1
|
Lin ECY, Davis MP, Lee MS, Ma G, Xu W, Chang YI, Li WJ. Advancing immunomodulatory functions in mesenchymal stem/stromal cells through targeting the GATA6-mediated pathway. Cytotherapy 2025; 27:85-97. [PMID: 39207345 PMCID: PMC11668624 DOI: 10.1016/j.jcyt.2024.08.001] [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: 12/05/2023] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AIMS The immunomodulatory capacity of mesenchymal stem/stromal cells (MSCs) is a key feature that makes them particularly valuable for regenerative medicine. However, this potential is affected by the chronological aging of the donors and the cell expansion procedures in culture. We have demonstrated that GATA binding protein 6 (GATA6) plays a pivotal role in the aging of MSCs and inhibiting GATA6 rejuvenates the characteristics of MSCs. METHODS In this study, we compared the immunomodulatory capabilities of young and old MSC models, using induced pluripotent stem cells-derived rejuvenated MSCs (rMSCs) and their parental MSCs (pMSCs), respectively, to identify a key mechanism involved in the differential regulation of these capabilities. Additionally, we explored the role of GATA6 in mediating the mechanism. RESULTS Our results demonstrated that rMSCs exhibited downregulated aging-associated regulators, including p53, p21 and GATA6, and showed enhanced suppression of T cell proliferation compared to pMSCs. Through analyzing our previous RNA-seq data and employing target gene knockdown, we determined both suppressors of cytokine signaling 3 (SOCS3) and interleukin 6 were involved in GATA6-induced regulation, collectively affecting the expression of programmed death ligand 1 (PDL1) in both pMSCs and rMSCs. CONCLUSIONS Our findings underline the significance of the GATA6/SOCS3/PDL1 pathway in regulating aging-associated changes in MSC immunomodulatory activity, providing valuable insights into the potential use of rMSCs in the treatment of immune diseases and regenerative medicine.
Collapse
Affiliation(s)
- Eric Chang-Yi Lin
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA; Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Madison P Davis
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ming-Song Lee
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Gui Ma
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yuan-I Chang
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Wan-Ju Li
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
2
|
Han D, Wang W, Gong J, Ma Y, Li Y. Controlled delivery of mesenchymal stem cells via biodegradable scaffolds for fracture healing. Nanomedicine (Lond) 2024:1-18. [PMID: 39686770 DOI: 10.1080/17435889.2024.2439242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Biodegradable controlled delivery systems for mesenchymal stem cells (MSCs) have emerged as novel advancements in the field of regenerative medicine, particularly for accelerating bone fracture healing. This detailed study emphasizes the importance of quick and adequate fracture treatment and the limitations of existing methods. New approaches employing biodegradable scaffolds can be placed within a fracture to serve as a mechanical support and allow controlled release of in situ MSCs and bioactive agents. They are made up of polymers and composites which degrade over time, aiding in natural tissue regrowth. The fabrication methods, including 3D printing, electrospinning, and solvent casting, with particulate leaching that enable precise control over scaffold architecture and properties, are discussed. Progress in controlled drug delivery systems including encapsulation techniques and release kinetics is described, highlighting the potential of such strategies to maintain therapeutic benefits over a prolonged time as well as improving outcomes for fracture repair. MSCs play a role in bone regeneration through differentiation using biodegradable scaffolds, paracrine effects, and regulation of inflammation focusing on fracture healing. Current trends and future directions in scaffold technology and MSC delivery, including smart scaffolds with growth factor incorporation and innovative delivery approaches for fracture healing are also discussed.
Collapse
Affiliation(s)
- Dong Han
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Weijiao Wang
- Otolaryngology Department, Yantaishan Hospital, Yantai, China
| | - Jinpeng Gong
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Yupeng Ma
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Yu Li
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| |
Collapse
|
3
|
Wang W, Tai S, Tao J, Yang L, Cheng X, Zhou J. Innovative hydrogel-based therapies for ischemia-reperfusion injury: bridging the gap between pathophysiology and treatment. Mater Today Bio 2024; 29:101295. [PMID: 39493810 PMCID: PMC11528235 DOI: 10.1016/j.mtbio.2024.101295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/21/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Ischemia-reperfusion injury (IRI) commonly occurs in clinical settings, particularly in medical practices such as organ transplantation, cardiopulmonary resuscitation, and recovery from acute trauma, posing substantial challenges in clinical therapies. Current systemic therapies for IRI are limited by poor drug targeting, short efficacy, and significant side effects. Owing to their exceptional biocompatibility, biodegradability, excellent mechanical properties, targeting capabilities, controlled release potential, and properties mimicking the extracellular matrix (ECM), hydrogels not only serve as superior platforms for therapeutic substance delivery and retention, but also facilitate bioenvironment cultivation and cell recruitment, demonstrating significant potential in IRI treatment. This review explores the pathological processes of IRI and discusses the roles and therapeutic outcomes of various hydrogel systems. By categorizing hydrogel systems into depots delivering therapeutic agents, scaffolds encapsulating mesenchymal stem cells (MSCs), and ECM-mimicking hydrogels, this article emphasizes the selection of polymers and therapeutic substances, and details special crosslinking mechanisms and physicochemical properties, as well as summarizes the application of hydrogel systems for IRI treatment. Furthermore, it evaluates the limitations of current hydrogel treatments and suggests directions for future clinical applications.
Collapse
Affiliation(s)
- Weibo Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Supeng Tai
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Junyue Tao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Lexing Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Xi Cheng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
4
|
Tsadaris SA, Komatsu DE, Grubisic V, Ramos RL, Hadjiargyrou M. A GCaMP reporter mouse with chondrocyte specific expression of a green fluorescent calcium indicator. Bone 2024; 188:117234. [PMID: 39147354 PMCID: PMC11392458 DOI: 10.1016/j.bone.2024.117234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
One of the major processes occurring during the healing of a fractured long bone is chondrogenesis, leading to the formation of the soft callus, which subsequently undergoes endochondral ossification and ultimately bridges the fracture site. Thus, understanding the molecular mechanisms of chondrogenesis can enhance our knowledge of the fracture repair process. One such molecular process is calciun (Ca++) signaling, which is known to play a critical role in the development and regeneration of multiple tissues, including bone, in response to external stimuli. Despite the existence of various mouse models for studying Ca++ signaling, none of them were designed to specifically examine the skeletal system or the various musculoskeletal cell types. As such, we generated a genetically engineered mouse model that is specific to cartilage (crossed with Col2a1 Cre mice) to study chondrocytes. Herein, we report on the characterization of this transgenic mouse line using conditional expression of GCaMP6f, a Ca++-indicator protein. Specifically, this mouse line exhibits increased GCaMP6f fluorescence following Ca++ binding in chondrocytes. Using this model, we show real-time Ca++ signaling in embryos, newborn and adult mice, as well as in fracture calluses. Further, robust expression of GCaMP6f in chondrocytes can be easily detected in embryos, neonates, adults, and fracture callus tissue sections. Finally, we also report on Ca++ signaling pathway gene expression, as well as real-time Ca++ transient measurements in fracture callus chondrocytes. Taken together, these mice provide a new experimental tool to study chondrocyte-specific Ca++ signaling during skeletal development and regeneration, as well as various in vitro perturbations.
Collapse
Affiliation(s)
- Sotirios A Tsadaris
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | - David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, Stony Brook, NY, USA
| | - Vladimir Grubisic
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, USA; Center for Biomedical Innovation, College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Raddy L Ramos
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Michael Hadjiargyrou
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA.
| |
Collapse
|
5
|
Shiwaku Y, Okawa H, Suzuki I, Sakai S, Egusa H, Suzuki O. Induced pluripotent stem cell-derived neural stem cells promote bone formation in mice with calvarial defects. Acta Biomater 2024; 188:93-102. [PMID: 39241820 DOI: 10.1016/j.actbio.2024.08.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/27/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
Nerve-derived factors have attracted attention in bone regeneration therapy due to their ability to promote bone regeneration and nerve innervation. Mesenchymal stem cells transported to target sites promote osteogenesis. However, there are few reports on the effects of neural stem cells on bone regeneration. Therefore, the aim of this study was to investigate the role of neural stem cells in osteogenesis. Here, embryoid bodies (EB) or primary neurospheres (1NS) were generated using mouse induced pluripotent stem cells (iPS cells), which were then seeded onto gelatin (Gel) sponges. The seeded Gel sponges were then transplanted into mouse calvarial bone defects. We noted that 1NS-seeded Gel promoted bone regeneration and the presence of tartrate-resistant acid phosphatase (TRAP)-positive cells, whereas the EB-seeded Gel did not. RNA-sequencing of the 1NS-seeded and EB seeded Gels showed an upregulation of the transforming growth factor (TGF)-β signaling pathway in the 1NS-seeded Gel group. Immunostaining confirmed the presence of Id3 positive cells in mice with bone defects treated with the 1NS-seeded Gel. These findings suggest that the transplantation of neural stem cells may contribute to the promotion of bone regeneration. STATEMENT OF SIGNIFICANCE: This study aimed to investigate whether neural stem cells, when seeded in Gel sponges, promoted bone regeneration. It has been well documented that bone is tightly linked with the nervous systems. Bioscaffolds comprising factors that promote innervation and bone regeneration have been investigated for use in bone therapy. However, there is limited research on the use of neural stem cells for promoting bone formation. To assess this relationship, we conducted both in vivo and in vitro assays to determine whether neural stem cells promoted bone formation. We noted that 1NS-seeded Gel sponges promoted bone formation significantly in mice with calvarial defects after 4 weeks. This study provides a novel approach of neural stem cells for bone therapy.
Collapse
Affiliation(s)
- Yukari Shiwaku
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Hiroko Okawa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai 982-8577, Japan
| | - Susumu Sakai
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Osamu Suzuki
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| |
Collapse
|
6
|
Kolliopoulos V, Tiffany A, Polanek M, Harley BAC. Donor Sex and Passage Conditions Influence MSC Osteogenic Response in Mineralized Collagen Scaffolds. Adv Healthc Mater 2024; 13:e2400039. [PMID: 39036820 PMCID: PMC11518655 DOI: 10.1002/adhm.202400039] [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: 01/04/2024] [Revised: 06/13/2024] [Indexed: 07/23/2024]
Abstract
Contemporary tissue engineering efforts often seek to use mesenchymal stem cells (MSCs) due to their multi-potent potential and ability to generate a pro-regenerative secretome. While many have reported the influence of matrix environment on MSC osteogenic response, few have investigated the effects of donor and sex. Here, a well-defined mineralized collagen scaffold is used to study the influence of passage number and donor-reported sex on MSC proliferation and osteogenic potential. A library of bone marrow and adipose tissue-derived stem cells from eight donors to examine donor viability in osteogenic capacity in mineralized collagen scaffolds is obtained. MSCs displayed reduced proliferative capacity as a function of passage duration. Further, MSCs showed significant sex-associated variability in osteogenic capacity. Notably, MSCs from male donors displayed significantly higher cell proliferation while MSCs from female donors displayed significantly higher osteogenic response via increased alkaline phosphate activity, osteoprotegerin release, and mineral formation in vitro. The study highlights the essentiality of including donor-reported sex as an experimental variable and reporting culture expansion in future studies of biomaterial regenerative potential.
Collapse
Affiliation(s)
- Vasiliki Kolliopoulos
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Aleczandria Tiffany
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Maxwell Polanek
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
7
|
Nguyen Van Q, Akiba Y, Eguchi K, Akiba N, Uoshima K. Controlling redox state by edaravone at transplantation site enhances bone regeneration. Biomed Pharmacother 2024; 177:117032. [PMID: 38941894 DOI: 10.1016/j.biopha.2024.117032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024] Open
Abstract
In cell-based bone augmentation, transplanted cell dysfunction and apoptosis can occur due to oxidative stress caused by the overproduction of reactive oxygen species (ROS). Edaravone (EDA) is a potent free radical scavenger with potential medical applications. This study aimed to investigate the effect of controlling oxidative stress on bone regeneration using EDA. Bone marrow-derived cells were collected from 4-week-old rats, and EDA effects on cell viability and osteogenic differentiation were evaluated. Collagen gels containing PKH26-prelabeled cells were implanted into the calvarial defects of 12-week-old rats, followed by daily subcutaneous injections of normal saline or 500 μM EDA for 4 d. Bone formation was examined using micro-computed tomography and histological staining. Immunofluorescence staining was performed for markers of oxidative stress, macrophages, osteogenesis, and angiogenesis. EDA suppressed ROS production and hydrogen peroxide-induced apoptosis, recovering cell viability and osteoblast differentiation. EDA treatment in vivo increased new bone formation. EDA induced the transition of the macrophage population toward the M2 phenotype. The EDA group also exhibited stronger immunofluorescence for vascular endothelial growth factor and CD31. In addition, more PKH26-positive and PKH26-osteocalcin-double-positive cells were observed in the EDA group, indicating that transplanted cell survival was prolonged, and they differentiated into bone-forming cells. This could be attributed to oxidative stress suppression at the transplantation site by EDA. Collectively, local administration using EDA facilitates bone regeneration by improving the local environment and angiogenesis, prolonging survival, and enhancing the osteogenic capabilities of transplanted cells.
