferative phase and transmigration of inflammatory cells to the wound area. One approach to circumvent delayed neovascularisation is the exogenous use of pro-angiogenic factors, which is expensive, highly dose-dependent, and the delivery of them requires a very well-controlled system to avoid leaky, highly permeable and haemorrhagic blood vessel formation. In this study, we decorated polycaprolactone (PCL)-based polymerised high internal phase emulsion (PolyHIPE) scaffolds with fibroblast-derived ECM to assess fibroblast, endothelial cell and keratinocyte activity in vitro and angiogenesis in ex ovo chick chorioallantoic membrane (CAM) assays. Our results showed that the inclusion of ECM in the scaffolds increased the metabolic activity of three types of cells that play a key role in wound healing and stimulated angiogenesis in ex ovo CAM assays over 7 days. Herein, we demonstrated that fibroblast-ECM functionalised PCL PolyHIPE scaffolds appear to have great potential to be used as an active wound dressing to promote angiogenesis and wound healing. Decellularisation of in vitro generated extracellular matrix (ECM) provides an effective way to stimulate angiogenesis and wound healing.![]()
Collapse
Affiliation(s)
- Serkan Dikici
- Department of Bioengineering, Izmir Institute of Technology, Izmir, 35430, Turkey. .,Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield, S3 7HQ, UK.
| | - Betül Aldemir Dikici
- Department of Bioengineering, Izmir Institute of Technology, Izmir, 35430, Turkey. .,Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield, S3 7HQ, UK.
| | - Sheila MacNeil
- Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield, S3 7HQ, UK.
| | - Frederik Claeyssens
- Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield, S3 7HQ, UK.
| |
Collapse
|
23
|
Liu X, Camilleri ET, Li L, Gaihre B, Rezaei A, Park S, Miller Ii AL, Tilton M, Waletzki BE, Terzic A, Elder BD, Yaszemski MJ, Lu L. Injectable catalyst-free "click" organic-inorganic nanohybrid (click-ON) cement for minimally invasive in vivo bone repair. Biomaterials 2021; 276:121014. [PMID: 34280821 PMCID: PMC8916681 DOI: 10.1016/j.biomaterials.2021.121014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/20/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022]
Abstract
Injectable polymers have attracted intensive attention in tissue engineering and drug delivery applications. Current injectable polymer systems often require free-radical or heavy-metal initiators and catalysts for the crosslinking process, which may be extremely toxic to the human body. Here, we report a novel polyhedral oligomeric silsesquioxane (POSS) based strain-promoted alkyne-azide cycloaddition (SPAAC) "click" organic-inorganic nanohybrids (click-ON) system that can be click-crosslinked without any toxic initiators or catalysts. The click-ON scaffolds supported excellent adhesion, proliferation, and osteogenesis of stem cells. In vivo evaluation using a rat cranial defect model showed outstanding bone formation with minimum cytotoxicity. Essential osteogenic alkaline phosphatase (ALP) and vascular CD31 marker expression were detected on the defect site, indicating excellent support of in vivo osteogenesis and vascularization. Using salt leaching techniques, an injectable porous click-ON cement was developed to create porous structures and support better in vivo bone regeneration. Beyond defect filling, the click-ON cement also showed promising application for spinal fusion using rabbits as a model. Compared to the current clinically used poly (methyl methacrylate) (PMMA) cement, this click-ON cement showed great advantages of low heat generation, better biocompatibility and biodegradability, and thus has great potential for bone and related tissue engineering applications.
Collapse
Affiliation(s)
- Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Emily T Camilleri
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Linli Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Asghar Rezaei
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sungjo Park
- Department of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - A Lee Miller Ii
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Maryam Tilton
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Brian E Waletzki
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Andre Terzic
- Department of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Benjamin D Elder
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA; Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Michael J Yaszemski
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
24
|
Safdari M, Bibak B, Soltani H, Hashemi J. Recent advancements in decellularized matrix technology for bone tissue engineering. Differentiation 2021; 121:25-34. [PMID: 34454348 DOI: 10.1016/j.diff.2021.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022]
Abstract
The native extracellular matrix (ECM) provides a matrix to hold tissue/organ, defines the cellular fate and function, and retains growth factors. Such a matrix is considered as a most biomimetic scaffold for tissue engineering due to the biochemical and biological components, 3D hierarchical structure, and physicomechanical properties. Several attempts have been performed to decellularize allo- or xeno-graft tissues and used them for bone repairing and regeneration. Decellularized ECM (dECM) technology has been developed to create an in vivo-like microenvironment to promote cell adhesion, growth, and differentiation for tissue repair and regeneration. Decellularization is mediated through physical, chemical, and enzymatic methods. In this review, we describe the recent progress in bone decellularization and their applications as a scaffold, hydrogel, bioink, or particles in bone tissue engineering. Furthermore, we address the native dECM limitations and the potential of non-bone dECM, cell-based ECM, and engineered ECM (eECM) for in vitro osteogenic differentiation and in vivo bone regeneration.
Collapse
Affiliation(s)
- Mohammadreza Safdari
- Department of Surgery, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Bahram Bibak
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Research Center of Natural Products Safety and Medicinal Plants, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hoseinali Soltani
- Department of General Surgery, Imam Ali Hospital, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Javad Hashemi
- Research Center of Natural Products Safety and Medicinal Plants, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| |
Collapse
|
25
|
Dziedzic DSM, Mogharbel BF, Irioda AC, Stricker PEF, Perussolo MC, Franco CRC, Chang HW, Abdelwahid E, de Carvalho KAT. Adipose-Derived Stromal Cells and Mineralized Extracellular Matrix Delivery by a Human Decellularized Amniotic Membrane in Periodontal Tissue Engineering. MEMBRANES 2021; 11:membranes11080606. [PMID: 34436369 PMCID: PMC8401540 DOI: 10.3390/membranes11080606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 12/21/2022]
Abstract
Periodontitis is a prevalent disease characterized by the loss of periodontal supporting tissues, bone, periodontal ligament, and cementum. The application of a bone tissue engineering strategy with Decellularized Human Amniotic Membrane (DAM) with adipose-derived stromal cells (ASCs) has shown to be convenient and valuable. This study aims to investigate the treatments of a rat periodontal furcation defect model with DAM, ASCs, and a mineralized extracellular matrix (ECM). Rat ASCs were expanded, cultivated on DAM, and with a bone differentiation medium for four weeks, deposited ECM on DAM. Periodontal healing for four weeks was evaluated by micro-computed tomography and histological analysis after treatments with DAM, ASCs, and ECM and compared to untreated defects on five consecutive horizontal levels, from gingival to apical. The results demonstrate that DAM preserves its structure during cultivation and healing periods, supporting cell attachment, permeation, bone deposition on DAM, and periodontal regeneration. DAM and DAM+ASCs enhance bone healing compared to the control on the gingival level. In conclusion, DAM with ASC or without cells and the ECM ensures bone tissue healing. The membrane supported neovascularization and promoted osteoconduction.
Collapse
Affiliation(s)
- Dilcele Silva Moreira Dziedzic
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba, Paraná 80250-060, Brazil; (D.S.M.D.); (B.F.M.); (A.C.I.); (P.E.F.S.); (M.C.P.)
| | - Bassam Felipe Mogharbel
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba, Paraná 80250-060, Brazil; (D.S.M.D.); (B.F.M.); (A.C.I.); (P.E.F.S.); (M.C.P.)
| | - Ana Carolina Irioda
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba, Paraná 80250-060, Brazil; (D.S.M.D.); (B.F.M.); (A.C.I.); (P.E.F.S.); (M.C.P.)
| | - Priscila Elias Ferreira Stricker
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba, Paraná 80250-060, Brazil; (D.S.M.D.); (B.F.M.); (A.C.I.); (P.E.F.S.); (M.C.P.)
| | - Maiara Carolina Perussolo
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba, Paraná 80250-060, Brazil; (D.S.M.D.); (B.F.M.); (A.C.I.); (P.E.F.S.); (M.C.P.)
| | | | - Hsueh-Wen Chang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| | - Eltyeb Abdelwahid
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Katherine Athayde Teixeira de Carvalho
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba, Paraná 80250-060, Brazil; (D.S.M.D.); (B.F.M.); (A.C.I.); (P.E.F.S.); (M.C.P.)