Collapse
Affiliation(s)
- Quang Nguyen Van
- Division of Bio-Prosthodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, 2-5274, Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
| | - Yosuke Akiba
- Division of Bio-Prosthodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, 2-5274, Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan.
| | - Kaori Eguchi
- Division of Bio-Prosthodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, 2-5274, Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
| | - Nami Akiba
- Division of Bio-Prosthodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, 2-5274, Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
| | - Katsumi Uoshima
- Division of Bio-Prosthodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, 2-5274, Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
| |
Collapse
|
8
|
Saberian E, Jenča A, Zafari Y, Jenča A, Petrášová A, Zare-Zardini H, Jenčová J. Scaffold Application for Bone Regeneration with Stem Cells in Dentistry: Literature Review. Cells 2024; 13:1065. [PMID: 38920693 PMCID: PMC11202130 DOI: 10.3390/cells13121065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Bone tissue injuries within oral and dental contexts often present considerable challenges because traditional treatments may not be able to fully restore lost or damaged bone tissue. Novel approaches involving stem cells and targeted 3D scaffolds have been investigated in the search for workable solutions. The use of scaffolds in stem cell-assisted bone regeneration is a crucial component of tissue engineering techniques designed to overcome the drawbacks of traditional bone grafts. This study provides a detailed review of scaffold applications for bone regeneration with stem cells in dentistry. This review focuses on scaffolds and stem cells while covering a broad range of studies explaining bone regeneration in dentistry through the presentation of studies conducted in this field. The role of different stem cells in regenerative medicine is covered in great detail in the reviewed literature. These studies have addressed a wide range of subjects, including the effects of platelet concentrates during dental surgery or specific combinations, such as human dental pulp stem cells with scaffolds for animal model bone regeneration, to promote bone regeneration in animal models. Noting developments, research works consider methods to improve vascularization and explore the use of 3D-printed scaffolds, secretome applications, mesenchymal stem cells, and biomaterials for oral bone tissue regeneration. This thorough assessment outlines possible developments within these crucial regenerative dentistry cycles and provides insights and suggestions for additional study. Furthermore, alternative creative methods for regenerating bone tissue include biophysical stimuli, mechanical stimulation, magnetic field therapy, laser therapy, nutritional supplements and diet, gene therapy, and biomimetic materials. These innovative approaches offer promising avenues for future research and development in the field of bone tissue regeneration in dentistry.
Collapse
Affiliation(s)
- Elham Saberian
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| | - Yaser Zafari
- Lyles School of Civil Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| | - Adriána Petrášová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| | - Hadi Zare-Zardini
- Department of Biomedical Engineering, Meybod University, Meybod 89616-99557, Iran
| | - Janka Jenčová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia
| |
Collapse
|
9
|
Guo T, Pei F, Zhang M, Yamada T, Feng J, Jing J, Ho TV, Chai Y. Vascular architecture regulates mesenchymal stromal cell heterogeneity via P53-PDGF signaling in the mouse incisor. Cell Stem Cell 2024; 31:904-920.e6. [PMID: 38703771 PMCID: PMC11162319 DOI: 10.1016/j.stem.2024.04.011] [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: 08/05/2023] [Revised: 02/17/2024] [Accepted: 04/15/2024] [Indexed: 05/06/2024]
Abstract
Mesenchymal stem cells (MSCs) reside in niches to maintain tissue homeostasis and contribute to repair and regeneration. Although the physiological functions of blood and lymphatic vasculature are well studied, their regulation of MSCs as niche components remains largely unknown. Using adult mouse incisors as a model, we uncover the role of Trp53 in regulating vascular composition through THBS2 to maintain mesenchymal tissue homeostasis. Loss of Trp53 in GLI1+ progeny increases arteries and decreases other vessel types. Platelet-derived growth factors from arteries deposit in the MSC region and interact with PDGFRA and PDGFRB. Significantly, PDGFRA+ and PDGFRB+ cells differentially contribute to defined cell lineages in the adult mouse incisor. Collectively, our results highlight Trp53's importance in regulating the vascular niche for MSCs. They also shed light on how different arterial cells provide unique cues to regulate MSC subpopulations and maintain their heterogeneity. Furthermore, they provide mechanistic insight into MSC-vasculature crosstalk.
Collapse
Affiliation(s)
- Tingwei Guo
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Fei Pei
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Mingyi Zhang
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Takahiko Yamada
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Jifan Feng
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Junjun Jing
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
10
|
Muttanahally KS, Yadav S, Freilich MA, Tadinada A. Does the Outcome of Graft Materials at Dental Implant Sites Differ Between Patients With Normal and Compromised Bone Health? J ORAL IMPLANTOL 2024; 50:238-244. [PMID: 38624039 DOI: 10.1563/aaid-joi-d-23-00078] [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: 04/17/2024]
Abstract
The objective of this paper was to assess the outcome of bone graft material at alveolar bone augmentation sites combined with dental implants in postmenopausal women with compromised bone health by evaluating cone beam computerized tomography (CBCT) scans at multiple time points.CBCT scans were analyzed on 55 postmenopausal women with compromised bone health status to determine the fate of alveolar bone augmentation. CBCT scans were taken immediately after surgery and 9 and 24 months postoperatively. The patient's medication regimens and durations were recorded, and the pixel intensity value (PIV) was measured and standardized using scoring criteria and visual assessment. Statistical analyses included 2-sample t tests for continuous variables and Fisher's exact tests for categorical variables.Among the normal patients, 73% received a grade 2 visual score, and 27% received a grade 1 visual score. After 24 months, 45% of patients received a grade 2 score, and 27% received a grade 3 score. In the osteoporotic group receiving medication, 77% of participants received a grade 1 visual score at the 9-month postoperative evaluation, while 23% received a grade 2 score. At the 24-month assessment, 55% of patients received a grade 1 score, 41% received a grade 2 score, and only 5% received a grade 3 score. Notably, although the graft material did not remodel into native bone, it was a scaffold for implants in controlled osteoporotic patients. The study's results show that the pixel intensity values of particulate graft materials are similar across the three different time points, suggesting that the graft material's pixel intensity value remains constant in postmenopausal women with osteoporosis. The study's limitations include a small sample size and a restricted 24-month follow-up period. This limited time frame may need to capture long-term changes or variations in graft materials adequately. Future research should include a larger sample size and have a longer follow-up duration to provide a more comprehensive understanding of the change in graft materials between patients with normal and compromised bone health.
Collapse
Affiliation(s)
- Kavya Shankar Muttanahally
- Oral and Maxillofacial Radiology, Department of Growth and Development, University of Nebraska Medical Center (UNMC) College of Dentistry, Lincoln, Nebraska, USA
| | - Sumit Yadav
- Department of Growth and Development, UNMC College of Dentistry, Lincoln, Nebraska, USA
| | - Martin A Freilich
- Department of Prosthodontics, University of Connecticut (UConn) School of Dental Medicine, Farmington, Connecticut, USA
| | - Aditya Tadinada
- UConn School of Dental Medicine, Farmington, Connecticut, USA
| |
Collapse
|
11
|
Prasad P, Cancelas JA. From Marrow to Bone and Fat: Exploring the Multifaceted Roles of Leptin Receptor Positive Bone Marrow Mesenchymal Stromal Cells. Cells 2024; 13:910. [PMID: 38891042 PMCID: PMC11171870 DOI: 10.3390/cells13110910] [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: 05/05/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
The bone marrow (BM) stromal cell microenvironment contains non-hematopoietic stromal cells called mesenchymal stromal cells (MSCs). MSCs are plastic adherent, form CFU-Fs, and give rise to osteogenic, adipogenic, chondrogenic progenitors, and most importantly provide HSC niche factor chemokine C-X-C motif ligand 12 (CXCL12) and stem cell factor (SCF). Different authors have defined different markers for mouse MSC identification like PDGFR+Sca-1+ subsets, Nestin+, or LepR+ cells. Of these, the LepR+ cells are the major source of SCF and CXCL12 in the BM microenvironment and play a major role in HSC maintenance and hematopoiesis. LepR+ cells give rise to most of the bones and BM adipocytes, further regulating the microenvironment. In adult BM, LepR+ cells are quiescent but after fracture or irradiation, they proliferate and differentiate into mesenchymal lineage osteogenic, adipogenic and/or chondrogenic cells. They also play a crucial role in the steady-state hematopoiesis process, as well as hematopoietic regeneration and the homing of hematopoietic stem cells (HSCs) after myeloablative injury and/or HSC transplantation. They line the sinusoidal cavities, maintain the trabeculae formation, and provide the space for HSC homing and retention. However, the LepR+ cell subset is heterogeneous; some subsets have higher adipogenic potential, while others express osteollineage-biased genes. Different transcription factors like Early B cell factor 3 (EBF3) or RunX2 help maintain this balance between the self-renewing and committed states, whether osteogenic or adipogenic. The study of LepR+ MSCs holds immense promise for advancing our understanding of HSC biology, tissue regeneration, metabolic disorders, and immune responses. In this review, we will discuss the origin of the BM resident LepR+ cells, different subtypes, and the role of LepR+ cells in maintaining hematopoiesis, osteogenesis, and BM adipogenesis following their multifaceted impact.
Collapse
Affiliation(s)
| | - Jose A. Cancelas
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA;
| |
Collapse
|
12
|
Tateiwa D, Iwamoto M, Kodama J, Ukon Y, Hirai H, Ikuta M, Kitahara T, Furuichi T, Bun M, Otsuru S, Okada S, Kaito T. A synthetic retinoic acid receptor γ antagonist (7C)-loaded nanoparticle enhances bone morphogenetic protein-induced bone regeneration in a rat spinal fusion model. Spine J 2024; 24:899-908. [PMID: 38092193 PMCID: PMC11610515 DOI: 10.1016/j.spinee.2023.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/19/2023] [Accepted: 11/27/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND CONTEXT Bone morphogenetic proteins (BMPs) have potent osteoinductivity and have been applied clinically for challenging musculoskeletal conditions. However, the supraphysiological doses of BMPs used in clinical settings cause various side effects that prevent widespread use, and therefore the BMP dosage needs to be reduced. PURPOSE To address this problem, we synthesized 7C, a retinoic acid receptor γ antagonist-loaded nanoparticle (NP), and investigated its potential application in BMP-based bone regeneration therapy using a rat spinal fusion model. STUDY DESIGN An experimental animal study. METHODS Fifty-three male 8-week-old Sprague-Dawley rats underwent posterolateral spinal fusion and were divided into the following five treatment groups: (1) no recombinant human (rh)BMP-2 and blank-NP (Control), (2) no rhBMP-2 and 1 μg 7C-NP (7C group), (3) low-dose rhBMP-2 (0.5 μg) and 1 μg blank-NP (L-BMP group), (4) low-dose rhBMP-2 (0.5 μg) and 1 μg 7C-NP (L-BMP + 7C group), and (5) high-dose rhBMP-2 (5.0 μg) and 1 μg blank-NP (H-BMP group). Micro-computed tomography and histologic analysis were performed 2 and 6 weeks after the surgery. RESULTS The spinal fusion rates of the Control and 7C groups were both 0%, and those of the L-BMP, L-BMP + 7C, and H-BMP groups were 55.6%, 94.4%, and 100%, respectively. The L-BMP + 7C group markedly promoted cartilaginous tissue formation during BMP-induced endochondral bone formation that resulted in a significantly better spinal fusion rate and bone formation than in the L-BMP group. Although spinal fusion was slower in the L-BMP + 7C group, the L-BMP + 7C group formed a spinal fusion mass with better bone quality than the spinal fusion mass in the H-BMP group. CONCLUSIONS The combined use of 7C-NP with rhBMP-2 in a rat posterolateral lumbar fusion model increased spinal fusion rate and new bone volume without deteriorating the quality of newly formed bone. CLINICAL SIGNIFICANCE 7C-NP potentiates BMP-2-induced bone regeneration and has the potential for efficient bone regeneration with low-dose BMP-2, which can reduce the dose-dependent side effects of BMP-2 in clinical settings.