- Correspondence: ; Tel.: +55-41-3310-1719
| |
Collapse
|
26
|
Vascularization Strategies in Bone Tissue Engineering. Cells 2021; 10:cells10071749. [PMID: 34359919 PMCID: PMC8306064 DOI: 10.3390/cells10071749] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Bone is a highly vascularized tissue, and its development, maturation, remodeling, and regeneration are dependent on a tight regulation of blood vessel supply. This condition also has to be taken into consideration in the context of the development of artificial tissue substitutes. In classic tissue engineering, bone-forming cells such as primary osteoblasts or mesenchymal stem cells are introduced into suitable scaffolds and implanted in order to treat critical-size bone defects. However, such tissue substitutes are initially avascular. Because of the occurrence of hypoxic conditions, especially in larger tissue substitutes, this leads to the death of the implanted cells. Therefore, it is necessary to devise vascularization strategies aiming at fast and efficient vascularization of implanted artificial tissues. In this review article, we present and discuss the current vascularization strategies in bone tissue engineering. These are based on the use of angiogenic growth factors, the co-implantation of blood vessel forming cells, the ex vivo microfabrication of blood vessels by means of bioprinting, and surgical methods for creating surgically transferable composite tissues.
Collapse
|
27
|
Effect of 3D Printing Temperature on Bioactivity of Bone Morphogenetic Protein-2 Released from Polymeric Constructs. Ann Biomed Eng 2021; 49:2114-2125. [PMID: 33560466 DOI: 10.1007/s10439-021-02736-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022]
Abstract
Growth factors such as bone morphogenetic protein-2 (BMP-2) are potent tools for tissue engineering. Three-dimensional (3D) printing offers a potential strategy for delivery of BMP-2 from polymeric constructs; however, these biomolecules are sensitive to inactivation by the elevated temperatures commonly employed during extrusion-based 3D printing. Therefore, we aimed to correlate printing temperature to the bioactivity of BMP-2 released from 3D printed constructs composed of a model polymer, poly(propylene fumarate). Following encapsulation of BMP-2 in poly(DL-lactic-co-glycolic acid) particles, growth factor-loaded fibers were fabricated at three different printing temperatures. Resulting constructs underwent 28 days of aqueous degradation for collection of released BMP-2. Supernatants were then assayed for the presence of bioactive BMP-2 using a cellular assay for alkaline phosphatase activity. Cumulative release profiles indicated that BMP-2 released from constructs that were 3D printed at physiologic and intermediate temperatures exhibited comparable total amounts of bioactive BMP-2 release as those encapsulated in non-printed particulate delivery vehicles. Meanwhile, the elevated printing temperature of 90 °C resulted in a decreased amount of total bioactive BMP-2 release from the fibers. These findings elucidate the effects of elevated printing temperatures on BMP-2 bioactivity during extrusion-based 3D printing, and enlighten polymeric material selection for 3D printing with growth factors.
Collapse
|
28
|
Jasuja H, Kar S, Katti DR, Katti K. Perfusion bioreactor enabled fluid-derived shear stress conditions for novel bone metastatic prostate cancer testbed. Biofabrication 2021; 13. [PMID: 33418550 DOI: 10.1088/1758-5090/abd9d6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 01/08/2021] [Indexed: 12/27/2022]
Abstract
Critical understanding of the complex metastatic cascade of prostate cancer is necessary for the development of a therapeutic interventions for treating metastatic prostate cancer. Increasing evidence supports the synergistic role of biochemical and biophysical cues in cancer progression at metastases. The biochemical factors such as cytokines have been extensively studied in relation to prostate cancer progression to the bone; however, the role of shear stress-induced by interstitial fluid around bone extracellular matrix has not been fully explored as a driving factor for prostate cancer metastasis. Shear stress governs various cellular processes, including cell proliferation and migration. Thus, it is essential to understand the impact of fluid-derived shear stress on the aggressiveness of prostate cancer at the metastatic stage. Here, we report development of a three-dimensional (3D) in-vitro dynamic cell culture system to recapitulate the microenvironment of prostate cancer bone metastasis, to understand the cause of modulation in cell response under fluid-derived shear stress. We observed an increased human mesenchymal stem cells (hMSCs) proliferation and differentiation rate under dynamic culture. We observed that hMSCs under static culture form cell agglutinates, whereas under dynamic culture, hMSCs exhibited a directional alignment with broad and flattened morphology. Next, we observed increased expression of mesenchymal to epithelial transition (MET) biomarkers in bone metastasized prostate cancer models as well as large changes in cellular and tumoroid morphologies with shear stress. Evaluation of cell adhesion proteins indicated that the altered cancer cell morphologies resulted from the constant force pulling due to increased E-Cadherin and phosphorylated Focal adhesion kinase (FAK) proteins under shear stress. Collectively, we have successfully developed a 3D in-vitro dynamic model to recapitulate the behavior of bone metastatic prostate cancer under dynamic conditions.
Collapse
Affiliation(s)
- Haneesh Jasuja
- North Dakota State University, 1410 14th Ave N, North Dakota State University, Fargo, North Dakota, 58105, UNITED STATES
| | - Sumanta Kar
- North Dakota State University, 1410 14th Ave N, North Dakota State University, Fargo, North Dakota, 58108-6050, UNITED STATES
| | - Dinesh R Katti
- Department of Civil Engineering, North Dakota State University, 1410 14th Ave N, Fargo, North Dakota, 58108-6050, UNITED STATES
| | - Kalpana Katti
- Department of Civil and Environmental Engineering, North Dakota State University, 1410 14th Ave N, North Dakota State University, Fargo, North Dakota, 58105, UNITED STATES
| |
Collapse
|
29
|
Yamada S, Yassin MA, Schwarz T, Hansmann J, Mustafa K. Induction of osteogenic differentiation of bone marrow stromal cells on 3D polyester-based scaffolds solely by subphysiological fluidic stimulation in a laminar flow bioreactor. J Tissue Eng 2021; 12:20417314211019375. [PMID: 34262684 PMCID: PMC8243246 DOI: 10.1177/20417314211019375] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/04/2021] [Indexed: 01/09/2023] Open
Abstract
The fatal determination of bone marrow mesenchymal stem/stromal cells (BMSC) is closely associated with mechano-environmental factors in addition to biochemical clues. The aim of this study was to induce osteogenesis in the absence of chemical stimuli using a custom-designed laminar flow bioreactor. BMSC were seeded onto synthetic microporous scaffolds and subjected to the subphysiological level of fluid flow for up to 21 days. During the perfusion, cell proliferation was significantly inhibited. There were also morphological changes, with F-actin polymerisation and upregulation of ROCK1. Notably, in BMSC subjected to flow, mRNA expression of osteogenic markers was significantly upregulated and RUNX2 was localised in the nuclei. Further, under perfusion, there was greater deposition of collagen type 1 and calcium onto the scaffolds. The results confirm that an appropriate level of fluid stimuli preconditions BMSC towards the osteoblastic lineage on 3D scaffolds in the absence of chemical stimulation, which highlights the utility of flow bioreactors in bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Mohammed Ahmed Yassin
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Thomas Schwarz
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
| | - Jan Hansmann
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
- Chair of Tissue Engineering and
Regenerative Medicine, University Hospital Würzburg, Germany
- Department Electrical Engineering,
University of Applied Sciences Würzburg-Schweinfurt, Germany
| | - Kamal Mustafa
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| |
Collapse
|
30
|
Zhang Y, Wu D, Zhao X, Pakvasa M, Tucker AB, Luo H, Qin KH, Hu DA, Wang EJ, Li AJ, Zhang M, Mao Y, Sabharwal M, He F, Niu C, Wang H, Huang L, Shi D, Liu Q, Ni N, Fu K, Chen C, Wagstaff W, Reid RR, Athiviraham A, Ho S, Lee MJ, Hynes K, Strelzow J, He TC, El Dafrawy M. Stem Cell-Friendly Scaffold Biomaterials: Applications for Bone Tissue Engineering and Regenerative Medicine. Front Bioeng Biotechnol 2020; 8:598607. [PMID: 33381499 PMCID: PMC7767872 DOI: 10.3389/fbioe.2020.598607] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is a dynamic organ with high regenerative potential and provides essential biological functions in the body, such as providing body mobility and protection of internal organs, regulating hematopoietic cell homeostasis, and serving as important mineral reservoir. Bone defects, which can be caused by trauma, cancer and bone disorders, pose formidable public health burdens. Even though autologous bone grafts, allografts, or xenografts have been used clinically, repairing large bone defects remains as a significant clinical challenge. Bone tissue engineering (BTE) emerged as a promising solution to overcome the limitations of autografts and allografts. Ideal bone tissue engineering is to induce bone regeneration through the synergistic integration of biomaterial scaffolds, bone progenitor cells, and bone-forming factors. Successful stem cell-based BTE requires a combination of abundant mesenchymal progenitors with osteogenic potential, suitable biofactors to drive osteogenic differentiation, and cell-friendly scaffold biomaterials. Thus, the crux of BTE lies within the use of cell-friendly biomaterials as scaffolds to overcome extensive bone defects. In this review, we focus on the biocompatibility and cell-friendly features of commonly used scaffold materials, including inorganic compound-based ceramics, natural polymers, synthetic polymers, decellularized extracellular matrix, and in many cases, composite scaffolds using the above existing biomaterials. It is conceivable that combinations of bioactive materials, progenitor cells, growth factors, functionalization techniques, and biomimetic scaffold designs, along with 3D bioprinting technology, will unleash a new era of complex BTE scaffolds tailored to patient-specific applications.