Collapse
Affiliation(s)
- Daisuke Tateiwa
- Department of Orthopaedic Surgery, Osaka General Medical Center, 3-1-56, Mandaihigashi, Sumiyoshi, Osaka, Japan
| | - Masahiro Iwamoto
- Department of Orthopaedic, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD, USA
| | - Joe Kodama
- Department of Orthopaedic, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD, USA
| | - Yuichiro Ukon
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiromasa Hirai
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masato Ikuta
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kitahara
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takuya Furuichi
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masayuki Bun
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Satoru Otsuru
- Department of Orthopaedic, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD, USA
| | - Seiji Okada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takashi Kaito
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
13
|
Juan C, Bancroft AC, Choi JH, Nunez JH, Pagani CA, Lin YS, Hsiao EC, Levi B. Intersections of Fibrodysplasia Ossificans Progressiva and Traumatic Heterotopic Ossification. Biomolecules 2024; 14:349. [PMID: 38540768 PMCID: PMC10968060 DOI: 10.3390/biom14030349] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 11/11/2024] Open
Abstract
Heterotopic ossification (HO) is a debilitating pathology where ectopic bone develops in areas of soft tissue. HO can develop as a consequence of traumatic insult or as a result of dysregulated osteogenic signaling, as in the case of the orphan disease fibrodysplasia ossificans progressiva (FOP). Traumatic HO (tHO) formation is mediated by the complex interplay of signaling between progenitor, inflammatory, and nerve cells, among others, making it a challenging process to understand. Research into the pathogenesis of genetically mediated HO (gHO) in FOP has established a pathway involving uninhibited activin-like kinase 2 receptor (ALK2) signaling that leads to downstream osteogenesis. Current methods of diagnosis and treatment lag behind pre-mature HO detection and progressive HO accumulation, resulting in irreversible decreases in range of motion and chronic pain for patients. As such, it is necessary to draw on advancements made in the study of tHO and gHO to better diagnose, comprehend, prevent, and treat both.
Collapse
Affiliation(s)
- Conan Juan
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| | - Alec C. Bancroft
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
- Baylor College of Medicine, Houston, TX 77030, USA
| | - Ji Hae Choi
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| | - Johanna H. Nunez
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| | - Chase A. Pagani
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| | - Yen-Sheng Lin
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Edward C. Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine, the Institute for Human Genetics, and the Program in Craniofacial Biology, University of California San Francisco Medical Center, San Francisco, CA 94143, USA;
| | - Benjamin Levi
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| |
Collapse
|
14
|
Hu W, Wang W, Chen Z, Chen Y, Wang Z. Engineered exosomes and composite biomaterials for tissue regeneration. Theranostics 2024; 14:2099-2126. [PMID: 38505616 PMCID: PMC10945329 DOI: 10.7150/thno.93088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/25/2024] [Indexed: 03/21/2024] Open
Abstract
Exosomes, which are small vesicles enclosed by a lipid bilayer and released by many cell types, are widely dispersed and have garnered increased attention in the field of regenerative medicine due to their ability to serve as indicators of diseases and agents with therapeutic potential. Exosomes play a crucial role in mediating intercellular communication through the transfer of many biomolecules, including proteins, lipids, RNA, and other molecular constituents, between cells. The targeted transport of proteins and nucleic acids to specific cells has the potential to enhance or impair specific biological functions. Exosomes have many applications, and they can be used alone or in combination with other therapeutic approaches. The examination of the unique attributes and many functions of these factors has emerged as a prominent field of study in the realm of biomedical research. This manuscript summarizes the origins and properties of exosomes, including their structural, biological, physical, and chemical aspects. This paper offers a complete examination of recent progress in tissue repair and regenerative medicine, emphasizing the possible implications of these methods in forthcoming tissue regeneration attempts.
Collapse
Affiliation(s)
- Weikang Hu
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Ministry of Education Key Laboratory of the Green Preparation and Application for Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Wang Wang
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zesheng Chen
- Ministry of Education Key Laboratory of the Green Preparation and Application for Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Yun Chen
- Department of Biomedical Engineering, Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Zijian Wang
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Ministry of Education Key Laboratory of the Green Preparation and Application for Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
- Department of Biomedical Engineering, Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| |
Collapse
|
15
|
Travnickova M, Filova E, Slepicka P, Slepickova Kasalkova N, Kocourek T, Zaloudkova M, Suchy T, Bacakova L. Titanium-Doped Diamond-like Carbon Layers as a Promising Coating for Joint Replacements Supporting Osteogenic Differentiation of Mesenchymal Stem Cells. Int J Mol Sci 2024; 25:2837. [PMID: 38474083 DOI: 10.3390/ijms25052837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/17/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Diamond-like carbon (DLC) layers are known for their high corrosion and wear resistance, low friction, and high biocompatibility. However, it is often necessary to dope DLC layers with additional chemical elements to strengthen their adhesion to the substrate. Ti-DLC layers (doped with 0.4, 2.1, 3.7, 6.6, and 12.8 at.% of Ti) were prepared by dual pulsed laser deposition, and pure DLC, glass, and polystyrene (PS) were used as controls. In vitro cell-material interactions were investigated with an emphasis on cell adhesion, proliferation, and osteogenic differentiation. We observed slightly increasing roughness and contact angle and decreasing surface free energy on Ti-DLC layers with increasing Ti content. Three-week biological experiments were performed using adipose tissue-derived stem cells (ADSCs) and bone marrow mesenchymal stem cells (bmMSCs) in vitro. The cell proliferation activity was similar or slightly higher on the Ti-doped materials than on glass and PS. Osteogenic cell differentiation on all materials was proved by collagen and osteocalcin production, ALP activity, and Ca deposition. The bmMSCs exhibited greater initial proliferation potential and an earlier onset of osteogenic differentiation than the ADSCs. The ADSCs showed a slightly higher formation of focal adhesions, higher metabolic activity, and Ca deposition with increasing Ti content.
Collapse
Affiliation(s)
- Martina Travnickova
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic
| | - Elena Filova
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic
- Faculty of Materials and Technology, VSB-Technical University of Ostrava, 17. Listopadu 2172/15, 708 00 Ostrava, Czech Republic
| | - Petr Slepicka
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, Czech Republic
| | - Nikola Slepickova Kasalkova
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, Czech Republic
| | - Tomas Kocourek
- Institute of Physics of the Czech Academy of Sciences, Na Slovance 2, 182 21 Prague, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Nam. Sitna 3105, 272 01 Kladno, Czech Republic
| | - Margit Zaloudkova
- Institute of Rock Structure and Mechanics, Czech Academy of Sciences, V Holesovickach 94/41, 182 09 Prague, Czech Republic
| | - Tomas Suchy
- Institute of Rock Structure and Mechanics, Czech Academy of Sciences, V Holesovickach 94/41, 182 09 Prague, Czech Republic
| | - Lucie Bacakova
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic
- Faculty of Materials and Technology, VSB-Technical University of Ostrava, 17. Listopadu 2172/15, 708 00 Ostrava, Czech Republic
| |
Collapse
|
16
|
Tan Y, Sun H, Lan Y, Khan HM, Zhang H, Zhang L, Zhang F, Cui Y, Zhang L, Huang D, Chen X, Zhou C, Sun J, Zhou X. Study on 3D printed MXene-berberine-integrated scaffold for photo-activated antibacterial activity and bone regeneration. J Mater Chem B 2024; 12:2158-2179. [PMID: 38323437 DOI: 10.1039/d3tb02306k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
The repair of mandibular defects is a challenging clinical problem, and associated infections often hinder the treatment, leading to failure in bone regeneration. Herein, a multifunctional platform is designed against the shortages of existing therapies for infected bone deficiency. 2D Ti3C2 MXene and berberine (BBR) are effectively loaded into 3D printing biphasic calcium phosphate (BCP) scaffolds. The prepared composite scaffolds take the feature of the excellent photothermal capacity of Ti3C2 as an antibacterial, mediating NIR-responsive BBR release under laser stimuli. Meanwhile, the sustained release of BBR enhances its antibacterial effect and further accelerates the bone healing process. Importantly, the integration of Ti3C2 improves the mechanical properties of the 3D scaffolds, which are beneficial for new bone formation. Their remarkable biomedical performances in vitro and in vivo present the outstanding antibacterial and osteogenic properties of the Ti3C2-BBR functionalized BCP scaffolds. The synergistic therapy makes it highly promising for repairing infected bone defects and provides insights into a wide range of applications of 2D nanosheets in biomedicine.
Collapse
Affiliation(s)
- Yi Tan
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Huan Sun
- National Engineering Research Centre for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Yuanchen Lan
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Haider Mohammed Khan
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hui Zhang
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Linli Zhang
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Fengying Zhang
- West China Hospital/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Yujia Cui
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Paediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lan Zhang
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Dingming Huang
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xinmei Chen
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Changchun Zhou
- National Engineering Research Centre for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Jianxun Sun
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
17
|
Boscaro D, Sikorski P. Spheroids as a 3D in vitro model to study bone and bone mineralization. BIOMATERIALS ADVANCES 2024; 157:213727. [PMID: 38101067 DOI: 10.1016/j.bioadv.2023.213727] [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: 10/10/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Traumas, fractures, and diseases can severely influence bone tissue. Insight into bone mineralization is essential for the development of therapies and new strategies to enhance bone regeneration. 3D cell culture systems, in particular cellular spheroids, have gained a lot of interest as they can recapitulate crucial aspects of the in vivo tissue microenvironment, such as the extensive cell-cell and cell-extracellular matrix (ECM) interactions found in tissue. The potential of combining spheroids and various classes of biomaterials opens also new opportunities for research within bone tissue engineering. Characterizing cellular organization, ECM structure, and ECM mineralization is a fundamental step for understanding the biological processes involved in bone tissue formation in a spheroid-based model system. Still, many experimental techniques used in this field of research are optimized for use with monolayer cell cultures. There is thus a need to develop new and improving existing experimental techniques, for applications in 3D cell culture systems. In this review, bone composition and spheroids properties are described. This is followed by an insight into the techniques that are currently used in bone spheroids research and how these can be used to study bone mineralization. We discuss the application of staining techniques used with optical and confocal fluorescence microscopy, molecular biology techniques, second harmonic imaging microscopy, Raman spectroscopy and microscopy, as well as electron microscopy-based techniques, to evaluate osteogenic differentiation, collagen production and mineral deposition. Challenges in the applications of these methods in bone regeneration and bone tissue engineering are described. STATEMENT OF SIGNIFICANCE: 3D cell cultures have gained a lot of interest in the last decades as a possible technique that can be used to recreate in vitro in vivo biological process. The importance of 3D environment during bone mineralization led scientists to use this cell culture to study this biological process, to obtain a better understanding of the events involved. New and improved techniques are also required for a proper analysis of this cell model and the process under investigation. This review summarizes the state of the art of the techniques used to study bone mineralization and how 3D cell cultures, in particular spheroids, are tested and analysed to obtain better resolved results related to this complex biological process.
Collapse
Affiliation(s)
- Diamante Boscaro
- Department of Physics, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, Trondheim 7034, Norway.
| | - Pawel Sikorski
- Department of Physics, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, Trondheim 7034, Norway.
| |
Collapse
|
18
|
Paresishvili T, Kakabadze Z. Freeze-Dried Mesenchymal Stem Cells: From Bench to Bedside. Review. Adv Biol (Weinh) 2024; 8:e2300155. [PMID: 37990389 DOI: 10.1002/adbi.202300155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/09/2023] [Indexed: 11/23/2023]
Abstract
This review describes the freeze-dried mesenchymal stem cells (MSCs) and their ability to restore damaged tissues and organs. An analysis of the literature shows that after the lyophilization MSCs retain >80% of paracrine factors and that the mechanism of their action on the restoration of damaged tissues and organs is similar to the mechanism of action of paracrine factors in fresh and cryopreserved mesenchymal stem cells. Based on the own materials, the use of paracrine factors of freeze-dried MSCs in vivo and in vitro for the treatment of various diseases of organs and tissues has shown to be effective. The study also discusses about the advantages and disadvantages of freeze-dried MSCs versus cryopreserved MSCs. However, for the effective use of freeze-dried MSCs in clinical practice, a more detailed study of the mechanism of interaction of paracrine factors of freeze-dried MSCs with target cells and tissues is required. It is also necessary to identify possible other specific paracrine factors of freeze-dried MSCs. In addition, develop new therapeutic strategies for the use of freeze-dried MSCs in regenerative medicine and tissue bioengineering.