Collapse
Affiliation(s)
- Yongtao Zhang
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Di Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Xia Zhao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mikhail Pakvasa
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Andrew Blake Tucker
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Huaxiu Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Kevin H. Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Daniel A. Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Eric J. Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Alexander J. Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Meng Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yukun Mao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Maya Sabharwal
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Fang He
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Changchun Niu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Laboratory Diagnostic Medicine, The Affiliated Hospital of the University of Chinese Academy of Sciences, Chongqing General Hospital, Chongqing, China
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Linjuan Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Deyao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Spine Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Kai Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Surgery Section of Plastic and Reconstructive Surgery, The University of Chicago Medical Center, Chicago, IL, United States
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Sherwin Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Kelly Hynes
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mostafa El Dafrawy
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
31
|
Liang J, Li W, Zhuang N, Wen S, Huang S, Lu W, Zhou Y, Liao G, Zhang B, Liu C. Experimental study on bone defect repair by BMSCs combined with a light-sensitive material: g-C 3N 4/rGO. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 32:248-265. [PMID: 32975477 DOI: 10.1080/09205063.2020.1827923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Bone marrow mesenchymal stem cells (BMSCs), as seed cells, have played an important role in bone defect repair. However, efficiently amplifying and inducing BMSCs in vitro or vivo remains an urgent problem to be solved. Electrical stimulation has been beneficial to the proliferation and differentiation of BMSCs, but current electrical stimulation methods have a critical disadvantage in that they usually burn the skin. g-C3N4/rGO, a new photosensitive material, can produce photocurrent under natural light irradiation, thus reducing energy consumption. Our purpose was to explore whether this photocurrent can promote the proliferation and differentiation of BMSCs. g-C3N4/rGO synthesised under high temperature and pressure had negligible cytotoxicity as confirmed by methyl thiazolyl tetrazolium to BMSCs. Better osteogenesis was found in the blue light material group than in the light-shielding material group, exhibited by alizarin red staining, alkaline phosphatase activity, Western-Blot, and RT-qPCR. Animal experiments showed that the bone repair potential of the material group was significantly higher than that of the non-material group. Overall, we conclude that g-C3N4/rGO is a new non-toxic photosensitive material which can rapidly induce BMSCs into osteoblasts, accelerating bone regeneration and providing us with a feasible method of rapid bone repair.
Collapse
Affiliation(s)
- Jie Liang
- Department of Trauma and Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wenhua Li
- Department of Trauma and Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Zhuang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Sini Wen
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shijia Huang
- Department of Trauma and Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weizhi Lu
- Laboratory of Biosafety, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yezhen Zhou
- Laboratory of Biosafety, School of Public Health, Southern Medical University, Guangzhou, China
| | - Gaozu Liao
- School of Environment, South China Normal University, Guangzhou, China
| | - Bao Zhang
- Laboratory of Biosafety, School of Public Health, Southern Medical University, Guangzhou, China
| | - Chenglong Liu
- Department of Trauma and Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
32
|
Soheilmoghaddam M, Padmanabhan H, Cooper-White JJ. Biomimetic cues from poly(lactic-co-glycolic acid)/hydroxyapatite nano-fibrous scaffolds drive osteogenic commitment in human mesenchymal stem cells in the absence of osteogenic factor supplements. Biomater Sci 2020; 8:5677-5689. [PMID: 32915185 DOI: 10.1039/d0bm00946f] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mimicking the complex hierarchical architecture of the 'osteon', the functional unit of cortical bone, from the bottom-up offers the possibility of generating mature bone tissue in tissue engineered bone substitutes. In this work, a modular 'bottom-up' approach has been developed to assemble bone niche-mimicking nanocomposite scaffolds composed of aligned electrospun nanofibers of poly(lactic-co-glycolic acid) (PLGA) encapsulating aligned rod-shape nano-sized hydroxyapatite (nHA). By encoding axial orientation of the nHA within these aligned nanocomposite fibers, significant improvements in mechanical properties, surface roughness, hydrophilicity and in vitro simulated body fluid (SBF) mineral deposition were achieved. Moreover, these hierarchical scaffolds induced robust formation of bone hydroxyapatite and osteoblastic maturation of human bone marrow-derived mesenchymal stem cells (hBMSCs) in growth media that was absent of any soluble osteogenic differentiation factors. The results of this investigation confirm that these tailored, aligned nanocomposite fibers, in the absence of media-bone inductive factors, offer the requisite biophysical and biochemical cues to hBMSCs to promote and support their differentiation into mature osteoblast cells and form early bone-like tissue in vitro.
Collapse
Affiliation(s)
- Mohammad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory (TE&M), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St Lucia, QLD, Australia.
| | | | | |
Collapse
|
33
|
Afflerbach AK, Kiri MD, Detinis T, Maoz BM. Mesenchymal Stem Cells as a Promising Cell Source for Integration in Novel In Vitro Models. Biomolecules 2020; 10:E1306. [PMID: 32927777 PMCID: PMC7565384 DOI: 10.3390/biom10091306] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023] Open
Abstract
The human-relevance of an in vitro model is dependent on two main factors-(i) an appropriate human cell source and (ii) a modeling platform that recapitulates human in vivo conditions. Recent years have brought substantial advancements in both these aspects. In particular, mesenchymal stem cells (MSCs) have emerged as a promising cell source, as these cells can differentiate into multiple cell types, yet do not raise the ethical and practical concerns associated with other types of stem cells. In turn, advanced bioengineered in vitro models such as microfluidics, Organs-on-a-Chip, scaffolds, bioprinting and organoids are bringing researchers ever closer to mimicking complex in vivo environments, thereby overcoming some of the limitations of traditional 2D cell cultures. This review covers each of these advancements separately and discusses how the integration of MSCs into novel in vitro platforms may contribute enormously to clinical and fundamental research.
Collapse
Affiliation(s)
- Ann-Kristin Afflerbach
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
- Faculty of Biosciences, Universität Heidelberg, 69120 Heidelberg, Germany
| | - Mark D. Kiri
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
| | - Tahir Detinis
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
| | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
34
|
Junka R, Yu X. Polymeric nanofibrous scaffolds laden with cell-derived extracellular matrix for bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 113:110981. [PMID: 32487395 PMCID: PMC7292471 DOI: 10.1016/j.msec.2020.110981] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022]
Abstract
Bone tissue engineering aims to alleviate the shortage of available autograft material and the biological/mechanical incompatibility of allografts through fabrication of bioactive synthetic bone graft substitutes. However, these substitute grafting materials have insufficient biological potency that limits their clinical efficacy in regenerating large defects. Extracellular matrix, a natural tissue scaffold laden with biochemical and structural cues regulating cell adhesion and tissue morphogenesis, may be a versatile supplement that can extend its biological functionality to synthetic grafts. Embedding decellularized extracellular matrix (dECM) into synthetic polymers offers a promising strategy to enhance cellular response to synthetic materials, mitigate physical and mechanical limitations of dECMs, and improve clinical utility of synthetic bone grafts. Enriched with dECM biochemical cues, synthetic polymers can be readily fabricated into complex biocomposite grafts that mimic bone structure and stimulate endogenous cells to regenerate bone. In this study, cell-derived dECMs from osteoblast and endothelial cells were incorporated into polycaprolactone (PCL) solutions for electrospinning dual-layer nanofibrous scaffolds with osteogenic and vascular cues. The study examined the bioactivity of dECM scaffolds in osteoblast cultures for cell number, mineral deposits, and osteogenic markers, as well as regeneration of cortical bone defect in a rat femur. Scaffolds with osteoblast dECM had a significantly robust osteoblast proliferation, Alizarin Red staining/concentration, and osteopontin-positive extracellular deposits. Implanted scaffolds increased bone growth in femoral defects, and constructs with both osteogenic and vascular cues significantly improved cortical width. These findings demonstrate the potential to fabricate tailored biomimetic grafts with dECM cues and fibrous architecture for bone applications.
Collapse
Affiliation(s)
- Radoslaw Junka
- Department Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America
| | - Xiaojun Yu
- Department Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America.
| |
Collapse
|
35
|
Liu X, George MN, Park S, Miller Ii AL, Gaihre B, Li L, Waletzki BE, Terzic A, Yaszemski MJ, Lu L. 3D-printed scaffolds with carbon nanotubes for bone tissue engineering: Fast and homogeneous one-step functionalization. Acta Biomater 2020; 111:129-140. [PMID: 32428680 DOI: 10.1016/j.actbio.2020.04.047] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/29/2020] [Accepted: 04/25/2020] [Indexed: 02/03/2023]
Abstract
Three-dimensional (3D) printing is a promising technology for tissue engineering. However, 3D-printing methods are limited in their ability to produce desired microscale features or electrochemical properties in support of robust cell adhesion, proliferation, and differentiation. This study addresses this deficiency by proposing an integrated, one-step, method to increase the cytocompatibility of 3D-printed scaffolds through functionalization leveraging conductive carbon nanotubes (CNTs). To this end, CNTs were first sonicated with water-soluble single-stranded deoxyribonucleic acid (ssDNA) to generate a negatively charged ssDNA@CNT nano-complex. Concomitantly, 3D-printed poly(propylene fumarate) (PPF) scaffolds were ammonolyzed to introduce free amine groups, which can take on a positive surface charge in water. The ssDNA@CNT nano-complex was then applied to 3D-printed scaffolds through a simple one-step coating utilizing electric-static force. This fast and facile functionalization step resulted in a homogenous and non-toxic coating of CNTs to the surface, which significantly improved the adhesion, proliferation, and differentiation of pre-osteoblast cells. In addition, the CNT based conductive coating layer enabled modulation of cell behavior through electrical stimuli (ES) leading to cellular proliferation and osteogenic gene marker expression, including alkaline phosphatase (ALP), osteocalcin (OCN), and osteopontin (OPN). Collectively, these data provide the foundation for a one-step functionalization method for simple, fast, and effective functionalization of 3D printed scaffolds that support enhanced cell adhesion, proliferation, and differentiation, especially when employed in conjunction with ES. STATEMENT OF SIGNIFICANCE: Three-dimensional (3D) printing is a promising technology for tissue engineering. However, 3D-printing methods have limited ability to produce desired features or electrochemical properties in support of robust cell behavior. To address this deficiency, the current study proposed an integrated, one-step method to increase the cytocompatibility of 3D-printed scaffolds through functionalization leveraging conductive carbon nanotubes (CNTs). This fast and facile functionalization resulted in a homogenous and non-toxic coating of CNTs to the surface, which significantly improved the adhesion, proliferation, and differentiation of cells on the 3D-printed scaffolds.
Collapse
Affiliation(s)
- Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew N George
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Sungjo Park
- Department of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - A Lee Miller Ii
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Linli Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Brian E Waletzki
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre Terzic
- Department of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael J Yaszemski
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
36
|
In Vitro Production of Calcified Bone Matrix onto Wool Keratin Scaffolds via Osteogenic Factors and Electromagnetic Stimulus. MATERIALS 2020; 13:ma13143052. [PMID: 32650489 PMCID: PMC7411850 DOI: 10.3390/ma13143052] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/15/2022]
Abstract
Pulsed electromagnetic field (PEMF) has drawn attention as a potential tool to improve the ability of bone biomaterials to integrate into the surrounding tissue. We investigated the effects of PEMF (frequency, 75 Hz; magnetic induction amplitude, 2 mT; pulse duration, 1.3 ms) on human osteoblast-like cells (SAOS-2) seeded onto wool keratin scaffolds in terms of proliferation, differentiation, and production of the calcified bone extracellular matrix. The wool keratin scaffold offered a 3D porous architecture for cell guesting and nutrient diffusion, suggesting its possible use as a filler to repair bone defects. Here, the combined approach of applying a daily PEMF exposure with additional osteogenic factors stimulated the cells to increase both the deposition of bone-related proteins and calcified matrix onto the wool keratin scaffolds. Also, the presence of SAOS-2 cells, or PEMF, or osteogenic factors did not influence the compression behavior or the resilience of keratin scaffolds in wet conditions. Besides, ageing tests revealed that wool keratin scaffolds were very stable and showed a lower degradation rate compared to commercial collagen sponges. It is for these reasons that this tissue engineering strategy, which improves the osteointegration properties of the wool keratin scaffold, may have a promising application for long term support of bone formation in vivo.
Collapse
|
37
|
Park JH, Gillispie GJ, Copus JS, Zhang W, Atala A, Yoo JJ, Yelick PC, Lee SJ. The effect of BMP-mimetic peptide tethering bioinks on the differentiation of dental pulp stem cells (DPSCs) in 3D bioprinted dental constructs. Biofabrication 2020; 12:035029. [PMID: 32428889 PMCID: PMC7641314 DOI: 10.1088/1758-5090/ab9492] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The goal of this study was to use 3D bioprinting technology to create a bioengineered dental construct containing human dental pulp stem cells (hDPSCs). To accomplish this, we first developed a novel bone morphogenetic protein (BMP) peptide-tethering bioink formulation and examined its rheological properties, its printability, and the structural stability of the bioprinted construct. Second, we evaluated the survival and differentiation of hDPSCs in the bioprinted dental construct by measuring cell viability, proliferation, and gene expression, as well as histological and immunofluorescent analyses. Our results showed that the peptide conjugation into the gelatin methacrylate-based bioink formulation was successfully performed. We determined that greater than 50% of the peptides remained in the bioprinted construct after three weeks in vitro cell culture. Human DPSC viability was >90% in the bioprinted constructs immediately after the printing process. Alizarin Red staining showed that the BMP peptide construct group exhibited the highest calcification as compared to the growth medium, osteogenic medium, and non-BMP peptide construct groups. In addition, immunofluorescent and quantitative reverse transcription-polymerase chain reaction analyses showed robust expression of dentin sialophosphoprotein and osteocalcin in the BMP peptide dental constructs. Together, these results strongly suggested that BMP peptide-tethering bioink could accelerate the differentiation of hDPSCs in 3D bioprinted dental constructs.