Collapse
Affiliation(s)
- Teona Paresishvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, 0186, Georgia
| | - Zurab Kakabadze
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, 0186, Georgia
| |
Collapse
|
19
|
Koukoulias NE, Germanou E, Koukoulias D, Kannas TM, Dimitriadis T. Percutaneous Intramedullary Application of Stem Cells for Fifth Metatarsal Fractures Treated With a Cannulated Screw. Cureus 2024; 16:e55185. [PMID: 38558576 PMCID: PMC10980830 DOI: 10.7759/cureus.55185] [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] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Non-union and refracture of fifth metatarsal fractures are common and devastating complications in the athletic population. Stem cell application at the fracture site, for biologic enhancement, is utilized to address this challenge. We present a simple technique to approach both the endosteum and the periosteum percutaneously, under a local anesthetic, in cases of cannulated screw intramedullary fixation.
Collapse
Affiliation(s)
- Nikolaos E Koukoulias
- Department of Sports Trauma and Orthopaedics, St. Luke's Hospital, Thessaloniki, GRC
| | - Evangelia Germanou
- Department of Physical Education and Sport Science, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | - Dimitris Koukoulias
- Department of Physiotherapy, International Hellenic University, Thessaloniki, GRC
| | - Theodoros M Kannas
- Laboratory of Neuromechanics, Department of Physical Education and Sport Science, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | - Thefilos Dimitriadis
- Department of Sports Trauma and Orthopaedics, St. Luke's Hospital, Thessaloniki, GRC
| |
Collapse
|
20
|
Li S, Yuan Q, Yang M, Long X, Sun J, Yuan X, Liu L, Zhang W, Li Q, Deng Z, Tian R, Xu R, Xie L, Yuan J, He Y, Liu Y, Liu H, Yuan Z. Enhanced cartilage regeneration by icariin and mesenchymal stem cell-derived extracellular vesicles combined in alginate-hyaluronic acid hydrogel. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 55:102723. [PMID: 38007064 DOI: 10.1016/j.nano.2023.102723] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/09/2023] [Accepted: 10/31/2023] [Indexed: 11/27/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is characterized by progressive cartilage degeneration and absence of curative therapies. Therefore, more efficient therapies are compellingly needed. Both mesenchymal stem cells (MSCs)-derived extracellular vesicles (EVs) and Icariin (ICA) are promising for repair of cartilage defect. This study proposes that ICA may be combined to potentiate the cartilage repair capacity of MSC-EVs. MATERIALS AND METHODS MSC-EVs were isolated from sodium alginate (SA) and hyaluronic acid (HA) composite hydrogel (SA-HA) cell spheroid culture. EVs and ICA were combined in SA-HA hydrogel to test therapeutic efficacy on cartilage defect in vivo. RESULTS EVs and ICA were synergistic for promoting both proliferation and migration of MSCs and inflammatory chondrocytes. The combination therapy led to strikingly enhanced repair on cartilage defect in rats, with mechanisms involved in the concomitant modulation of both cartilage degradation and synthesis makers. CONCLUSION The MSC-EVs-ICA/SA-HA hydrogel potentially constitutes a novel therapy for cartilage defect in OA.
Collapse
Affiliation(s)
- Shuyi Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Qian Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Minghui Yang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Xinyi Long
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Jianwu Sun
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Xin Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Lang Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Wanting Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Quanjiang Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Zhujie Deng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Rui Tian
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Renhao Xu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, 510317 Guangzhou, PR China.
| | - Lingna Xie
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Jingna Yuan
- Jinhang Bio-science and Biotechnology Co. Ltd, Guangzhou 510663, PR China.
| | - Yue He
- Jinhang Bio-science and Biotechnology Co. Ltd, Guangzhou 510663, PR China.
| | - Yi Liu
- Orthopedics Department, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China.
| | - Hongmei Liu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, 510317 Guangzhou, PR China.
| | - Zhengqiang Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
21
|
Bouman K, Dittrich ATM, Groothuis JT, van Engelen BGM, Zweers-van Essen H, de Baaij-Daalmeyer A, Janssen MCH, Erasmus CE, Draaisma JMT, Voermans NC. Bone quality in LAMA2-related muscular dystrophy and SELENON-related congenital myopathy, a one-year prospective natural history study. Neuromuscul Disord 2024; 34:105-113. [PMID: 38160563 DOI: 10.1016/j.nmd.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/09/2023] [Accepted: 11/26/2023] [Indexed: 01/03/2024]
Abstract
Fragility fractures are frequently reported in neuromuscular diseases and negatively influence functional prognosis, quality of life and survival. In LAMA2-related muscular dystrophy (LAMA2-MD) and SELENON(SEPN1)-related congenital myopathy (SELENON-RM) cross-sectional and prospective natural history studies on bone quality and fragility long bone fractures (LBFs) are lacking. We therefore aim to systematically assess bone quality and provide recommendations for clinical care. We performed a one-year prospective natural history study in 21 LAMA2-MD and 10 SELENON-RM patients including a standardized fracture history and bone quality assessment through dual energy Xray absorptiometry scan (DEXA-scan) and/or bone health index (BHI). Ninety percent of the LAMA2-MD and SELENON-RM patients showed low bone quality. Eight (38%) LAMA2-MD and five (50%) SELENON-RM patients had a history of fragility LBFs. During the one-year follow-up period, one LAMA2-MD patient (female, 3 years) experienced a fragility LBF of the right humerus. We found no difference in bone mineral density between baseline and one-year follow-up. Based on general international guidelines for osteoporosis, we advise adequate vitamin D and calcium intake, and standardized clinical follow-up through a DEXA-scan or BHI in all LAMA2-MD and SELENON-RM patients. On indication, patients should be referred to the pediatrics or internal medicine for consideration of additional treatments.
Collapse
Affiliation(s)
- Karlijn Bouman
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Pediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Anne T M Dittrich
- Department of Pediatrics, Radboud Institute for Health Sciences, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jan T Groothuis
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Baziel G M van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Heidi Zweers-van Essen
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anja de Baaij-Daalmeyer
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mirian C H Janssen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Corrie E Erasmus
- Department of Pediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jos M T Draaisma
- Department of Pediatrics, Radboud Institute for Health Sciences, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nicol C Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
22
|
Anerillas LO, Wiberg M, Kingham PJ, Kelk P. Platelet lysate for expansion or osteogenic differentiation of bone marrow mesenchymal stem cells for 3D tissue constructs. Regen Ther 2023; 24:298-310. [PMID: 37588134 PMCID: PMC10425714 DOI: 10.1016/j.reth.2023.07.011] [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: 03/07/2023] [Revised: 06/13/2023] [Accepted: 07/26/2023] [Indexed: 08/18/2023] Open
Abstract
Background The use of mesenchymal stem cells (MSCs) for the development of tissue-engineered constructs has advanced in recent years. However, future clinically approved products require following good manufacturing practice (GMP) guidelines. This includes using alternatives to xenogeneic-derived cell culture supplements to avoid rejection of the transplants. Consequently, human platelet lysate (PLT) has been adopted as an affordable and effective alternative to foetal bovine serum (FBS) in traditional 2D cultures. However, little is known about its effect in more advanced 3D culture systems. Methods We evaluated bone marrow MSCs (BMSCs) proliferation and CD marker expression in cells expanded in FBS or PLT-supplemented media. Differentiation capacity of the BMSCs expanded in the presence of the different supplements was evaluated in 3D type I collagen hydrogels. Furthermore, the effects of the supplements on the process of differentiation were analyzed by using qPCR and histological staining. Results Cell proliferation was greater in PLT-supplemented media versus FBS. BMSCs expanded in PLT showed similar osteogenic differentiation capacity in 3D compared with FBS expanded cells. In contrast, when cells were 3D differentiated in PLT they showed lower osteogenesis versus the traditional FBS protocol. This was also the case for adipogenic differentiation, in which FBS supplementation was superior to PLT. Conclusions PLT is a superior alternative to FBS for the expansion of MSCs without compromising their subsequent differentiation capacity in 3D. However, differentiation in PLT is impaired. Thus, PLT can be used to reduce the time required to expand the necessary cell numbers for development of 3D tissue engineered MSC constructs.
Collapse
Affiliation(s)
| | - Mikael Wiberg
- Department of Integrative Medical Biology, Umeå University, 901 87 Umeå, Sweden
- Department of Surgical & Perioperative Sciences, Section for Hand and Plastic Surgery, Umeå University, 901 87 Umeå, Sweden
| | - Paul J. Kingham
- Department of Integrative Medical Biology, Umeå University, 901 87 Umeå, Sweden
| | - Peyman Kelk
- Department of Integrative Medical Biology, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
23
|
Kolliopoulos V, Tiffany A, Polanek M, Harley BAC. DONOR VARIABILITY IN HUMAN MESENCHYMAL STEM CELL OSTEOGENIC RESPONSE AS A FUNCTION OF PASSAGE CONDITIONS AND DONOR SEX. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.12.566781. [PMID: 38014316 PMCID: PMC10680622 DOI: 10.1101/2023.11.12.566781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Contemporary tissue engineering efforts often seek to use mesenchymal stem cells (MSCs) due to their potential to differentiate to various tissue-specific cells and generate a pro-regenerative secretome. While MSC differentiation and therapeutic potential can differ as a function of matrix environment, it may also be widely influenced as a function of donor-to-donor variability. Further, effects of passage number and donor sex may further convolute the identification of clinically effective MSC-mediated regeneration technologies. We report efforts to adapt a well-defined mineralized collagen scaffold platform to study the influence of MSC proliferation and osteogenic potential as a function of passage number and donor sex. Mineralized collagen scaffolds broadly support MSC osteogenic differentiation and regenerative potency in the absence of traditional osteogenic supplements for a wide range of MSCs (rabbit, rat, porcine, human). We obtained a library of bone marrow and adipose tissue derived stem cells to examine donor-variability of regenerative potency in mineralized collagen scaffolds. MSCs displayed reduced proliferative capacity as a function of passage duration. Further, MSCs showed significant sex-based differences. Notably, MSCs from male donors displayed significantly higher metabolic activity and proliferation while MSCs from female donor displayed significantly higher osteogenic response via increased alkaline phosphate activity, osteoprotegerin release, and mineral formation in vitro. Our study highlights the essentiality of considering MSC donor sex and culture expansion in future studies of biomaterial regenerative potential.
Collapse
|
24
|
Baltatu MS, Vizureanu P, Sandu AV, Solcan C, Hritcu LD, Spataru MC. Research Progress of Titanium-Based Alloys for Medical Devices. Biomedicines 2023; 11:2997. [PMID: 38001997 PMCID: PMC10669585 DOI: 10.3390/biomedicines11112997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/16/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
Biomaterials are currently a unique class of materials that are essential to improving the standard of human life and extending it. In the assent of the appearance of biomaterials that contain non-toxic elements, in this study, we examine a system of Ti25Mo7Zr15TaxSi (x = 0, 0.5, 0.75, 1 wt.%) for future medical applications. The alloys were developed in a vacuum electric arc furnace and then studied from a structural, mechanical and in vivo assessment (on rabbits) perspective. The effect of the silicon addition was clearly seen in both the structural and the mechanical characteristics, standing out as beta alloys with a dendritic structure and lowering the mechanical properties as a result of the silicon addition. In experimental rabbits, the proliferation of mesenchymal stem cells was observed in the periosteum and peri-implant area, differentiating into osteoblasts and then into osteocytes. Osteoclasts were discovered within the cartilaginous islands that provide structural support to newly formed bone, playing a primary role in bone remodeling. The newly formed spongy tissue adhered to the fibrous capsule that surrounds the alloy, ensuring good osseointegration of metallic implants. The overexpression of Osteopontin, Metalloproteinase-2 (also known as gelatinase A), and Metallopeptidase-9 (also known as gelatinase B) underscores the processes of osteogenesis, bone mineralization, and normal bone remodeling.