Collapse
Affiliation(s)
- Ji Hoon Park
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Gregory J. Gillispie
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Joshua S. Copus
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Weibo Zhang
- Department of Orthodontics, Tufts University, Boston MA 02111
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | | | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
38
|
Tang SW, Tong WY, Pang SW, Voelcker NH, Lam YW. Deconstructing, Replicating, and Engineering Tissue Microenvironment for Stem Cell Differentiation. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:540-554. [PMID: 32242476 DOI: 10.1089/ten.teb.2020.0044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One of the most crucial components of regenerative medicine is the controlled differentiation of embryonic or adult stem cells into the desired cell lineage. Although most of the reported protocols of stem cell differentiation involve the use of soluble growth factors, it is increasingly evident that stem cells also undergo differentiation when cultured in the appropriate microenvironment. When cultured in decellularized tissues, for instance, stem cells can recapitulate the morphogenesis and functional specialization of differentiated cell types with speed and efficiency that often surpass the traditional growth factor-driven protocols. This suggests that the tissue microenvironment (TME) provides stem cells with a holistic "instructive niche" that harbors signals for cellular reprogramming. The translation of this into medical applications requires the decoding of these signals, but this has been hampered by the complexity of TME. This problem is often addressed by a reductionist approach, in which cells are exposed to substrates decorated with simple, empirically designed geometries, textures, and chemical compositions ("bottom-up" approach). Although these studies are invaluable in revealing the basic principles of mechanotransduction mechanisms, their physiological relevance is often uncertain. This review examines the recent progress of an alternative, "top-down" approach, in which the TME is treated as a holistic biological entity. This approach is made possible by recent advances in systems biology and fabrication technologies that enable the isolation, characterization, and reconstitution of TME. It is hoped that these new techniques will elucidate the nature of niche signals so that they can be extracted, replicated, and controlled. This review summarizes these emerging techniques and how the data they generated are changing our view on TME. Impact statement This review summarizes the current state of art of the understanding of instructive niche in the field of tissue microenvironment. Not only did we survey the use of different biochemical preparations as stimuli of stem cell differentiation and summarize the recent effort in dissecting the biochemical composition of these preparations, through the application of extracellular matrix (ECM) arrays and proteomics, but we also introduce the use of open-source, high-content immunohistochemistry projects in contributing to the understanding of tissue-specific composition of ECM. We believe this review would be highly useful for our peer researching in the same field. "Mr. Tulkinghorn is always the same… so oddly out of place and yet so perfectly at home." -Charles Dickens, Bleak House.
Collapse
Affiliation(s)
- Sze Wing Tang
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| | - Wing Yin Tong
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Stella W Pang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong
| | - Nicolas H Voelcker
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
39
|
Aldemir Dikici B, Reilly GC, Claeyssens F. Boosting the Osteogenic and Angiogenic Performance of Multiscale Porous Polycaprolactone Scaffolds by In Vitro Generated Extracellular Matrix Decoration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:12510-12524. [PMID: 32100541 PMCID: PMC7146758 DOI: 10.1021/acsami.9b23100] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 02/26/2020] [Indexed: 05/05/2023]
Abstract
Tissue engineering (TE)-based bone grafts are favorable alternatives to autografts and allografts. Both biochemical properties and the architectural features of TE scaffolds are crucial in their design process. Synthetic polymers are attractive biomaterials to be used in the manufacturing of TE scaffolds, due to various advantages, such as being relatively inexpensive, enabling precise reproducibility, possessing tunable mechanical/chemical properties, and ease of processing. However, such scaffolds need modifications to improve their limited interaction with biological tissues. Structurally, multiscale porosity is advantageous over single-scale porosity; therefore, in this study, we have considered two key points in the design of a bone repair material; (i) manufacture of multiscale porous scaffolds made of photocurable polycaprolactone (PCL) by a combination of emulsion templating and three-dimensional (3D) printing and (ii) decoration of these scaffolds with the in vitro generated bone-like extracellular matrix (ECM) to create biohybrid scaffolds that have improved biological performance compared to PCL-only scaffolds. Multiscale porous scaffolds were fabricated, bone cells were cultured on them, and then they were decellularized. The biological performance of these constructs was tested in vitro and in vivo. Mesenchymal progenitors were seeded on PCL-only and biohybrid scaffolds. Cells not only showed improved attachment on biohybrid scaffolds but also exhibited a significantly higher rate of cell growth and osteogenic activity. The chick chorioallantoic membrane (CAM) assay was used to explore the angiogenic potential of the biohybrid scaffolds. The CAM assay indicated that the presence of the in vitro generated ECM on polymeric scaffolds resulted in higher angiogenic potential and a high degree of tissue infiltration. This study demonstrated that multiscale porous biohybrid scaffolds present a promising approach to improve bioactivity, encourage precursors to differentiate into mature bones, and to induce angiogenesis.
Collapse
Affiliation(s)
- Betül Aldemir Dikici
- Department
of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Department
of Materials Science and Engineering, INSIGNEO Institute for In Silico
Medicine, University of Sheffield, The Pam Liversidge Building, Sheffield S1 3JD, United Kingdom
| | - Gwendolen C. Reilly
- Department
of Materials Science and Engineering, INSIGNEO Institute for In Silico
Medicine, University of Sheffield, The Pam Liversidge Building, Sheffield S1 3JD, United Kingdom
| | - Frederik Claeyssens
- Department
of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Department
of Materials Science and Engineering, INSIGNEO Institute for In Silico
Medicine, University of Sheffield, The Pam Liversidge Building, Sheffield S1 3JD, United Kingdom
| |
Collapse
|
40
|
Sharifi F, Atyabi SM, Irani S, Bakhshi H. Bone morphogenic protein-2 immobilization by cold atmospheric plasma to enhance the osteoinductivity of carboxymethyl chitosan-based nanofibers. Carbohydr Polym 2020; 231:115681. [DOI: 10.1016/j.carbpol.2019.115681] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 01/29/2023]
|
41
|
Molla MDS, Katti DR, Iswara J, Venkatesan R, Paulmurugan R, Katti KS. Prostate Cancer Phenotype Influences Bone Mineralization at Metastasis: A Study Using an In Vitro Prostate Cancer Metastasis Testbed. JBMR Plus 2020; 4:e10256. [PMID: 32083238 PMCID: PMC7017885 DOI: 10.1002/jbm4.10256] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/01/2019] [Accepted: 11/13/2019] [Indexed: 12/18/2022] Open
Abstract
In this study, two types of prostate cancer cell lines, highly metastatic PC-3 and low metastatic MDA PCa 2b (PCa) were cultured on bone mimetic scaffolds to recapitulate metastasis to bone. A unique in vitro 3D tumor model that uses a sequential culture (SC) of human mesenchymal stem cells followed by seeding with cancer cells after bone formation was initiated to study the phenotype-specific interaction between prostate cancer cells and bone microenvironment. The PCa cells were observed to be less prolific and less metastatic, and to form multicellular tumoroids in the bone microenvironment, whereas PC-3 cells were more prolific and were highly metastatic, and did not form multicellular tumoroids in the bone microenvironment. The metastatic process exhibited by these two prostate cancer cell lines showed a significant and different effect on bone mineralization and extracellular matrix formation. Excessive bone formation in the presence of PC-3 and significant osteolysis in the presence of PCa were observed, which was also indicated by osteocalcin and MMP-9 expression as measured by ELISA and qRT-PCR. The field emission scanning electron microscopy images revealed that the structure of mineralized collagen in the presence of PC-3 is different than the one observed in healthy bone. All experimental results indicated that both osteolytic and osteoblastic bone lesions can be recapitulated in our tumor testbed model and that different cancer phenotypes have a very different influence on bone at metastasis. The 3D in vitro model presented in this study provides an improved, reproducible, and controllable system that is a useful tool to elucidate osteotropism of prostate cancer cells. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- MD Shahjahan Molla