Collapse
Affiliation(s)
- Madalina Simona Baltatu
- Faculty of Materials Science and Engineering, “Gheorghe Asachi” Technical University of Iasi, 41 “D. Mangeron” Street, 700050 Iasi, Romania; (M.S.B.); (A.V.S.)
| | - Petrica Vizureanu
- Faculty of Materials Science and Engineering, “Gheorghe Asachi” Technical University of Iasi, 41 “D. Mangeron” Street, 700050 Iasi, Romania; (M.S.B.); (A.V.S.)
- Technical Sciences Academy of Romania, Dacia Blvd 26, 030167 Bucharest, Romania
| | - Andrei Victor Sandu
- Faculty of Materials Science and Engineering, “Gheorghe Asachi” Technical University of Iasi, 41 “D. Mangeron” Street, 700050 Iasi, Romania; (M.S.B.); (A.V.S.)
- Romanian Inventors Forum, Str. Sf. P. Movila 3, 700089 Iasi, Romania
- Academy of Romanian Scientists, 54 Splaiul Independentei, 050094 Bucharest, Romania
- National Institute for Research and Development in Environmental Protection, 294 Splaiul Independentei, 060031 Bucharest, Romania
| | - Carmen Solcan
- Department of Public Health, Faculty of Veterinary Medicine, Iasi University of Life Sciences, Mihail Sadoveanu Street, No 3, 700490 Iasi, Romania; (L.D.H.); (M.C.S.)
| | - Luminița Diana Hritcu
- Department of Public Health, Faculty of Veterinary Medicine, Iasi University of Life Sciences, Mihail Sadoveanu Street, No 3, 700490 Iasi, Romania; (L.D.H.); (M.C.S.)
| | - Mihaela Claudia Spataru
- Department of Public Health, Faculty of Veterinary Medicine, Iasi University of Life Sciences, Mihail Sadoveanu Street, No 3, 700490 Iasi, Romania; (L.D.H.); (M.C.S.)
| |
Collapse
|
25
|
Khotib J, Marhaeny HD, Miatmoko A, Budiatin AS, Ardianto C, Rahmadi M, Pratama YA, Tahir M. Differentiation of osteoblasts: the links between essential transcription factors. J Biomol Struct Dyn 2023; 41:10257-10276. [PMID: 36420663 DOI: 10.1080/07391102.2022.2148749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/12/2022] [Indexed: 11/27/2022]
Abstract
Osteoblasts, cells derived from mesenchymal stem cells (MSCs) in the bone marrow, are cells responsible for bone formation and remodeling. The differentiation of osteoblasts from MSCs is triggered by the expression of specific genes, which are subsequently controlled by pro-osteogenic pathways. Mature osteoblasts then differentiate into osteocytes and are embedded in the bone matrix. Dysregulation of osteoblast function can cause inadequate bone formation, which leads to the development of bone disease. Various key molecules are involved in the regulation of osteoblastogenesis, which are transcription factors. Previous studies have heavily examined the role of factors that control gene expression during osteoblastogenesis, both in vitro and in vivo. However, the systematic relationship of these transcription factors remains unknown. The involvement of ncRNAs in this mechanism, particularly miRNAs, lncRNAs, and circRNAs, has been shown to influence transcriptional factor activity in the regulation of osteoblast differentiation. Here, we discuss nine essential transcription factors involved in osteoblast differentiation, including Runx2, Osx, Dlx5, β-catenin, ATF4, Ihh, Satb2, and Shn3. In addition, we summarize the role of ncRNAs and their relationship to these essential transcription factors in order to improve our understanding of the transcriptional regulation of osteoblast differentiation. Adequate exploration and understanding of the molecular mechanisms of osteoblastogenesis can be a critical strategy in the development of therapies for bone-related diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Junaidi Khotib
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Honey Dzikri Marhaeny
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Andang Miatmoko
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Aniek Setiya Budiatin
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Mahardian Rahmadi
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Yusuf Alif Pratama
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Muhammad Tahir
- Department of Pharmaceutical Science, Kulliyah of Pharmacy, International Islamic University Malaysia, Pahang, Malaysia
| |
Collapse
|
26
|
Degorska B, Sterna J, Bonecka J, Sobczak-Filipiak M, Jacewicz J. Successful treatment of a benign, non-infected cyst in a dog by bone marrow injections. VET MED-CZECH 2023; 68:337-342. [PMID: 37982124 PMCID: PMC10646538 DOI: 10.17221/19/2023-vetmed] [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: 02/13/2023] [Accepted: 07/19/2023] [Indexed: 11/21/2023] Open
Abstract
Bone cysts are rare orthopaedic problems in dogs. There are no clear treatment guidelines. A young male Shih Tzu was referred to Small Animal Clinic with fifth-degree lameness (5/5) of the left thoracic limb, and with swelling and deformation of the distal humeral region. The radiological assessment revealed an enlargement of the distal brachium and an extensive hypodense osteolytic lesion in the distal metaphyseal region of the humerus. Diagnosis of the bone cyst was formulated and treated with a mini-invasive method using autologous non-concentrated bone marrow injections. The treatment was successful, and at the three weeks, the cyst significantly changed its structure. The follow-up at 12 weeks after the first injection, and at one year revealed complete recovery. To our knowledge, this is the first evidence of a bone cyst in a young dog successfully treated with a minimally-invasive method by using a non-concentrated autologous bone marrow injection.
Collapse
Affiliation(s)
- Beata Degorska
- Department of Small Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences – SGGW, Warsaw, Poland
| | - Jacek Sterna
- Department of Small Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences – SGGW, Warsaw, Poland
| | - Joanna Bonecka
- Department of Small Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences – SGGW, Warsaw, Poland
| | - Malgorzata Sobczak-Filipiak
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences – SGGW, Warsaw, Poland
| | - Jowita Jacewicz
- Department of Small Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences – SGGW, Warsaw, Poland
| |
Collapse
|
27
|
Shou Y, Liu L, Liu Q, Le Z, Lee KL, Li H, Li X, Koh DZ, Wang Y, Liu TM, Yang Z, Lim CT, Cheung C, Tay A. Mechano-responsive hydrogel for direct stem cell manufacturing to therapy. Bioact Mater 2023; 24:387-400. [PMID: 36632503 PMCID: PMC9817177 DOI: 10.1016/j.bioactmat.2022.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 01/04/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cell (MSC) is one of the most actively studied cell types due to its regenerative potential and immunomodulatory properties. Conventional cell expansion methods using 2D tissue culture plates and 2.5D microcarriers in bioreactors can generate large cell numbers, but they compromise stem cell potency and lack mechanical preconditioning to prepare MSC for physiological loading expected in vivo. To overcome these challenges, in this work, we describe a 3D dynamic hydrogel using magneto-stimulation for direct MSC manufacturing to therapy. With our technology, we found that dynamic mechanical stimulation (DMS) enhanced matrix-integrin β1 interactions which induced MSCs spreading and proliferation. In addition, DMS could modulate MSC biofunctions including directing MSC differentiation into specific lineages and boosting paracrine activities (e.g., growth factor secretion) through YAP nuclear localization and FAK-ERK pathway. With our magnetic hydrogel, complex procedures from MSC manufacturing to final clinical use, can be integrated into one single platform, and we believe this 'all-in-one' technology could offer a paradigm shift to existing standards in MSC therapy.
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Ling Liu
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| | - Qimin Liu
- School of Civil Engineering and Architecture, Wuhan University of Technology, 430070, Wuhan, China
| | - Zhicheng Le
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Khang Leng Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
| | - Hua Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 639798, Singapore
| | - Xianlei Li
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Dion Zhanyun Koh
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Yuwen Wang
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Tong Ming Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Zheng Yang
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| |
Collapse
|
28
|
Zhang M, Liu C, Zhao L, Zhang X, Su Y. The Emerging Role of Protein Phosphatase in Regeneration. Life (Basel) 2023; 13:life13051216. [PMID: 37240861 DOI: 10.3390/life13051216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Maintaining normal cellular behavior is essential for the survival of organisms. One of the main mechanisms to control cellular behavior is protein phosphorylation. The process of protein phosphorylation is reversible under the regulation of protein kinases and protein phosphatases. The importance of kinases in numerous cellular processes has been well recognized. In recent years, protein phosphatases have also been demonstrated to function actively and specifically in various cellular processes and thus have gained more and more attention from researchers. In the animal kingdom, regeneration frequently occurs to replace or repair damaged or missing tissues. Emerging evidence has revealed that protein phosphatases are crucial for organ regeneration. In this review, after providing a brief overview of the classification of protein phosphatases and their functions in several representative developmental processes, we highlight the critical roles that protein phosphatases play in organ regeneration by summarizing the most recent research on the function and underlying mechanism of protein phosphatase in the regeneration of the liver, bone, neuron, and heart in vertebrates.
Collapse
Affiliation(s)
- Meiling Zhang
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Chenglin Liu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Long Zhao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Xuejiao Zhang
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Ying Su
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
29
|
Smith N, Shirazi S, Cakouros D, Gronthos S. Impact of Environmental and Epigenetic Changes on Mesenchymal Stem Cells during Aging. Int J Mol Sci 2023; 24:ijms24076499. [PMID: 37047469 PMCID: PMC10095074 DOI: 10.3390/ijms24076499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Many crucial epigenetic changes occur during early skeletal development and throughout life due to aging, disease and are heavily influenced by an individual’s lifestyle. Epigenetics is the study of heritable changes in gene expression as the result of changes in the environment without any mutation in the underlying DNA sequence. The epigenetic profiles of cells are dynamic and mediated by different mechanisms, including histone modifications, non-coding RNA-associated gene silencing and DNA methylation. Given the underlining role of dysfunctional mesenchymal tissues in common age-related skeletal diseases such as osteoporosis and osteoarthritis, investigations into skeletal stem cells or mesenchymal stem cells (MSC) and their functional deregulation during aging has been of great interest and how this is mediated by an evolving epigenetic landscape. The present review describes the recent findings in epigenetic changes of MSCs that effect growth and cell fate determination in the context of aging, diet, exercise and bone-related diseases.
Collapse
Affiliation(s)
- Nicholas Smith
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5001, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Suzanna Shirazi
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5001, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Dimitrios Cakouros
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5001, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
- Correspondence: (D.C.); (S.G.); Tel.: +61-8-8128-4395 (S.G.)
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5001, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
- Correspondence: (D.C.); (S.G.); Tel.: +61-8-8128-4395 (S.G.)
| |
Collapse
|
30
|
Prado-Prone G, Silva-Bermudez P, Rodil SE, Ganjkhani Y, Moradi AR, Méndez FJ, García-Macedo JA, Bazzar M, Almaguer-Flores A. ZnO nanoparticles-modified polycaprolactone-gelatin membranes for guided/bone tissue regeneration, antibacterial and osteogenic differentiation properties. Biomed Phys Eng Express 2023; 9. [PMID: 36821850 DOI: 10.1088/2057-1976/acbe47] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/23/2023] [Indexed: 02/25/2023]
Abstract
Periodontitis is a highly prevalent infectious disease that causes the progressive destruction of the periodontal supporting tissues. If left untreated, it can lead to tooth loss impairing oral function, aesthetics, and the patient's overall quality of life. Guided and Bone Tissue Regeneration (GTR/BTR) are surgical therapies based on the placement of a membrane that prevents epithelial growth into the defect, allowing the periodontal/bone cells (including stem cells) to regenerate or restore the affected tissues. The success of these therapies is commonly affected by the local bacterial colonization of the membrane area and its fast biodegradation, causing postoperative infections and a premature rupture of the membrane limiting the regeneration process. This study presents the antibacterial and osteogenic differentiation properties of polycaprolactone-gelatin (PCL-G) electrospun membranes modified with ZnO nanoparticles (ZnO-NPs). The membranes´ chemical composition, surface roughness, biodegradation, water wettability, and mechanical properties under simulated physiological conditions, were analyzed by the close relationship with their biological properties. The PCL-G membranes modified with 1, 3, and 6% w/w of ZnO-NPs showed a significant reduction in the planktonic and biofilm formation of four clinically relevant bacteria;A. actinomycetemcomitansserotype b, P. gingivalis,E. coli, andS. epidermidis. Additionally, the membranes presented appropriate mechanical properties and biodegradation rates to be potentially used in clinical treatments. Notably, the membranes modified with the lowest concentration of ZnO-NPs (1% w/w) stimulated the production of osteoblast markers and calcium deposits in human bone marrow-derived mesenchymal stem cells (BM-MSC) and were biocompatible to human osteoblasts cells (hFOB). These results suggest that the PCL-G membranes with 1% w/w of ZnO-NPs are high-potential candidates for GTR/BTR treatments, as they were the most effective in terms of better antibacterial effectiveness at a lower NPs-concentration while creating a favorable cellular microenvironment for bone growth.
Collapse
Affiliation(s)
- Gina Prado-Prone
- Facultad de Odontología, División de Estudios de Posgrado e Investigación, Universidad Nacional Autónoma de México. Circuito exterior s/n, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Phaedra Silva-Bermudez
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa; Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra; Av. México Xochimilco No. 289 Col. Arenal de Guadalupe C.P. 14389, Ciudad de México, Mexico
| | - Sandra E Rodil
- Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México; Ciudad Universitaria No. 3000, C.P. 04360, Ciudad de México, Mexico
| | - Yasaman Ganjkhani
- Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran.,Institut für Technische Optik, Universitat Stuttgart, Pfaffenwaldring 9, 70569, Stuttgart, Germany
| | - Ali-Reza Moradi
- Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Franklin J Méndez
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada, CICATA-Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología 1036 Z-1 P 2/2, Atlacholoaya 62790, Xochitepec, Mexico
| | - Jorge A García-Macedo
- Departamento de Estado Sólido, Instituto de Física, Universidad Nacional Autónoma de México; Circuito exterior s/n, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Masoomeh Bazzar
- School of Chemistry, University of East Anglia, Norwich Research Park, NR4 7TJ Norwich, United Kingdom
| | - Argelia Almaguer-Flores
- Facultad de Odontología, División de Estudios de Posgrado e Investigación, Universidad Nacional Autónoma de México. Circuito exterior s/n, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| |
Collapse
|
31
|
Mehta D, Dankert J, Yim N, Leclerc K, Leucht P. Rosiglitazone induces adipogenesis of both marrow and periosteum derived mesenchymal stem cells during endochondral fracture healing. J Orthop Sci 2023; 28:460-467. [PMID: 34879982 PMCID: PMC9167886 DOI: 10.1016/j.jos.2021.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/21/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) afflicts about six percent of the global population, and these patients suffer from a two-fold increased fracture risk. Thiazolidinediones (TZDs), including rosiglitazone, are commonly used medications in T2DM because they have a low incidence of monotherapy failure. It is known that rosiglitazone is associated with secondary osteoporosis, further increasing the fracture risk in an already susceptible population. However, it is not yet understood how rosiglitazone impacts endochondral bone healing after fracture. The aim of this study is to elucidate how rosiglitazone treatment impacts endochondral fracture healing, and how rosiglitazone influences the differentiation of skeletal stem and progenitor cells from the bone marrow and the periosteum. METHODS An in-vivo mouse femur fracture model was employed to evaluate differences in fracture healing between mice treated with and without rosiglitazone chow. Fracture healing was assessed with histology and micro computed tomography (μCT). In-vitro assays utilized isolated mouse bone marrow stromal cells and periosteal cells to investigate how rosiglitazone impacts the osteogenic capability and adipogenicity of these cells. RESULTS The in-vivo mouse femur fracture model showed that fracture callus in mice treated with rosiglitazone had significantly more adipose content than those of control mice that did not receive rosiglitazone. In addition, μCT analysis showed that rosiglitazone treated mice had significantly greater bone volume, but overall greater porosity when compared to control mice. In-vitro experimentation showed significantly less osteogenesis and more adipogenesis in bone marrow derived progenitor cells that were cultured in osteogenic media. In addition, rosiglitazone treatment alone caused significant increases in adipogenesis in both bone marrow and periosteum derived cells. CONCLUSION Rosiglitazone impairs endochondral fracture healing in mice by increasing adipogenesis and decreasing osteogenesis of both bone marrow and periosteum derived skeletal progenitor cells.
Collapse
Affiliation(s)
- Devan Mehta
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| | - John Dankert
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| | - Nury Yim
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| | - Kevin Leclerc
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| | - Philipp Leucht
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| |
Collapse
|
32
|
Shi Y, Liu Y, Zhang B, Li X, Lin J, Yang C. Human Menstrual Blood-Derived Endometrial Stem Cells Promote Functional Recovery by Improving the Inflammatory Microenvironment in a Mouse Spinal Cord Injury Model. Cell Transplant 2023; 32:9636897231154579. [PMID: 36786359 PMCID: PMC9932767 DOI: 10.1177/09636897231154579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Spinal cord injury (SCI) is a traumatic injury of the central nervous system. Because neurons are damaged and difficult to regenerate after SCI, its repair remains challenging. However, recent research on stem cell therapy have favored its use after SCI. In this study, based on the establishment of a mouse SCI model, human menstrual blood-derived endometrial stem cells (MenSCs) were intrathecally injected to explore the role and molecular mechanism of MenSCs in SCI. MenSCs were transplanted following SCI in the animal model, and behavioral evaluations showed that MenSC transplantation improved functional recovery. Therefore, samples were collected after 7 days, and transcriptome sequencing was performed. Gene Ontology (GO) enrichment analysis revealed that SCI is closely related to immune system processes. After transplantation of MenSCs, the immune response was significantly activated. In the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, MenSC transplantation was found to be closely related to Th1, Th2, and Th17 cell differentiation pathways. Neuronal damage and glial cell proliferation and activation in the different groups were detected by fluorescence immunohistochemistry and Western blotting 7 days after SCI. Simultaneously, the activation of different types of microglia was detected and the expression of pro-inflammatory and anti-inflammatory factors was quantitatively analyzed. The results showed that MenSC transplantation and sonic hedgehog (Shh)-induced MenSCs accelerated neuronal recovery at the injured site, inhibited the formation of glial cells and microglial activation at the injured site, inhibited the expression of inflammatory factors, and improved the inflammatory microenvironment to achieve functional recovery of SCI. This study provides an experimental basis for the study of the role and molecular mechanism of MenSCs in SCI repair, and a reference for the role of Shh-induced MenSCs in SCI repair.
Collapse
Affiliation(s)
- Yaping Shi
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China
| | - Yunfei Liu
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China
| | - Bichao Zhang
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China
| | - Xiaoying Li
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China
| | - Juntang Lin
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China,Henan Key Laboratory of Medical Tissue
Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Ciqing Yang
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China,Henan Key Laboratory of Medical Tissue
Regeneration, Xinxiang Medical University, Xinxiang, China,Henan Key Laboratory of
Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University,
Xinxiang, China,Ciqing Yang, Stem Cells and Biotherapy
Engineering Research Center of Henan, National Joint Engineering Laboratory of
Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang
Medical University, Xinxiang 453003, China.
| |
Collapse
|
33
|
Cha KY, Cho W, Park S, Ahn J, Park H, Baek I, Lee M, Lee S, Arai Y, Lee SH. Generation of bioactive MSC-EVs for bone tissue regeneration by tauroursodeoxycholic acid treatment. J Control Release 2023; 354:45-56. [PMID: 36586671 DOI: 10.1016/j.jconrel.2022.12.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 01/02/2023]
Abstract
Extracellular vesicles (EVs) are nano-sized carriers that reflect the parent cell's information and are known to mediate cell-cell communication. In order to overcome the disadvantages of mesenchymal stem cells (MSCs) in cell therapy, such as unexpected differentiation leading to tumorization, immune rejection, and other side effects, EVs derived from MSCs (MSC-EVs) with the tissue regenerative function have been studied as new cell-free therapeutics. However, therapeutic applications of EVs require overcoming several challenges. First, the production efficiency of MSC-EVs should be increased at least as much as the quantity of them are required to their clinical application; second, MSC-EVs needs to show various functionality further, thereby increasing tissue regeneration efficiency. In this study, we treated tauroursodeoxycholic acid (TUDCA), a biological derivative known to regulate cholesterol, to MSCs and investigated whether TUDCA treatment would be able to increase EV production efficiency and tissue regenerative capacity of EVs. Indeed, it appears that TUDCA priming to MSC increases the yield of MSC-EVs >2 times by reducing the cellular cholesterol level in MSCs and increasing the exocytosis-related CAV1 expression. Interestingly, it was found that the EVs derived from TUDCA-primed MSCs (T-EV) contained higher amounts of anti-inflammatory cytokines (IL1RN, IL6, IL10, and IL11) and osteogenic proteins (ALP, RUNX2, BMP2, BMPR1, and BMPR2) than those in control MSC-EVs (C-EV). Besides, it was shown that T-EV not only regulated M1/M2 macrophages differentiation of monocytes, also effectively increased the osteogenic differentiation of MSCs as well as bone tissue regeneration in a bone defect rat model. Based on these results, it is concluded that TUDCA treatment to MSC as a new approach endows EV with high-yield production and functionality. Thus, we strongly believe T-EV would be a powerful therapeutic material for bone tissue regeneration and potentially could be expanded to other types of tissue regeneration for clinical applications.
Collapse
Affiliation(s)
- Kyung-Yup Cha
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Woongjin Cho
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Sunghyun Park
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Jinsung Ahn
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Hyoeun Park
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Inho Baek
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Minju Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Sunjun Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Yoshie Arai
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea.
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, 04620 Seoul, South Korea.
| |
Collapse
|
34
|
Hyväri L, Vanhatupa S, Ojansivu M, Kelloniemi M, Pakarinen TK, Hupa L, Miettinen S. Heat Shock Protein 27 Is Involved in the Bioactive Glass Induced Osteogenic Response of Human Mesenchymal Stem Cells. Cells 2023; 12:cells12020224. [PMID: 36672159 PMCID: PMC9856363 DOI: 10.3390/cells12020224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/15/2022] [Accepted: 12/26/2022] [Indexed: 01/06/2023] Open
Abstract
Bioactive glass (BaG) materials are increasingly used in clinics, but their regulatory mechanisms on osteogenic differentiation remain understudied. In this study, we elucidated the currently unknown role of the p38 MAPK downstream target heat shock protein 27 (HSP27), in the osteogenic commitment of human mesenchymal stem cells (hMSCs), derived from adipose tissue (hASCs) and bone marrow (hBMSCs). Osteogenesis was induced with ionic extract of an experimental BaG in osteogenic medium (OM). Our results showed that BaG OM induced fast osteogenesis of hASCs and hBMSCs, demonstrated by enhanced alkaline phosphatase (ALP) activity, production of extracellular matrix protein collagen type I, and matrix mineralization. BaG OM stimulated early and transient activation of p38/HSP27 signaling by phosphorylation in hMSCs. Inhibition of HSP27 phosphorylation with SB202190 reduced the ALP activity, mineralization, and collagen type I production induced by BaG OM. Furthermore, the reduced pHSP27 protein by SB202190 corresponded to a reduced F-actin intensity of hMSCs. The phosphorylation of HSP27 allowed its co-localization with the cytoskeleton. In terminally differentiated cells, however, pHSP27 was found diffusely in the cytoplasm. This study provides the first evidence that HSP27 is involved in hMSC osteogenesis induced with the ionic dissolution products of BaG. Our results indicate that HSP27 phosphorylation plays a role in the osteogenic commitment of hMSCs, possibly through the interaction with the cytoskeleton.