- Center for Engineered Cancer TestbedsNorth Dakota State UniversityFargoNDUSA
- Department of Civil and Environmental EngineeringNorth Dakota State UniversityFargoNDUSA
- Scintillon InstituteSan DiegoUSA
| | - Dinesh R Katti
- Center for Engineered Cancer TestbedsNorth Dakota State UniversityFargoNDUSA
- Department of Civil and Environmental EngineeringNorth Dakota State UniversityFargoNDUSA
| | - Jairam Iswara
- Department of Urology, Saint Elizabeth's Medical CenterTufts UniversityBostonMAUSA
| | - Renugopalkrishnan Venkatesan
- Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMAUSA
- Center for Life SciencesBoston Children's Hospital, Harvard Medical School, BostonMassachusettsUSA
| | - Ramasamy Paulmurugan
- Department of RadiologyCellular Pathway Imaging Laboratory (CPIL), Stanford University School of MedicinePalo AltoCAUSA
| | - Kalpana S Katti
- Center for Engineered Cancer TestbedsNorth Dakota State UniversityFargoNDUSA
| |
Collapse
|
42
|
Silva JC, Carvalho MS, Udangawa RN, Moura CS, Cabral JMS, L da Silva C, Ferreira FC, Vashishth D, Linhardt RJ. Extracellular matrix decorated polycaprolactone scaffolds for improved mesenchymal stem/stromal cell osteogenesis towards a patient-tailored bone tissue engineering approach. J Biomed Mater Res B Appl Biomater 2020; 108:2153-2166. [PMID: 31916699 DOI: 10.1002/jbm.b.34554] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/05/2019] [Accepted: 12/20/2019] [Indexed: 01/20/2023]
Abstract
The clinical demand for tissue-engineered bone is growing due to the increase of non-union fractures and delayed healing in an aging population. Herein, we present a method combining additive manufacturing (AM) techniques with cell-derived extracellular matrix (ECM) to generate structurally well-defined bioactive scaffolds for bone tissue engineering (BTE). In this work, highly porous three-dimensional polycaprolactone (PCL) scaffolds with desired size and architecture were fabricated by fused deposition modeling and subsequently decorated with human mesenchymal stem/stromal cell (MSC)-derived ECM produced in situ. The successful deposition of MSC-derived ECM onto PCL scaffolds (PCL-MSC ECM) was confirmed after decellularization using scanning electron microscopy, elemental analysis, and immunofluorescence. The presence of cell-derived ECM within the PCL scaffolds significantly enhanced MSC attachment and proliferation, with and without osteogenic supplementation. Additionally, under osteogenic induction, PCL-MSC ECM scaffolds promoted significantly higher calcium deposition and elevated relative expression of bone-specific genes, particularly the gene encoding osteopontin, when compared to pristine scaffolds. Overall, our results demonstrated the favorable effects of combining MSC-derived ECM and AM-based scaffolds on the osteogenic differentiation of MSC, resulting from a closer mimicry of the native bone niche. This strategy is highly promising for the development of novel personalized BTE approaches enabling the fabrication of patient defect-tailored scaffolds with enhanced biological performance and osteoinductive properties.
Collapse
Affiliation(s)
- João C Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.,Department of Chemistry and Chemical Biology, Biological Sciences and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - Marta S Carvalho
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.,Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - Ranodhi N Udangawa
- Department of Chemistry and Chemical Biology, Biological Sciences and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - Carla S Moura
- CDRSP-Centre for Rapid and Sustainable Product Development, Polytechnic Institute of Leiria, Rua de Portugal-Zona Industrial, Marinha Grande, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Deepak Vashishth
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Biological Sciences and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York.,Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
43
|
Chahal AS, Schweikle M, Lian AM, Reseland JE, Haugen HJ, Tiainen H. Osteogenic potential of poly(ethylene glycol)-amorphous calcium phosphate composites on human mesenchymal stem cells. J Tissue Eng 2020; 11:2041731420926840. [PMID: 32537121 PMCID: PMC7268109 DOI: 10.1177/2041731420926840] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/22/2020] [Indexed: 01/02/2023] Open
Abstract
Synthetic hydrogel-amorphous calcium phosphate composites are promising candidates to substitute biologically sourced scaffolds for bone repair. While the hydrogel matrix serves as a template for stem cell colonisation, amorphous calcium phosphate s provide mechanical integrity with the potential to stimulate osteogenic differentiation. Here, we utilise composites of poly(ethylene glycol)-based hydrogels and differently stabilised amorphous calcium phosphate to investigate potential effects on attachment and osteogenic differentiation of human mesenchymal stem cells. We found that functionalisation with integrin binding motifs in the form of RGD tripeptide was necessary to allow adhesion of large numbers of cells in spread morphology. Slow dissolution of amorphous calcium phosphate mineral in the scaffolds over at least 21 days was observed, resulting in the release of calcium and zinc ions into the cell culture medium. While we qualitatively observed an increasingly mineralised extracellular matrix along with calcium deposition in the presence of amorphous calcium phosphate-loaded scaffolds, we did not observe significant changes in the expression of selected osteogenic markers.
Collapse
Affiliation(s)
- Aman S Chahal
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway
| | - Manuel Schweikle
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway
| | - Aina-Mari Lian
- Oral Research Laboratory, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway
| | - Janne E Reseland
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway
| | - Håvard J Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway
| | - Hanna Tiainen
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
44
|
Ji X, Yuan X, Ma L, Bi B, Zhu H, Lei Z, Liu W, Pu H, Jiang J, Jiang X, Zhang Y, Xiao J. Mesenchymal stem cell-loaded thermosensitive hydroxypropyl chitin hydrogel combined with a three-dimensional-printed poly(ε-caprolactone) /nano-hydroxyapatite scaffold to repair bone defects via osteogenesis, angiogenesis and immunomodulation. Theranostics 2020; 10:725-740. [PMID: 31903147 PMCID: PMC6929983 DOI: 10.7150/thno.39167] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/08/2019] [Indexed: 12/01/2022] Open
Abstract
Chitin-derived hydrogels are commonly used in bone regeneration because of their high cell compatibility; however, their poor mechanical properties and little knowledge of the interaction between the materials and host cells have limited their practical application. Methods: To evaluate osteoinductivity and enhance the mechanical properties of a newly synthesized thermosensitive hydroxypropyl chitin hydrogel (HPCH), a mesenchymal stem cell (MSC)-encapsulated HPCH was infused into a three-dimensional-printed poly (ε-caprolactone) (PCL)/ nano-hydroxyapatite (nHA) scaffold to form a hybrid scaffold. The mechanical properties and cell compatibility of the scaffold were tested. The interaction between macrophages and scaffold for angiogenesis and osteogenesis were explored in vitro and in vivo. Results: The hybrid scaffold showed improved mechanical properties and high cell viability. When MSCs were encapsulated in HPCH, osteo-differentiation was promoted properly via endochondral ossification. The co-culture experiments showed that the hybrid scaffold facilitated growth factor secretion from macrophages, thus promoting vascularization and osteoinduction. The Transwell culture proved that MSCs modulated the inflammatory response of HPCH. Additionally, subcutaneous implantation of MSC-encapsulated HPCH confirmed M2 activation. In situ evaluation of calvarial defects confirmed that the repair was optimal in the MSC-loaded HPCH + PCL/nHA group. Conclusions: PCL/nHA + HPCH hybrid scaffolds effectively promoted vascularization and osteoinduction via osteogenesis promotion and immunomodulation, which suggests promising applications for bone regeneration.