Collapse
Affiliation(s)
- Laura Hyväri
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Elämänaukio, Kuntokatu 2, 33520 Tampere, Finland
| | - Sari Vanhatupa
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Elämänaukio, Kuntokatu 2, 33520 Tampere, Finland
| | - Miina Ojansivu
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Elämänaukio, Kuntokatu 2, 33520 Tampere, Finland
| | - Minna Kelloniemi
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, Elämänaukio, Kuntokatu 2, 33520 Tampere, Finland
| | - Toni-Karri Pakarinen
- Regea Cell and Tissue Center, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
| | - Leena Hupa
- Johan Gadolin Process Chemistry Centre, Åbo Akademi University, Henrikinkatu 2, 20500 Turku, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Elämänaukio, Kuntokatu 2, 33520 Tampere, Finland
- Correspondence: ; Tel.: +358-40-1901789
| |
Collapse
|
35
|
Barik A, Kirtania MD. In-Vitro and In-Vivo Tracking of Cell-Biomaterial Interaction to Monitor the Process of Bone Regeneration. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
36
|
Mechanical strain induces ex vivo expansion of periosteum. PLoS One 2022; 17:e0279519. [PMID: 36584151 PMCID: PMC9803115 DOI: 10.1371/journal.pone.0279519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/02/2022] [Indexed: 12/31/2022] Open
Abstract
Segmental bone defects present complex clinical challenges. Nonunion, malunion, and infection are common sequalae of autogenous bone grafts, allografts, and synthetic bone implants due to poor incorporation with the patient's bone. The current project explores the osteogenic properties of periosteum to facilitate graft incorporation. As tissue area is a natural limitation of autografting, mechanical strain was implemented to expand the periosteum. Freshly harvested, porcine periosteum was strained at 5 and 10% per day for 10 days with non-strained and free-floating samples serving as controls. Total tissue size, viability and histologic examination revealed that strain increased area to a maximum of 1.6-fold in the 10% daily strain. No change in tissue anatomy or viability via MTT or Ki67 staining and quantification was observed among groups. The osteogenic potential of the mechanical expanded periosteum was then examined in vivo. Human cancellous allografts were wrapped with 10% per day strained, fresh, free-floating, or no porcine periosteum and implanted subcutaneously into female, athymic mice. Tissue was collected at 8- and 16-weeks. Gene expression analysis revealed a significant increase in alkaline phosphatase and osteocalcin in the fresh periosteum group at 8-weeks post implantation compared to all other groups. Values among all groups were similar at week 16. Additionally, histological assessment with H&E and Masson-Goldner Trichrome staining showed that all periosteal groups outperformed the non-periosteal allograft, with fresh periosteum demonstrating the highest levels of new tissue mineralization at the periosteum-bone interface. Overall, mechanical expansion of the periosteum can provide increased area for segmental healing via autograft strategies, though further studies are needed to explore culture methodology to optimize osteogenic potential.
Collapse
|
37
|
Nikolova MP, Apostolova MD. Advances in Multifunctional Bioactive Coatings for Metallic Bone Implants. MATERIALS (BASEL, SWITZERLAND) 2022; 16:183. [PMID: 36614523 PMCID: PMC9821663 DOI: 10.3390/ma16010183] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Abstract
To fix the bone in orthopedics, it is almost always necessary to use implants. Metals provide the needed physical and mechanical properties for load-bearing applications. Although widely used as biomedical materials for the replacement of hard tissue, metallic implants still confront challenges, among which the foremost is their low biocompatibility. Some of them also suffer from excessive wear, low corrosion resistance, infections and shielding stress. To address these issues, various coatings have been applied to enhance their in vitro and in vivo performance. When merged with the beneficial properties of various bio-ceramic or polymer coatings remarkable bioactive, osteogenic, antibacterial, or biodegradable composite implants can be created. In this review, bioactive and high-performance coatings for metallic bone implants are systematically reviewed and their biocompatibility is discussed. Updates in coating materials and formulations for metallic implants, as well as their production routes, have been provided. The ways of improving the bioactive coating performance by incorporating bioactive moieties such as growth factors, osteogenic factors, immunomodulatory factors, antibiotics, or other drugs that are locally released in a controlled manner have also been addressed.
Collapse
Affiliation(s)
- Maria P. Nikolova
- Department of Material Science and Technology, University of Ruse “A. Kanchev”, 8 Studentska Str., 7017 Ruse, Bulgaria
| | - Margarita D. Apostolova
- Medical and Biological Research Lab., “Roumen Tsanev” Institute of Molecular Biology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| |
Collapse
|
38
|
Yüce M, Albayrak E. Hyperthermia-stimulated tonsil-mesenchymal stromal cells suppress hematological cancer cells through downregulation of IL-6. J Cell Biochem 2022; 123:1966-1979. [PMID: 36029519 DOI: 10.1002/jcb.30322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 08/01/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022]
Abstract
There are contradictory reports on the use of mesenchymal stromal cells (MSCs) in cancer therapy. Variable outcomes have been associated with several factors including cancer pathology, experimental procedure, MSC source tissue, and individual genetic differences. It is also known that MSCs exert their therapeutic effects with various paracrine factors released from these cells. The profiles of the factors released from MSCs are altered by heat shock, hypoxia, oxidative stress, starvation or various agents such as inflammatory cytokines, and their therapeutic potential is affected. In this study, the antitumor potential of conditioned media (CM), which contains paracrine factors, of mild hyperthermia-stimulated mesenchymal stromal cells derived from lymphoid organ tonsil tissue (T-MSC) was investigated in comparison with CM obtained from T-MSCs grew under normal culture conditions. CM was obtained from T-MSCs that were successfully isolated from palatine tonsil tissue and characterized. The cytotoxic effect of CM on the growth of hematological cancer cell lines at different concentrations (1:1 and 1:2) was demonstrated by methylthiazoldiphenyl-tetrazolium bromide analysis. In addition, the apoptotic effect of T-MSC-CM treatment was evaluated on the cancer cells using Annexin-V/PI detection method by flow cytometry. The pro/anti-apoptotic and cytokine-related gene expressions were also analyzed by real-time polymerase chain reaction post T-MSC-CM treatment. In conclusion, we demonstrated that the factors released from hyperthermia-stimulated T-MSCs induced apoptosis in hematological cancer cell lines in a dose-dependent manner. Importantly, our results at the transcriptional level support that the factors and cytokines released from hyperthermia-stimulated T-MSC may exert antitumoral effects in cancer cells by downregulation of IL-6 that promotes tumorigenesis. These findings reveal that T-MSC-CM can be a powerful cell-free therapeutical strategy for cancer therapy.
Collapse
Affiliation(s)
- Melek Yüce
- Stem Cell Research & Application Center, Ondokuz Mayıs University, Kurupelit Campus, Atakum/Samsun, Turkey
| | - Esra Albayrak
- Stem Cell Research & Application Center, Ondokuz Mayıs University, Kurupelit Campus, Atakum/Samsun, Turkey
| |
Collapse
|
39
|
Awale G, Kan HM, Laurencin CT, Lo KWH. Molecular Mechanisms Underlying the Short-Term Intervention of Forskolin-Mediated Bone Regeneration. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00285-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
40
|
Kulebyakin K, Tyurin-Kuzmin P, Sozaeva L, Voloshin N, Nikolaev M, Chechekhin V, Vigovskiy M, Sysoeva V, Korchagina E, Naida D, Vorontsova M. Dynamic Balance between PTH1R-Dependent Signal Cascades Determines Its Pro- or Anti-Osteogenic Effects on MSC. Cells 2022; 11:3519. [PMID: 36359914 PMCID: PMC9656268 DOI: 10.3390/cells11213519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 03/14/2024] Open
Abstract
Parathyroid hormone (PTH) is one of the key regulators of calcium and phosphate metabolism in the body, controlling bone metabolism and ion excretion by the kidneys. At present, attempts to use PTH as a therapeutic agent have been associated with side-effects, the nature of which is not always clear and predictable. In addition, it is known that in vivo impairment of PTH post-receptor signaling is associated with atypical differentiation behavior not only of bone cells, but also of connective tissues, including adipose tissue. In this work, we studied the functional responses of multipotent mesenchymal stromal cells (MSCs) to the action of PTH at the level of single cells. We used MSCs isolated from the periosteum and subcutaneous adipose tissue to compare characteristics of cell responses to PTH. We found that the hormone can activate three key responses via its receptor located on the surface of MSCs: single transients of calcium, calcium oscillations, and hormone-activated smooth increase in intracellular calcium. These types of calcium responses led to principally different cellular responses of MSCs. The cAMP-dependent smooth increase of intracellular calcium was associated with pro-osteogenic action of PTH, whereas phospholipase C dependent calcium oscillations led to a decrease in osteogenic differentiation intensity. Different variants of calcium responses are in dynamic equilibrium. Suppression of one type of response leads to increased activation of another type and, accordingly, to a change in the effect of PTH on cell differentiation.
Collapse
Affiliation(s)
- Konstantin Kulebyakin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Pyotr Tyurin-Kuzmin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Leila Sozaeva
- Endocrinology Research Center, 115478 Moscow, Russia
| | - Nikita Voloshin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Mikhail Nikolaev
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Vadim Chechekhin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Maxim Vigovskiy
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Veronika Sysoeva
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | | | - Daria Naida
- Burdenko Main Military Clinical Hospital, 105094 Moscow, Russia
| | - Maria Vorontsova
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 119234 Moscow, Russia
- Endocrinology Research Center, 115478 Moscow, Russia
| |
Collapse
|
41
|
Waqas K, Muller M, Koedam M, El Kadi Y, Zillikens MC, van der Eerden BCJ. Methylglyoxal - an advanced glycation end products (AGEs) precursor - Inhibits differentiation of human MSC-derived osteoblasts in vitro independently of receptor for AGEs (RAGE). Bone 2022; 164:116526. [PMID: 35995334 DOI: 10.1016/j.bone.2022.116526] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/27/2022] [Accepted: 08/16/2022] [Indexed: 11/02/2022]
Abstract
A major precursor of advanced glycation end-products (AGEs) - methylglyoxal (MG) - is a reactive carbonyl metabolite that originates from glycolytic pathways. MG formation and accumulation has been implicated in the pathogenesis of diabetes and age-related chronic musculoskeletal disorders. Human bone marrow-derived stromal cells (BMSCs) are multipotent cells that have the potential to differentiate into cells of mesenchymal origin including osteoblasts, but the role of MG on their differentiation is unclear. We therefore evaluated the effect of MG on proliferation and differentiation of BMSC-derived osteoblasts. Cells were treated with different concentrations of MG (600, 800 and 1000 μM). Cell viability was assessed using a Cell Counting Kit-8 assay. Alkaline phosphatase (ALP) activity and calcium deposition assays were performed to evaluate osteoblast differentiation and mineralization. Gene expression was measured using qRT-PCR, whereas AGE specific receptor (RAGE) and collagen 1 were examined by immunocytochemistry and Western blotting. RAGE knockdown was performed by transducing RAGE specific short hairpin RNAs (shRNAs) using lentivirus. During osteogenic differentiation, MG treatment resulted in reduction of cell viability (27.7 %), ALP activity (45.5 %) and mineralization (82.3 %) compared to untreated cells. MG significantly decreased expression of genes involved in osteogenic differentiation - RUNX2 (2.8 fold), ALPL (3.2 fold), MG detoxification through glyoxalase - GLO1 (3 fold) and collagen metabolism - COL1A1 (4.9 fold), COL1A2 (6.8 fold), LOX (5.4 fold) and PLOD1 (1.7 fold). MG significantly reduced expression of collagen 1 (53.3 %) and RAGE (43.1 %) at protein levels. Co-treatment with a MG scavenger - aminoguanidine - prevented all negative effects of MG. RAGE-specific knockdown during MG treatment did not reverse the effects on cell viability, osteogenic differentiation or collagen metabolism. In conclusion, MG treatment can negatively influence the collagen metabolism and differentiation of BMSCs-derived osteoblasts through a RAGE independent mechanism.
Collapse
Affiliation(s)
- Komal Waqas
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Max Muller
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marijke Koedam
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Youssra El Kadi
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - B C J van der Eerden
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
42
|
Fontelo R, da Costa DS, Reis RL, Novoa-Carballal R, Pashkuleva I. Block copolymer nanopatterns affect cell spreading: Stem versus cancer bone cells. Colloids Surf B Biointerfaces 2022; 219:112774. [PMID: 36067682 DOI: 10.1016/j.colsurfb.2022.112774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022]
Abstract
Bone healing after a tumor removal can be promoted by biomaterials that enhance the bone regeneration and prevent the tumor relapse. Herein, we obtained several nanopatterns by self-assembly of polystyrene-block-poly-(2-vinylpyridine) (PS-b-P2VP) with different molecular weights and investigated the adhesion and morphology of human bone marrow mesenchymal stem cells (BMMSC) and osteosarcoma cell line (SaOS-2) on these patterns aiming to identify topography and chemistry that promote bone healing. We analyzed > 2000 cells per experimental condition using imaging software and different morphometric descriptors, namely area, perimeter, aspect ratio, circularity, surface/area, and fractal dimension of cellular contour (FDC). The obtained data were used as inputs for principal component analysis, which showed distinct response of BMMSC and SaOS-2 to the surface topography and chemistry. Among the studied substrates, micellar nanopatterns assembled from the copolymer with high molecular weight promote the adhesion and spreading of BMMSC and have an opposite effect on SaOS-2. This nanopattern is thus beneficial for bone regeneration after injury or pathology, e.g. bone fracture or tumor removal.
Collapse
Affiliation(s)
- R Fontelo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - D Soares da Costa
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - R L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - R Novoa-Carballal
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - I Pashkuleva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
43
|
Aramini B, Masciale V, Radaelli LFZ, Sgarzani R, Dominici M, Stella F. The sternum reconstruction: Present and future perspectives. Front Oncol 2022; 12:975603. [PMID: 36387077 PMCID: PMC9649912 DOI: 10.3389/fonc.2022.975603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/12/2022] [Indexed: 11/22/2022] Open
Abstract
Sternectomy is a procedure mainly used for removing tumor masses infiltrating the sternum or treating infections. Moreover, the removal of the sternum involves the additional challenge of performing a functional reconstruction. Fortunately, various approaches have been proposed for improving the operation and outcome of reconstruction, including allograft transplantation, using novel materials, and developing innovative surgical approaches, which promise to enhance the quality of life for the patient. This review will highlight the surgical approaches to sternum reconstruction and the new perspectives in the current literature.
Collapse
Affiliation(s)
- Beatrice Aramini
- Division of Thoracic Surgery, Department of Experimental, Diagnostic and Specialty Medicine—DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni—L. Pierantoni Hospital, Forlì, Italy
- *Correspondence: Beatrice Aramini,
| | - Valentina Masciale
- Cell Therapy Laboratory, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Lorenzo Federico Zini Radaelli
- Division of Thoracic Surgery, Department of Experimental, Diagnostic and Specialty Medicine—DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni—L. Pierantoni Hospital, Forlì, Italy
| | - Rossella Sgarzani
- Center of Major Burns, Plastic Surgery Unit, Maurizio Bufalini Hospital, Cesena, Italy
| | - Massimo Dominici
- Cell Therapy Laboratory, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Franco Stella
- Division of Thoracic Surgery, Department of Experimental, Diagnostic and Specialty Medicine—DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni—L. Pierantoni Hospital, Forlì, Italy
| |
Collapse
|
44
|
Celik B, Cicek K, Leal AF, Tomatsu S. Regulation of Molecular Targets in Osteosarcoma Treatment. Int J Mol Sci 2022; 23:12583. [PMID: 36293439 PMCID: PMC9604206 DOI: 10.3390/ijms232012583] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
The most prevalent malignant bone tumor, osteosarcoma, affects the growth plates of long bones in adolescents and young adults. Standard chemotherapeutic methods showed poor response rates in patients with recurrent and metastatic phases. Therefore, it is critical to develop novel and efficient targeted therapies to address relapse cases. In this regard, RNA interference technologies are encouraging options in cancer treatment, in which small interfering RNAs regulate the gene expression following RNA interference pathways. The determination of target tissue is as important as the selection of tissue-specific promoters. Moreover, small interfering RNAs should be delivered effectively into the cytoplasm. Lentiviral vectors could encapsulate and deliver the desired gene into the cell and integrate it into the genome, providing long-term regulation of targeted genes. Silencing overexpressed genes promote the tumor cells to lose invasiveness, prevents their proliferation, and triggers their apoptosis. The uniqueness of cancer cells among patients requires novel therapeutic methods that treat patients based on their unique mutations. Several studies showed the effectiveness of different approaches such as microRNA, drug- or chemotherapy-related methods in treating the disease; however, identifying various targets was challenging to understanding disease progression. In this regard, the patient-specific abnormal gene might be targeted using genomics and molecular advancements such as RNA interference approaches. Here, we review potential therapeutic targets for the RNA interference approach, which is applicable as a therapeutic option for osteosarcoma patients, and we point out how the small interfering RNA method becomes a promising approach for the unmet challenge.
Collapse
Affiliation(s)
- Betul Celik
- Department of Biological Science, University of Delaware, Newark, DE 19716, USA
- Nemours/Alfred I. DuPont Hospital for Children, Wilmington, DE 19803, USA
| | - Kader Cicek
- Department of Biological Science, University of Delaware, Newark, DE 19716, USA
- Nemours/Alfred I. DuPont Hospital for Children, Wilmington, DE 19803, USA
| | - Andrés Felipe Leal
- Nemours/Alfred I. DuPont Hospital for Children, Wilmington, DE 19803, USA
| | - Shunji Tomatsu
- Nemours/Alfred I. DuPont Hospital for Children, Wilmington, DE 19803, USA
| |
Collapse
|
45
|
Jimenez IA, Pool RR, Gabrielson KL. Canine Idiopathic Arteriopathy, Appendicular Bone Infarcts, and Neoplastic Transformation of Bone Infarcts in 108 Dogs ( Canis lupus familiaris). Comp Med 2022; 72:306-319. [PMID: 36113969 PMCID: PMC9827601 DOI: 10.30802/aalas-cm-22-000037] [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: 04/09/2022] [Revised: 05/27/2022] [Accepted: 07/26/2022] [Indexed: 01/27/2023]
Abstract
Osteosarcoma (OSA) is the most common primary bone tumor in both dogs and humans. The dog is an important research model for OSA, yet dogs have much higher prevalence of bone tumors than do humans, a disparity that has yet to be explained. Neoplastic transformation of cells within or adjacent to bone infarcts into primary bone tumors has been described in humans but only sparsely characterized in the veterinary literature. In this study, 653 cases of canine bone infarcts were received through a referral veterinary osteopathology service over a 14-y period. We identified an idiopathic disorder affecting the nutrient artery, termed canine idiopathic arteriopathy (CIA), which to our knowledge has no direct counterpart in human medicine. This disorder was documented alongside ischemic necrosis of the medullary cavity in 114 bone infarcts in 108 dogs. We hypothesize that CIA precipitated an ischemic environment, resulting in development of a bone infarct down- stream of the abnormal artery. In 52% (59 of 114) of cases, bone infarcts demonstrated evidence of repair (termed reparative bone infarcts [RBI]), while in 48% (55 of 114) of infarcts, a bone tumor was also present, including pleomorphic sarcoma, OSA, fibrosarcoma, and chondrosarcoma. In some cases, a spectrum of tumors was present. We hypothesize that the ischemic infarct environment provoked bone marrow mesenchymal stem cells (MSCs) to attempt repair of the stroma, and in approximately half of cases, MSCs underwent neoplastic transformation (BINT) to produce tumors. The most common sites of bone infarcts were the distal femur, distal radius, proximal humerus, and distal tibia, coinciding with common sites of canine OSA. The authors propose that CIA leading to bone infarcts and infarct-derived tumors, in combination with possible underdiagnosis of canine bone infarcts and misdiagnosis of some RBI as neoplasia, may contribute to the higher reported proportion of bone tumors in dogs compared with humans.
Collapse
Affiliation(s)
- Isabel A Jimenez
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland;,
| | - Roy R Pool
- Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine & Biomedical Sciences, College Station, Texas
| | - Kathleen L Gabrielson
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
46
|
Damle A, Sundaresan R, Rajwade JM, Srivastava P, Naik A. A concise review on implications of silver nanoparticles in bone tissue engineering. BIOMATERIALS ADVANCES 2022; 141:213099. [PMID: 36088719 DOI: 10.1016/j.bioadv.2022.213099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 06/15/2023]
Abstract
Skeletal disorders represent a variety of degenerative diseases that affect bone and cartilage homeostasis. The regenerative capacity of bone is affected in osteoporosis, osteoarthritis, rheumatoid arthritis, bone fractures, congenital defects, and bone cancers. There is no viable, non-invasive treatment option and bone regeneration requires surgical intervention with the implantation of bone grafts. Incorporating nanoparticles in bone grafts have improved fracture healing by providing fine structures for bone tissue engineering. It is currently a revolutionary finding in the field of regenerative medicine. Silver nanoparticles (AgNPs) have garnered particular attention due to their well-known anti-microbial and potential osteoinductive properties. In addition, AgNPs have been demonstrated to regulate the proliferation and differentiation of mesenchymal stem cells (MSCs) involved in bone regeneration. Furthermore, AgNPs have shown toxicity towards cancer cells derived from bone. In the last decade, there have been multiple studies focusing on the effect of nanoparticles on the proliferation and/or differentiation of MSCs and bone cancer cells; however, the specific studies with AgNPs are limited. Although the reported investigations show promising in vitro and in vivo potential of AgNPs for application in bone regeneration, more studies are required to ensure their implications in bone tissue engineering. This review aims to highlight the current advances related to the production of AgNPs and their effect on MSCs and bone cancer cells, which will potentiate their possible implications in orthopedics. Moreover, this review article evaluates the future of AgNPs in bone tissue engineering.
Collapse
Affiliation(s)
- Atharva Damle
- School of BioSciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Rajapriya Sundaresan
- School of BioSciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Jyutika M Rajwade
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, Maharashtra, India
| | - Priyanka Srivastava
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| | - Amruta Naik
- National Centre for Cell Science, S. P. Pune University Campus, Pune 411007, Maharashtra, India.
| |
Collapse
|
47
|
Effects of Exercise or Mechanical Stimulation on Bone Development and Bone Repair. Stem Cells Int 2022; 2022:5372229. [PMID: 36213684 PMCID: PMC9534715 DOI: 10.1155/2022/5372229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022] Open
Abstract
The development and regeneration of the bone are tightly regulated by mechanical cues. Multiple cell types, including osteoblasts, osteocytes, osteoclasts, mesenchymal stem cells (MSCs), and recently found skeletal stem cells (SSCs), are responsible for efficient bone development and injury repair. The immune cells in the environment interact with bone cells to maintain homeostasis and facilitate bone regeneration. Investigation of the mechanism by which these cells sense and respond to mechanical signals in bone is fundamental for optimal clinical intervention in bone injury healing. We discuss the effects of exercise programs on fracture healing in animal models and human patients, which encouragingly suggest that carefully designed exercise prescriptions can improve the result of fracture healing during the remodeling phase. However, additional clinical tracing and date accumulation are still required for the pervasive application of exercise prescriptions to improve fracture healing.
Collapse
|
48
|
Evaluation of the osteoinductive potential of HDPSCs cultured on β-glycerol phosphate functionalized MWCNTs/PCL membranes for bone regeneration. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-021-03721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Local immune cell contributions to fracture healing in aged individuals - A novel role for interleukin 22. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1262-1276. [PMID: 36028760 PMCID: PMC9440089 DOI: 10.1038/s12276-022-00834-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/25/2022] [Accepted: 06/06/2022] [Indexed: 11/08/2022]
Abstract
With increasing age, the risk of bone fractures increases while regenerative capacity decreases. This variation in healing potential appears to be linked to adaptive immunity, but the underlying mechanism is still unknown. This study sheds light on immunoaging/inflammaging, which impacts regenerative processes in aging individuals. In an aged preclinical model system, different levels of immunoaging were analyzed to identify key factors that connect immunoaged/inflammaged conditions with bone formation after long bone fracture. Immunological facets, progenitor cells, the microbiome, and confounders were monitored locally at the injury site and systemically in relation to healing outcomes in 12-month-old mice with distinct individual levels of immunoaging. Bone tissue formation during healing was delayed in the immunoaged group and could be associated with significant changes in cytokine levels. A prolonged and amplified pro-inflammatory reaction was caused by upregulated immune cell activation markers, increased chemokine receptor availability and a lack of inhibitory signaling. In immunoaged mice, interleukin-22 was identified as a core cell signaling protein that played a central role in delayed healing. Therapeutic neutralization of IL-22 reversed this specific immunoaging-related disturbed healing. Immunoaging was found to be an influencing factor of decreased regenerative capacity in aged individuals. Furthermore, a novel therapeutic strategy of neutralizing IL-22 may successfully rejuvenate healing in individuals with advanced immune experiences.
Collapse
|
50
|
Awale GM, Barajaa MA, Kan HM, Lo KWH, Laurencin CT. Single-Dose Induction of Osteogenic Differentiation of Mesenchymal Stem Cells Using a Cyclic AMP Activator, Forskolin. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00262-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|