Collapse
Affiliation(s)
- Xiongfa Ji
- Department of Orthopedics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, PR China
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xi Yuan
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Limin Ma
- Department of Orthopedics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, PR China
| | - Bo Bi
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Hao Zhu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zehua Lei
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenbin Liu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - HongXu Pu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiawei Jiang
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xulin Jiang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, PR China
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
45
|
Raic A, Friedrich F, Kratzer D, Bieback K, Lahann J, Lee-Thedieck C. Potential of electrospun cationic BSA fibers to guide osteogenic MSC differentiation via surface charge and fibrous topography. Sci Rep 2019; 9:20003. [PMID: 31882795 PMCID: PMC6934613 DOI: 10.1038/s41598-019-56508-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 12/12/2019] [Indexed: 01/18/2023] Open
Abstract
Large or complex bone fractures often need clinical treatments for sufficient bone repair. New treatment strategies have pursued the idea of using mesenchymal stromal cells (MSCs) in combination with osteoinductive materials to guide differentiation of MSCs into bone cells ensuring complete bone regeneration. To overcome the challenge of developing such materials, fundamental studies are needed to analyze and understand the MSC behavior on modified surfaces of applicable materials for bone healing. For this purpose, we developed a fibrous scaffold resembling the bone/bone marrow extracellular matrix (ECM) based on protein without addition of synthetic polymers. With this biomimetic in vitro model we identified the fibrous structure as well as the charge of the material to be responsible for its effects on MSC differentiation. Positive charge was introduced via cationization that additionally supported the stability of the scaffold in cell culture, and acted as nucleation point for mineralization during osteogenesis. Furthermore, we revealed enhanced focal adhesion formation and osteogenic differentiation of MSCs cultured on positively charged protein fibers. This pure protein-based and chemically modifiable, fibrous ECM model allows the investigation of MSC behavior on biomimetic materials to unfold new vistas how to direct cells' differentiation for the development of new bone regenerating strategies.
Collapse
Affiliation(s)
- Annamarija Raic
- Leibniz University Hannover, Institute of Cell Biology and Biophysics, Hannover, 30419, Germany
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Eggenstein-Leopoldshafen, 76344, Germany
| | - Frank Friedrich
- Karlsruhe Institute of Technology (KIT), Competence Center for Material Moisture, Eggenstein-Leopoldshafen, 76344, Germany
| | - Domenic Kratzer
- Leibniz University Hannover, Institute of Cell Biology and Biophysics, Hannover, 30419, Germany
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Eggenstein-Leopoldshafen, 76344, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University; German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, 68167, Germany
| | - Joerg Lahann
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Eggenstein-Leopoldshafen, 76344, Germany
- Biointerfaces Institute and Departments of Chemical Engineering, Materials Science and Engineering, Macromolecular Science and Engineering and Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Cornelia Lee-Thedieck
- Leibniz University Hannover, Institute of Cell Biology and Biophysics, Hannover, 30419, Germany.
| |
Collapse
|
46
|
Smith BT, Bittner SM, Watson E, Smoak MM, Diaz-Gomez L, Molina ER, Kim YS, Hudgins CD, Melchiorri AJ, Scott DW, Grande-Allen KJ, Yoo JJ, Atala A, Fisher JP, Mikos AG. Multimaterial Dual Gradient Three-Dimensional Printing for Osteogenic Differentiation and Spatial Segregation. Tissue Eng Part A 2019; 26:239-252. [PMID: 31696784 DOI: 10.1089/ten.tea.2019.0204] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In this study of three-dimensional (3D) printed composite β-tricalcium phosphate (β-TCP)-/hydroxyapatite/poly(ɛ-caprolactone)-based constructs, the effects of vertical compositional ceramic gradients and architectural porosity gradients on the osteogenic differentiation of rabbit bone marrow-derived mesenchymal stem cells (MSCs) were investigated. Specifically, three different concentrations of β-TCP (0, 10, and 20 wt%) and three different porosities (33% ± 4%, 50% ± 4%, and 65% ± 3%) were examined to elucidate the contributions of chemical and physical gradients on the biochemical behavior of MSCs and the mineralized matrix production within a 3D culture system. By delaminating the constructs at the gradient transition point, the spatial separation of cellular phenotypes could be specifically evaluated for each construct section. Results indicated that increased concentrations of β-TCP resulted in upregulation of osteogenic markers, including alkaline phosphatase activity and mineralized matrix development. Furthermore, MSCs located within regions of higher porosity displayed a more mature osteogenic phenotype compared to MSCs in lower porosity regions. These results demonstrate that 3D printing can be leveraged to create multiphasic gradient constructs to precisely direct the development and function of MSCs, leading to a phenotypic gradient. Impact Statement In this study, three-dimensional (3D) printed ceramic/polymeric constructs containing discrete vertical gradients of both composition and porosity were fabricated to precisely control the osteogenic differentiation of mesenchymal stem cells. By making simple alterations in construct architecture and composition, constructs containing heterogenous populations of cells were generated, where gradients in scaffold design led to corresponding gradients in cellular phenotype. The study demonstrates that 3D printed multiphasic composite constructs can be leveraged to create complex heterogeneous tissues and interfaces.
Collapse
Affiliation(s)
- Brandon T Smith
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas.,Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
| | - Sean M Bittner
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas
| | - Emma Watson
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas.,Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
| | - Mollie M Smoak
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas
| | - Luis Diaz-Gomez
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas
| | - Eric R Molina
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas.,Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
| | - Yu Seon Kim
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas
| | - Carrigan D Hudgins
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas
| | - Anthony J Melchiorri
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas
| | - David W Scott
- Department of Statistics, Rice University, Houston, Texas
| | | | - James J Yoo
- NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas.,Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Anthony Atala
- NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas.,Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - John P Fisher
- NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas.,Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, Texas.,Biomaterials Lab, Rice University, Houston, Texas.,NIH/NIBIB Center for Engineering Complex Tissues, Houston, Texas
| |
Collapse
|
47
|
Molina ER, Chim LK, Salazar MC, Koons GL, Menegaz BA, Ruiz-Velasco A, Lamhamedi-Cherradi SE, Vetter AM, Satish T, Cuglievan B, Smoak MM, Scott DW, Ludwig JA, Mikos AG. 3D Tissue-Engineered Tumor Model for Ewing's Sarcoma That Incorporates Bone-like ECM and Mineralization. ACS Biomater Sci Eng 2019; 6:539-552. [PMID: 33463239 DOI: 10.1021/acsbiomaterials.9b01068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The tumor microenvironment harbors essential components required for cancer progression including biochemical signals and mechanical cues. To study the effects of microenvironmental elements on Ewing's sarcoma (ES) pathogenesis, we tissue-engineered an acellular three-dimensional (3D) bone tumor niche from electrospun poly(ε-caprolactone) (PCL) scaffolds that incorporate bone-like architecture, extracellular matrix (ECM), and mineralization. PCL-ECM constructs were generated by decellularizing PCL scaffolds harboring cultures of osteogenic human mesenchymal stem cells. The PCL-ECM constructs simulated in vivo-like tumor architecture and increased the proliferation of ES cells compared to PCL scaffolds alone. Compared to monolayer controls, 3D environments facilitated the downregulation of the canonical insulin-like growth factor 1 receptor (IGF-1R) signal cascade through mechanistic target of rapamycin (mTOR), both of which are targets of recent clinical trials. In addition to the downregulation of canonical IGF-1R signaling, 3D environments promoted a reduction in the clathrin-dependent nuclear localization and transcriptional activity of IGF-1R. In vitro drug testing revealed that 3D environments generated cell phenotypes that were resistant to mTOR inhibition and chemotherapy. Our versatile PCL-ECM constructs allow for the investigation of the roles of various microenvironmental elements in ES tumor growth, cancer cell morphology, and induction of resistant cell phenotypes.
Collapse
Affiliation(s)
| | | | | | | | - Brian A Menegaz
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas 77030, United States
| | - Alejandra Ruiz-Velasco
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas 77030, United States
| | - Salah-Eddine Lamhamedi-Cherradi
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas 77030, United States
| | - Amelia M Vetter
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas 77030, United States
| | | | - Branko Cuglievan
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas 77030, United States
| | | | | | - Joseph A Ludwig
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas 77030, United States
| | | |
Collapse
|
48
|
Kar S, Jasuja H, Katti DR, Katti KS. Wnt/β-Catenin Signaling Pathway Regulates Osteogenesis for Breast Cancer Bone Metastasis: Experiments in an In Vitro Nanoclay Scaffold Cancer Testbed. ACS Biomater Sci Eng 2019; 6:2600-2611. [PMID: 33463270 DOI: 10.1021/acsbiomaterials.9b00923] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Breast cancer shows a high affinity toward bone, causing bone-related complications, leading to a poor clinical prognosis. The Wnt/β-catenin signaling pathway has been well-documented for the bone regenerative process; however, the regulation of the Wnt/β-catenin pathway in breast cancer bone metastasis is poorly explored. Here, we report that the Wnt/β-catenin signaling pathway has a significant effect on osteogenesis during breast cancer bone metastasis. In this study, we have created a 3D in vitro breast cancer bone metastatic microenvironment using nanoclay-based scaffolds along with osteogenically differentiated human mesenchymal stem cells (MSCs) and human breast cancer cells (MCF-7 and MDA-MB-231). The results showed upregulation in expressions of Wnt-related factors (Wnt-5a, β-catenin, AXIN2, and LRP5) in sequential cultures of MSCs with MCF-7 as compared to sequential cultures of MSCs with MDA-MB-231. Sequential cultures of MSCs with MCF-7 also showed higher β-catenin expression on the protein levels than sequential cultures of MSCs with MDA-MB-231. Stimulation of Wnt/β-catenin signaling in sequential cultures of MSCs with MCF-7 by ET-1 resulted in increased bone formation, whereas inactivation of Wnt/β-catenin signaling by DKK-1 displayed a significant decrease in bone formation, mimicking bone lesions in breast cancer patients. These data collectively demonstrate that Wnt/β-catenin signaling governs osteogenesis within the tumor-harboring bone microenvironment, leading to bone metastasis. The nanoclay scaffold provides a unique testbed approach for analysis of the pathways of cancer metastasis.
Collapse
Affiliation(s)
- Sumanta Kar
- Center for Engineered Cancer Test Beds, Materials and Nanotechnology Program, and Department of Civil and Environmental Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Haneesh Jasuja
- Center for Engineered Cancer Test Beds, Materials and Nanotechnology Program, and Department of Civil and Environmental Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Dinesh R Katti
- Center for Engineered Cancer Test Beds, Materials and Nanotechnology Program, and Department of Civil and Environmental Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Kalpana S Katti
- Center for Engineered Cancer Test Beds, Materials and Nanotechnology Program, and Department of Civil and Environmental Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
| |
Collapse
|
49
|
Bracey DN, Jinnah AH, Willey JS, Seyler TM, Hutchinson ID, Whitlock PW, Smith TL, Danelson KA, Emory CL, Kerr BA. Investigating the Osteoinductive Potential of a Decellularized Xenograft Bone Substitute. Cells Tissues Organs 2019; 207:97-113. [PMID: 31655811 PMCID: PMC6935535 DOI: 10.1159/000503280] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
Bone grafting is the second most common tissue transplantation procedure worldwide. One of the alternative methods for bone repair under investigation is a tissue-engineered bone substitute. An ideal property of tissue-engineered bone substitutes is osteoinductivity, defined as the ability to stimulate primitive cells to differentiate into a bone-forming lineage. In the current study, we use a decellularization and oxidation protocol to produce a porcine bone scaffold and examine whether it possesses osteoinductive potential and can be used to create a tissue-engineered bone microenvironment. The decellularization protocol was patented by our lab and consists of chemical decellularization and oxidation steps using combinations of deionized water, trypsin, antimicrobials, peracetic acid, and triton-X100. To test if the bone scaffold was a viable host, preosteoblasts were seeded and analyzed for markers of osteogenic differentiation. The osteoinductive potential was observed in vitro with similar osteogenic markers being expressed in preosteoblasts seeded on the scaffolds and demineralized bone matrix. To assess these properties in vivo, scaffolds with and without preosteoblasts preseeded were subcutaneously implanted in mice for 4 weeks. MicroCT scanning revealed 1.6-fold increased bone volume to total volume ratio and 1.4-fold increase in trabecular thickness in scaffolds after implantation. The histological analysis demonstrates new bone formation and blood vessel formation with pentachrome staining demonstrating osteogenesis and angiogenesis, respectively, within the scaffold. Furthermore, CD31+ staining confirmed the endothelial lining of the blood vessels. These results demonstrate that porcine bone maintains its osteoinductive properties after the application of a patented decellularization and oxidation protocol developed in our laboratory. Future work must be performed to definitively prove osteogenesis of human mesenchymal stem cells, biocompatibility in large animal models, and osteoinduction/osseointegration in a relevant clinical model in vivo. The ability to create a functional bone microenvironment using decellularized xenografts will impact regenerative medicine, orthopedic reconstruction, and could be used in the research of multiple diseases.
Collapse
Affiliation(s)
- Daniel N. Bracey
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Alexander H. Jinnah
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Jeffrey S. Willey
- Wake Forest Baptist Medical Center, Radiation Oncology, Winston Salem, NC, USA
| | | | | | | | - Thomas L. Smith
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Kerry A. Danelson
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Cynthia L. Emory
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Bethany A. Kerr
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
- Virginia Tech-Wake Forest University School for Bioengineering and Sciences, Winston Salem, NC, USA
- Wake Forest School of Medicine, Cancer Biology, Winston Salem, NC, USA
| |
Collapse
|
50
|
Junka R, Quevada K, Yu X. Acellular polycaprolactone scaffolds laden with fibroblast/endothelial cell-derived extracellular matrix for bone regeneration. J Biomed Mater Res A 2019; 108:351-364. [PMID: 31618528 DOI: 10.1002/jbm.a.36821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 10/03/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022]
Abstract
Inconsistencies in graft osteoconduction and osteoinduction present a clinical challenge in regeneration of large bone defects. Deposition of decellularized extracellular matrix (dECM) on tissue engineered scaffolds offers an alternative approach that can enhance these properties by mimicking bone's molecular complexity and direct infiltrating cells to repair damaged bone. However, dECMs derived from homogenous cell populations do not adequately simulate the heterogeneous and vascularized microenvironment of the bone. In this study, successive culture and decellularization of fibroblasts and endothelial cells (ECs) grown on polycaprolactone microfibers was used to develop a bioactive scaffold with heterogeneous dECM mimicking endothelial basement membrane. These scaffolds had greater amount of protein and minimally increased nucleic acid content than scaffolds with homogenous culture dECM. Coomassie Blue and antibody staining revealed extensive tube formation by ECs on fibroblast dECM. Fibroblast/endothelial dECM significantly enhanced osteoblast attachment, alkaline phosphatase activity, and osteocalcin- and osteopontin-positive extracellular mineral deposits. We demonstrated that the osteoconduction of dECMs can be tailored with the appropriate combination of cells to accelerate osteoblast mineral secretion. The overall concept can be expanded to generate increasingly more complex tissue constructs for regeneration of bone defects and other vascularized tissues.
Collapse
Affiliation(s)
- Radoslaw Junka
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| | - Kristian Quevada
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| | - Xiaojun Yu
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| |
Collapse
|